You are on page 1of 14

Analytic Review

Journal of Intensive Care Medicine


1-14
Review of Biguanide (Metformin) Toxicity ª The Author(s) 2018
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1177/0885066618793385
journals.sagepub.com/home/jic

George Sam Wang, MD1,2 and Christopher Hoyte, MD1,3

Abstract
In the 1920s, guanidine, the active component of Galega officinalis, was shown to lower glucose levels and used to synthesize
several antidiabetic compounds. Metformin (1,1 dimethylbiguanide) is the most well-known and currently the only marketed
biguanide in the United States, United Kingdom, Canada, and Australia for the treatment of non-insulin-dependent diabetes
mellitus. Although phenformin was removed from the US market in the 1970s, it is still available around the world and can be
found in unregulated herbal supplements. Adverse events associated with therapeutic use of biguanides include gastrointestinal
upset, vitamin B12 deficiency, and hemolytic anemia. Although the incidence is low, metformin toxicity can lead to hyperlactatemia
and metabolic acidosis. Since metformin is predominantly eliminated from the body by the kidneys, toxicity can occur when
metformin accumulates due to poor clearance from renal insufficiency or in the overdose setting. The dominant source of
metabolic acidosis associated with hyperlactatemia in metformin toxicity is the rapid cytosolic adenosine triphosphate (ATP)
turnover when complex I is inhibited and oxidative phosphorylation cannot adequately recycle the vast quantity of Hþ from ATP
hydrolysis. Although metabolic acidosis and hyperlactatemia are markers of metformin toxicity, the degree of hyperlactatemia and
severity of acidemia have not been shown to be of prognostic value. Regardless of the etiology of toxicity, treatment should
include supportive care and consideration for adjunct therapies such as gastrointestinal decontamination, glucose and insulin,
alkalinization, extracorporeal techniques to reduce metformin body burden, and metabolic rescue.

Keywords
biguanide, metformin, phenformin, metabolic acidosis, hyperlactatemia, lactic acidosis

Introduction 1960s but is no longer available.7 We will focus on the toxicity


of metformin and phenformin in this review.
Biguanides are a class of drugs and chemicals based on the
biguanidine molecule (Figure 1). These compounds were ini-
tially derived from the Galega officinalis (French lilac or goat’s Available Formulations, Therapeutic Dosing,
rue) plant.1 In the 1920s, guanidine, the active component of and Pharmacokinetics
Galega officinalis, was shown to lower glucose levels and used
Metformin is available as a stand-alone medication in an
to synthesize several antidiabetic compounds. Metformin (1,1
immediate-release or extended-release tablet and liquid formu-
dimethylbiguanide) is probably the most well-known bigua-
lation. It is also available as a combination product with other
nide. Currently, it is the only marketed biguanide in the United
antihyperglycemic and antihyperlipidemic medications includ-
States, United Kingdom, Canada, and Australia for the treat-
ing pioglitazone, rosiglitazone, sitagliptin, linagliptin, saxa-
ment of non-insulin-dependent diabetes mellitus (NIDDM).2,3
gliptin, glipizide, and repaglinide.8,9 The US Food and Drug
Other biguanides include antidiabetic agents phenformin (phe- Administration (FDA)-labeled indication for metformin is type
nylethyl biguanide) and buformin (1-butylbiguanide) and the 2 diabetes mellitus, but also used for hyperinsular obesity,
antimalarial proguanide (N-(4-chlorophenyl)-N-(isopropyl)-
imidodicarbonimidic diamide). Phenformin was marketed in
the United States in 1950s. It was removed from the market 1
University of Colorado Anschutz Medical Campus, Aurora, CO, USA
2
in the late 1970s because there was a high incidence of hyper- Children’s Hospital Colorado, Aurora, CO, USA
3
lactatemia and metabolic acidosis associated with its therapeu- University of Colorado Hospital, Aurora, CO, USA
tic use.4,5 Proguanil and chlorproguanil are commercially Received June 26, 2018. Received revised July 16, 2018. Accepted July 18,
available antimalarials marketed as combination products with 2018.
atovaquone and dapsone, respectively. Polyaminopropyl
Corresponding Author:
biguanide, chlorhexidine, alexidine, and polyhexanide are George Sam Wang, University of Colorado at Denver—Anschutz Medical
examples of biguanides used as antiseptics and disinfectants.6 Campus, 13123 E 16th Ave B251, Aurora, CO 80401, USA.
Moroxydine was briefly used as a treatment for influenza in the Email: george.wang@childrenscolorado.org
2 Journal of Intensive Care Medicine XX(X)

Figure 1. Structures of biguanides.

hypersecretion of ovarian androgens, polycystic ovary syn- 1.5 mg/dL (132 mmol/L) in men and 1.4 mg/dL (123 mmol/L)
drome, and weight gain induced by antipsychotic therapy.1,8 in women and is contraindicated in patients with a glomerular
The recommended oral metformin dosing for adults is 500 to filtration rate <50 mL/min.9,10 The Canada Diabetes Associa-
2000 mg daily. Metformin has an oral bioavailability of 40% to tion, the National Institute for Health and Clinical Excellence,
60% after absorption in the small intestine, with complete gas- and the Australian Diabetes Society recommend that an esti-
trointestinal absorption by 6 hours, and a time to peak concen- mated glomerular filtration rate 29 mL/min/1.73 m2 should
tration between 4 to 8 hours.10 It is rapidly distributed and has be a contraindication for metformin use.11,12
minimal protein binding. Therapeutic blood or plasma metfor- Phenformin is available in Europe, South America, and Asia
min concentrations are 0.5 to 1 mg/L in a fasting state and 1 to 2 and can be obtained through Internet or mail orders.13-19
mg/L after a meal and are reached within 24 to 48 hours.9,10 Despite not being available in the US market, phenformin and
Metformin does not undergo hepatic metabolism. Approxi- buformin are still available in several countries in Europe,
mately 90% of the absorbed dose is renally excreted involving South America, and Asia and can be found in unregulated
glomerular filtration and tubular secretion. The plasma elimi- herbal supplements.13-19 In an analysis of 29 adulterated herbal
nation half-life is 4 to 8.7 hours, which can be prolonged in antidiabetic products in Hong Kong, 18 of them contained
patients with renal insufficiency. In blood, the elimination half- phenformin.19 When commercially available, the therapeutic
life is approximately 17.6 hours, suggesting that the erythro- dose is 50 to 200 mg/d. The pharmacokinetics of phenformin
cyte mass may be a compartment of distribution. The FDA does are different from metformin. Phenformin is more lipophilic
not recommend metformin use with creatinine concentrations and undergoes some hepatic metabolism with 33% eliminated
Wang and Hoyte 3

unchanged. Phenformin is 19% protein bound and is slower to risk for B12 deficiency include empiric vitamin B12 supplemen-
clear with an elimination half-life of approximately 11 hours, tation, monitoring B12 concentrations and/or blood counts for
and the majority of the drug in the therapeutic setting is elim- anemia, or regular examinations evaluation for neuropathy.34
inated within the first 24 hours.20,21 There are also case reports of vitamin B12 deficiency associated
with phenformin therapy.38-40
Metformin-associated hemolytic anemia is rarely
Mechanism of Action in Therapeutic Dosing
reported.41-48 The majority of case reports are associated with
In therapeutic doses, metformin appears to decrease blood therapeutic metformin dosing, and one case report in the setting
glucose levels by several mechanisms. Metformin enhances of an overdose.48 There are 2 case reports where hemolysis
suppression of gluconeogenesis by insulin and reduces recurred following metformin rechallenge.42,47 Metformin-
glucagon-stimulated gluconeogenesis.1 Metformin can posi- associated hemolytic anemia has been reported in patients with
tively affect insulin receptor phosphorylation and tyrosine comorbidities such as leukemia44 and glucose-6-phosphate
kinase activity. Metformin can also increase translocation of dehydrogenase deficiency.41,43,45 Serologic data suggest met-
the Glucose Transporter (GLUT)-1 and GLUT-4 isoforms formin hemolytic anemia can be immunologically
of glucose transporters as well as prevent the development of mediated.42,44,46,47 Although metformin-associated hemolytic
insulin resistance in hepatocytes and adipocytes.1,5 By increas- anemia can be fatal,46 patients usually recover with supportive
ing hepatic insulin sensitivity, metformin reduces basal hepatic care and discontinuation of metformin.
glucose output and fasting glucose concentrations.22,23 The
other antidiabetic biguanides, phenformin, and buformin have
similar mechanisms of action as metformin.
Epidemiology
In 2009, 37% of all Medicare beneficiaries with diabetes were
Adverse Events Associated With Therapeutic
on metformin therapy as part of their care, higher than insu-
Dosing lins, sulfonylureas, and thiazolidinediones.49 Despite this pre-
Adverse events associated with therapeutic use of all bigua- valent use of metformin, the reported rate of “lactic acidosis”
nides include gastrointestinal symptoms (eg, nausea, vomiting, is low. Salpeter and colleagues evaluated 194 comparative
diarrhea, and abdominal pain).8,9 Reports of acute hepatitis and trials or observation cohort studies that evaluated metformin
cholestasis are rarely reported with metformin.24-28 Although therapy alone or in combination with other treatments for at
there was an apparent temporal relationship to hepatotoxicity least 1 month.50 They found no cases of “lactic acidosis” in
and the initiation of metformin in case reports, all these patients 36 893 patient-years in the metformin group or 30 109 patient-
had significant comorbidities (eg, type 2 diabetes and hyper- years in the nonmetformin group. The estimated true incidence
tension), along with use of other potentially hepatotoxic med- of “lactic acidosis” in the metformin and the nonmetformin
ications including statins and angiotensin-converting enzyme was 8.1 and 9.9 per 100 000 patient-years, respectively.50 The
inhibitors. None of these patients were rechallenged with met- same authors published a Cochrane Review several years later
formin, and all reported cases resolved with discontinuity of all that included 347 comparative trials and cohort studies in
their medications. patients who received metformin for at least a month com-
Metformin has been associated with malabsorption syn- pared with placebo or other glucose-lowering therapy.51 There
dromes resulting in electrolyte abnormalities and vitamin B12 were no cases of fatal or nonfatal “lactic acidosis” in 70 490
deficiency.29-40 Associated diarrhea can lead to hypomagne- patient-years of metformin use or in 55 451 patient-years in
saemia, hypocalcemia, and hypokalemia.32,33 Vitamin B12 the nonmetformin group. Their estimate of the incidence of
deficiency can present clinically as megaloblastic anemia or “lactic acidosis” per 100 000 patient-years was 4.3 cases in the
neuropathies. The mechanism of vitamin B12 deficiency is metformin group and 5.4 cases in the nonmetformin group.
unclear but is proposed to be multifactorial including altera- However, the interobserver agreement was low on whether
tions in gut flora, motility, competitive inhibition of absorption, metformin was the most important causative factor in cases
or impairing calcium-dependent membrane actions on the ter- of “lactic acidosis,” and passive reporting in trials similar to
minal ileum.34 A case–control study concluded that each gram those reviewed may be limited due to underreporting.
per day metformin dose increment conferred an odds ratio of Chan and colleagues performed a literature search to find
2.88 and duration greater than 3 years of use conferred an odds metformin-associated “lactic acidosis” cases that were reported
ratio of 2.39 for developing vitamin B12 deficiency.36 When worldwide: 31 cases in the United Kingdom over 34 years (19
vitamin B12 concentrations in diabetics who received metfor- were fatal), 16 cases in 14 years in Sweden, 2 nonfatal cases in
min were compared to diabetics who received sulfonylureas, Switzerland over 5 years, 73 cases in France in 8 years (nearly
vitamin B12 concentrations were lower in patients receiving half were fatal), and none in Canada in 10 years.2 The reported
metformin as early as 4 months of initiating therapy.30 Vitamin incidence of metformin-associated “lactic acidosis” was 3
B12 deficiency has responded to B12 supplementation but may cases per 100 000 patient-years. However, these investigators
necessitate discontinuation of metformin. Recommendations did not qualify their search results and inclusion criteria for
for patients receiving metformin therapy who may be at high analysis. There was no determination if the “lactic acidosis”
4 Journal of Intensive Care Medicine XX(X)

Figure 2. Biochemical pathways involved in metformin pathophysiology.

cases were associated with therapeutic or overdose setting, and Pathophysiology of Metformin Toxicity
with most pooled data, it is subject to underreporting.
Hyperlactatemia and metabolic acidosis are the hallmark of
In contrast to “lactic acidosis” in the therapeutic setting,
biguanide toxicity, and it is often misrepresented as “lactic
regional poison centers review metformin-related toxicity in
acidosis,” that is, lactate production releases a proton and
both acute overdose and therapeutic settings. The Toxic Expo-
sure Surveillance System of the America Association of Poison causes acidosis. Biochemically, lactate production is always
Control Centers reported a total of 10 958 526 total exposures accompanied by an equivalent appearance of hydrogen ion;
from 1996 to 2000, of which 4072 were metformin exposures. an increase in blood lactate concentration (hyperlactatemia)
Approximately 1% of metformin exposures led to severe does not result in excess hydrogen ions and would not contrib-
adverse events.52 From 2009 through 2013 (excluding 2010, ute to the actual cause of acidosis. However, hyperlactatemia or
case fatalities were not reported), the American Association of an increase in lactate to pyruvate ratio is a biochemical marker
Poison Control Center National Poison Data System reported for a decrease in aerobic metabolism (Figure 2). To better
21, 30, 30, and 39 fatality cases, respectively, where metformin understand this, we will briefly review the biochemistry and
was noted to be at least contributory to the patient’s death.53-57 metabolic pathways involved in lactate metabolism. This is
The incidence of phenformin-associated “lactic acidosis” followed by a review and discussion for a mechanism of hyper-
was reported to be much higher than metformin. Between lactatemia and metabolic acidosis during metformin toxicity.
1965 and 1977, The Swedish Adverse Drug Reaction Commit- In normal (aerobic) conditions, glycolysis generates 2 ade-
tee reported 91 suspected adverse drug reactions to biguanides: nosine triphosphate (ATP) molecules, while 2 molecules of
50 reports of “lactic acidosis” and 19 deaths with phenformin, nicotinamide adenine dinucleotide (NADþ) are reduced to
compared with one nonfatal case of “lactic acidosis” with met- NADH, primary source of cytosolic NADH. An intermediary
formin.58 In 1979, a registry in Finland estimated 1 in 2300 to sequence is the oxidation of fructose 1,6-bisphosphate to gly-
5700 patients taking phenformin therapeutically developed ceraldehyde 3-phosphate and dihydroxyacetone phosphate
“lactic acidosis,” compared to an estimated 1 in 40 000 to (DHAP). Dihydroxyacetone phosphate is rapidly and reversi-
80 000 in patients taking metformin or buformin.59 A review bly isomerized by triosephosphate isomerase to glyceraldehyde
of 330 cases in 1978 of biguanide-associated “lactic acidosis” 3-phosphate, which is oxidized to 1,3-bisphosphoglycerate
reported 86% of the cases were due to phenformin and 50% of with the simultaneous reduction in NADþ to NADH; NADþ
these patients died.60 must be regenerated for glycolysis to continue. Two major
Wang and Hoyte 5

cytosolic reduction–oxidation (REDOX) reaction shuttle sys- In vitro and animal studies have shown metformin inhibits
tems, the glycerol 3-phosphate shuttle and the malate-aspartate mGPD but not cGPD at doses used to treat patients with
(MAL-ASP) shuttle, transport the electrons from cytoplasmic NIDDM, and at higher doses inhibits oxidative phosphoryla-
NADH into the mitochondria. The glycerol 3-phosphate shuttle tion complex I of the mitochondrial electron transport
is composed of cytosolic and mitochondrial isoforms of gly- chain. 66-69 Inhibition of mGPD would halt the glycerol
cerol 3-phosphate dehydrogenase (GPD). Cytoplasmic GPD 3-phosphate shuttle, disrupt glyceraldehyde 3-phosphate oxida-
(cGPD) catalyzes the reduction in DHAP to glycerol 3- tion to DHAP, and increase the cytosolic REDOX state.
phosphate, while one molecule of NADH is oxidized to Gluconeogenesis becomes impaired as a result of decreased
NADþ. Glycerol 3-phosphate is reoxidized back to DHAP DHAP and increased cytosolic NADH. The increase in cyto-
by mitochondrial GPD (mGPD), a flavin adenine dinucleotide solic NADH is unfavorable for the oxidation of lactate to pyr-
(FAD)-linked enzyme that transfers electron pairs to FADþ. uvate, thus decreasing the pyruvate pool for gluconeogenesis.
The resultant FADH2 transfers its electrons to the electron Lactate accumulates and hyperlactatemia is the result of the
carrier ubiquinone, which then enters the electron transport inability of lactate to participate in its entry into metabolism
chain as ubiquinol. The transport of electrons from cytosolic via the pyruvate pool. Tissues that normally rely on mitochon-
NADH into mitochondria by the MAL-ASP shuttle, in contrast drial respiration for ATP (eg, skeletal muscle) would be the
with the glycerol 3-phosphate shuttle, is readily reversible and dominant source of lactate production as cellular energetics
is principally driven by the mitochondrial transmembrane elec- become dependent on accelerated anaerobic glycolysis when
trical potential gradient established by the proton pump of the mitochondrial function is compromised; major source of lac-
electron transport chain.61-63 The end process of aerobic meta- tate is nonhepatic in origin. Although hyperlactatemia may be
bolism occurs when pyruvate is transported into the mitochon- viewed as an imbalance between lactate production and lactate
dria. Pyruvate is decarboxylated to acetyl-CoA in a reaction utilization, the relative clinical importance of these factors
catalyzed by pyruvate dehydrogenase complex with reduction associated with hyperlactatemia is controversial. However,
in NADþ to NADH and enters the citric acid cycle, which fuels there is little doubt that during severe acidosis, there is reduced
the electron transport chain by generating NADH and supply- lactate uptake by the liver and the liver becomes a source of
ing succinate to be oxidized to fumarate while FADþ is lactate production. Both blood lactate and pyruvate production
reduced to FADH2; FADH2 effectively passes its electron onto have been observed to increase with a disproportionally greater
ubiquinone reducing it to ubiquinol and regenerates FADþ. As increase in lactate production, resulting in a significant increase
electrons from NADH and FADH2 move along the electron in lactate to pyruvate ratio during severe phenformin toxi-
transport chain, hydrogen ions (Hþ) are pumped across the city.70-74
inner mitochondrial membrane into the mitochondrial inter- Inhibition of complex I compromises oxidative phosphory-
membrane space, creating a transmembrane electrochemical lation by impeding electron transport, slows NADþ regenera-
potential that drives ATP production (ie, chemiosmosis).64 tion, and retards the citric acid cycle and thus decreases the
Normally, pyruvate, NADH, Hþ produced from substrate availability of succinate and consequently electrons being
flux through glycolysis, and products of ATP hydrolysis (ADP, transferred to FADþ and processing through complex II of the
Pi, and Hþ) are predominantly consumed as substrates during electron transport chain. As electron transport is slowed, recy-
mitochondrial respiration. Oxidative phosphorylation recycles cling of Hþ becomes inept and the mitochondrial transmem-
vast quantities of Hþ for ATP synthesis, thereby maintaining a brane falters resulting in paralysis of the MAL-ASP shuttle and
virtually constant pH, and is the primary source of cellular decreased ATP production. A dysfunctional MAL-ASP shuttle
bioenergetics. When mitochondrial oxidative phosphorylation also slows the removal of cytoplasmic NADH and exacerbates
is compromised (eg, complex I inhibition), there is a precipi- the high cytoplasmic REDOX state created by mGPD inhibi-
tous decrease in ATP production, and in an attempt to sustain tion, which further favors the reduction of pyruvate to lactate.
cellular energy stores, ATP production becomes dependent on A compromised oxidative phosphorylation will not be able to
glycolysis. As glycolysis intensifies, there is an increase in keep pace with the demands of cellular energetics and it
glucose consumption, substrate flux to pyruvate, and reduction becomes dependent on glycolysis for ATP production. Glyco-
in pyruvate to lactate in a reaction catalyzed by lactate dehy- lytic ATP production is sustained by a corresponding increase
drogenase (LDH) with the oxidation of NADH to NADþ. A in glucose consumption and reduction in pyruvate to lactate
conceptually important aspect of this catalytic reaction is lac- while NADH is oxidized to NADþ. The increased rate of ATP
tate (not lactic acid). When pyruvate is reduced to lactate, 2 production has to be matched by a corresponding rate in glu-
electrons and a proton are removed from NADH, and a proton cose consumption with expectant hypoglycemia unless an ade-
is consumed from solution. There is no net Hþ released and a quate supply of glucose is maintained. A compromised
proton is consumed each time a molecule of lactate is pro- oxidative phosphorylation also cannot adequately recycle the
duced. Thus, the formation of lactate is not a source of net vast amount of Hþ that is released when ATP is hydrolyzed for
Hþ and lactate does not cause or contribute to metabolic acido- cellular energetics, and when the endogenous buffering capac-
sis.65 The disposition of lactate is its entry into metabolism ity is overwhelmed, metabolic acidosis ensues.
through LDH by a REDOX-dependent reaction into the pyru- As glycolysis intensifies and the supply of glucose
vate pool. becomes critical, cellular fuel selection switches from glucose
6 Journal of Intensive Care Medicine XX(X)

to fat (ie, glucose fatty acid or Randle cycle). Triacylglycerols harvested within 48 hours of patients diagnosed with hyperlac-
are mobilized and broken down to fatty acids and glycerol. tatemia and metabolic acidosis from metformin toxicity show
Fatty acid b-oxidation produces acetyl-CoA, which can serve significantly lower oxidative phosphorylation complex I and
as fuel when processed through the citric acid cycle. How- complex IV activity compared to healthy controls.83 Metabolic
ever, b-oxidation and acetyl-CoA entry into the citric acid profile from patients with metformin or phenformin toxicity is
cycle are hindered because of limited NADþ. Acetyl-CoA characterized by hyperlactatemia, metabolic acidosis, signifi-
accumulates and high levels of acetyl-CoA favors the forma- cant increase in lactate/pyruvate ratio (eg, 2- to 200-fold), and
tion of acetoacetate and a high mitochondrial REDOX state blood alanine concentration (eg, 4- to 13-fold).70,72-74,84,85
favors the reduction of acetoacetate to b-hydroxybutyrate. Hemodynamic studies of metformin or phenformin toxic
Glycerol can be phosphorylated to glycerol 3-phosphate and patients without cardiac or liver failure or sepsis show severe
enters the gluconeogenic pathway at DHAP, but this pathway hyperlactatemia and metabolic acidosis and abnormally low
is muted with the inhibition of mGPD and prevents the oxida- systemic oxygen consumption despite normal or increased glo-
tion of glycerol 3-phosphate to DHAP. However, if glycerol bal oxygen delivery.86 Resolution of drug intoxication is par-
flux becomes significant (eg, lipolysis), its oxidation to alleled by correction of hyperlactatemia and metabolic acidosis
DHAP by cGPD with an increase in cytosolic NADH with normalization of systemic oxygen consumption.
becomes favorable; production of DHAP and NADH by Another expected consequence of accelerated glycolysis
cGPD running in the opposite direction.75 Although this gly- and muted gluconeogenesis is cellular hypoglycemia. Data
cerol–gluconeogenic pathway may be possible, it is an unfa- from in vitro human platelets study suggest that metformin
vorable reaction because it has to occur at the expense of decreases plasma pH and increases cellular glucose consump-
exacerbating a high cytosolic REDOX state created by the tion and lactate production in a dose-dependent manner and
paralysis of glycerol phosphate and MAL-ASP shuttles. A that plasma glucose concentrations are inversely correlated
high cellular REDOX state limits fatty acid b-oxidation and with lactate concentrations. 83 Clinically, there have been
its usefulness as a fuel source. Cellular fuel selection turns to numerous case reports of hypoglycemia associated with met-
protein. Amino acids (eg, alanine) are released from muscles formin toxicity, and one case seems particularly relevant in
as a source of precursors for gluconeogenesis via pyruvate. that severe hypoglycemic episodes appeared proximate to
However, gluconeogenesis is an energy costly process and when hyperlactatemia and metabolic acidosis were most
cellular bioenergetics is relying on glycolysis for ATP pro- severe.87
duction. Processes that are ATP costly are unfavorable. As It may be expected that a state of relative or absolute insulin
alanine entry into gluconeogenesis is impeded, alanine deficiency (eg, diabetes mellitus), superimposed upon metfor-
accumulates.74,76 min toxicity, may potentiate hyperlactatemia, metabolic acido-
The dominant source of metabolic acidosis associated with sis, and cellular hypoglycemia. That is, hyperlactatemia,
hyperlactatemia in metformin toxicity is the rapid cytosolic (ie, metabolic acidosis, and cellular hypoglycemia may be more
nonmitochondrial) ATP turnover (ie, glycolytic generation of severe in metformin toxic patients with diabetes mellitus. Insu-
ATP coupled to ATP hydrolysis) when complex I is inhibited lin deficiency would also aggravate lipolysis and promote mus-
and oxidative phosphorylation cannot adequately recycle the cle breakdown with release of amino acids.
vast quantity of Hþ from ATP hydrolysis.65,77-82 The same mechanisms describing hyperlactatemia, meta-
Animal model of severe metformin toxicity and human cel- bolic acidosis, and cellular hypoglycemia in metformin toxicity
lular biochemical, metabolic, and hemodynamic data from apply to phenformin toxicity.66,88-90 However, phenformin
metformin and phenformin toxicity suggest the biochemistry inhibits oxidative phosphorylation complex I more effectively
and biochemical pathways involved in lactate metabolism and than metformin. This is probably because phenformin is sig-
mechanisms of hyperlactatemia and metabolic acidosis may be nificantly more hydrophobic, allowing it to more readily enter
clinically relevant. Data from a swine model of severe metfor- and accumulate in the mitochondria; positively charged bigua-
min toxicity suggest there is decreased global oxygen con- nides accumulate in the mitochondria in response to the poten-
sumption and mitochondrial dysfunction in heart, kidney, tial difference across its inner membrane.67,91
liver, skeletal muscle, and platelets. Hyperlactatemia per se
does not decrease whole-body respiration, and diffuse inhibi-
tion of cellular respiration and secondary lactate overproduc-
Acute Overdose
tion contribute to the observed metabolic acidosis and Large acute overdoses can lead to significant toxicity and can-
hyperlactatemia of metformin toxicity.83 In vitro healthy not be explained by any other major risk factor other than a
human donor platelets studies demonstrate metformin biguanide overdose.92,93 The toxic dose that leads to hyperlac-
increased lactate production and glucose consumption and tatemia and metabolic acidosis is unclear, but large doses are
decreased the activity of oxidative phosphorylation complex typically reported.94-100 A case series of acute metformin
I, mitochondrial membrane potential, and oxygen consumption ingestions over 20 years reported adult ingestions of 3.5 to
in a dose- and time-dependent manner. These changes occurred 22.5 g did not develop hyperlactatemia or metabolic acido-
independently from hypoxia and differences in platelet count sis. 101 Lacher described a 15-year-old girl who ingested
and mitochondrial density.83 Ex vivo studies of human platelets 38.25 g and developed “lactic acidosis” and moderate renal
Wang and Hoyte 7

failure.96 There have also been reports of survival after acute 4.3 in 100 000 (43/1 000 000) in patients with risk factors or
ingestions of 60 to 100 g of metformin and pH as low as comorbidities.105
6.38.97-100 Patients who develop hyperlactatemia and metabolic acido-
The clinical course of acute unintentional pediatric metfor- sis while on metformin therapy are often categorized into 2
min exposures appears to be generally benign. In a review of 55 different types of toxicity: the so-called metformin-associated
pediatric patients, age 15 months to 17 years, metformin expo- lactic acidosis (MALA) and metformin-induced lactic acidosis
sure from 8 regional poison centers reported that assessable (MILA). These terms are sometimes used interchangeably and
patients did not develop hypoglycemia, metabolic acidosis, or poorly defined. For the purposes of this review, MALA occurs
hyperlactatemia and exposure to metformin 1700 mg did not in the setting of a patient receiving therapeutic metformin and
lead to acidosis or significant clinical illness.95 However, none develops severe pathology but does not have significant met-
of these exposures were confirmed by metformin concentra- formin accumulation. Common comorbidities include shock
tions, 62% were not assessed for acidosis, and 31% did not states (eg, distributive, hypovolemic, and cardiogenic),
have serial glucose measurements. obstructive pulmonary disease, heart failure, respiratory fail-
Glucose abnormalities have been reported in acute overdose ure, and hepatic failure.105-109 In this setting, an etiology cannot
settings. Hyperglycemia was reported in an otherwise healthy be directly or solely attributed to metformin and is usually
young patient who ingested over 60 g of metformin and devel- associated with other coexisting states. The MILA is also in
oped renal insufficiency and severe “lactic acidosis.”102 A the setting of therapeutic metformin, but metformin accumula-
review of all metformin exposures reported to a regional poison tion has some degree of responsibility. This is of greatest con-
center found 1.5% of patients developed hypoglycemia.103 cern in patients with renal insufficiency whose metformin
Hypoglycemia was described in a 15-year-old girl who dosing has not been accordingly adjusted.21,110-112 Since met-
ingested 75 g of metformin and 3 g of quetiapine, who devel- formin is almost entirely eliminated by the kidneys, renal
oped “lactic acidosis,” hypotension, and severe hypoglyce- impairment is a major risk factor for development of MILA.
mia.87 In the setting of hypoglycemia, the clinician should Acute kidney injury leads to decreased metformin elimination
also remain vigilant for other etiologies of hypoglycemia or and increased metformin systemic concentrations during ther-
other exposures, as many patients taking metformin also take apeutic dosing. Cimetidine reduces metformin tubular secre-
sulfonylureas or insulin. tion, but is unclear how much this contributes to toxicity.2
Besides hyperlactatemia and metabolic acidosis, there
have also been rare reports of metformin toxicity leading to
Adverse Events in Therapeutic Setting encephalopathy, hyperglycemia, hypoglycemia, and pancreati-
Hyperlactatemia and metabolic acidosis are very rare during tis.113-117 In one case series, a patient developed encephalopa-
therapeutic metformin dosing in patients without comorbid- thy temporally related to metformin therapy that resolved after
ities. In an animal model, rats were fed 1, 200, 600, 900, or its withdrawal, while another patient had been treated with
1200 mg/kg/d for 13 weeks. Doses >600 mg/kg/d were associ- metformin for years prior to presentation and developed ence-
ated with minimal metabolic acidosis characterized by phalopathy following initiation of repaglinide.113 Interestingly,
increased serum lactate and b-hydroxybutyrate concentrations; this patient became asymptomatic when repaglinide and met-
however, doses >900 mg/kg/d were associated with some ani- formin were discontinued, but encephalopathy recurred when
mals being moribund. The no-observable-adverse-effect level patient was rechallenged with metformin. Others have reported
was 200 mg/kg/d, which corresponds to approximately twice encephalopathy and parkinsonian symptoms in patients who
the plasma metformin concentration and 8 times the maximum received metformin in the setting of end-stage renal disease
plasma concentration achieved in humans following a 2000 mg requiring dialysis.114-116 Symptoms were correlated with high
total daily dose.104 signal intensity in the basal ganglia on MRI T2-weight images
Within the first 13 months of metformin approval in the that resolved when metformin was discontinued and with clin-
United States, 47 patients on metformin therapy with con- ical improvement.115,116
firmed “lactic acidosis” (lactate >5 mmol/L) were reported to It is rare for patients on metformin therapy to experience
the FDA.105 Of these 47 patients, 43 had at least one risk factor hypoglycemia. The incidence of moderate to severe hypogly-
(eg, cardiac disease, renal insufficiency, pulmonary disease, cemia for patients on metformin therapy is 60 per 100 000, and
and older age) for “lactic acidosis” and 20 of these patients the odds ratio of developing moderate to severe hypoglycemia
died.106 Only 4 patients had no apparent risk factors and they associated with metformin use is 1.42 (95% confidence inter-
all recovered.105 Based on an estimate of 1 million Americans val: 1.22-1.64).117
were taking metformin during this time, the reported rate of
confirmed “lactic acidosis” is about 5 cases per 100 000
(47/1 000 000), which suggests the overall development of
Diagnosis of Metformin Toxicity
hyperlactatemia and metabolic acidosis in patients on metfor- History and physical examination are mainstays in diagnosing
min therapy to be very rare. This also suggests development of metformin toxicity. Of course, history of acute overdose should
hyperlactatemia and metabolic acidosis in patients without lead to appropriate evaluation. However, signs and symptoms
risk factors is 0.4 in 100 000 (4/1 000 000) compared to of metformin toxicity in the therapeutic setting can be
8 Journal of Intensive Care Medicine XX(X)

nonspecific and pose a diagnostic challenge.111,112 Signs and Table 1. Differential Diagnosis for Hyperlactatemia and Associated
symptoms include nausea, vomiting, fatigue, hypovolemia, Metabolic Acidosis.
altered mentation, poor urine output, tachycardia, tachypnea, Toxicological Etiology Nontoxicological Etiology
and hypotension or shock.
Serum biguanide concentrations in the acute overdose set- Cyanide Sepsis
ting are unhelpful in patient management. Serum metformin Hydrogen sulfide Shock
concentration may aid in determining the final diagnosis of Rotenone Status epilepticus
HIV antiretrovirals (nucleoside Inborn error of metabolism
a patient but will unlikely return in a timely fashion and
reverse transcriptase inhibitor)
unlikely affect acute clinical management. Overall, it is rare Isoniazid Antibacterial-induced D-lactic
to have significant metformin accumulation without acidosis
hyperlactatemia and metformin-induced hyperlactatemia with- Linezolid Liver failure
out metformin accumulation.118 Metformin concentrations Propylene glycol Warburg effect
typically associated with hyperlactatemia and metabolic acido- Propofol infusion syndrome
sis are >5 mg/mL.92,119,120 Adrenergic stimulants
(eg, theophylline, salbutamol)
Nalidixic acid
Prognostic Factors Acetaminophen (massive overdose)
Fructose
Acute Overdose
There are case reports of patients who survived acute serious
ketoacidosis, alcoholic ketoacidosis, and xenobiotic-induced
metformin overdoses with serum pH 6.38 and hyperlactatemia
myocardial suppression).
(>20 mmol/L).96-101 However, survival rates appear higher in
patients without profound acidemia and hyperlactatemia. Del-
l’Anglio and colleagues performed a literature review and found Treatment
22 cases of acute metformin overdoses with documented serum
There is no specific antidote for metformin toxicity and its
pH, lactate levels, and metformin concentrations and found no
mainstay of therapy is supportive care, which should include
patient with a nadir serum pH >6.9, lactate concentration
management of fluids, electrolytes, acid–base, respiratory,
<25 mmol/L, or peak serum metformin concentration <50 mg/mL
metabolic, renal, and hemodynamic derangements. Some
died.121 Seidowsky and colleagues reported the overall relative
adjunct therapies to consider include gastrointestinal deconta-
risk for death was higher in metformin overdose patients with a
mination, glucose and insulin, serum alkalinization, extracor-
serum pH 7.2 and arterial lactate 15.122 When compared
poreal techniques to reduce metformin body burden, and
with patients with incidental overdoses, patients with intentional
metabolic rescue. Gastrointestinal decontamination (eg, gastric
overdoses developed less acidosis, developed end-organ dys-
lavage and activated charcoal) should be considered following
function, and had better survival rates.122 Prothrombin time has
a serious acute metformin overdose provided there is no
also been evaluated as a prognostic factor. Prolonged prothrom-
contraindications.
bin time appears to be a good predictor of mortality in patients
with metformin toxicity.122-124
Extracorporeal Techniques
Toxicity in Therapeutic Setting Extracorporeal techniques (eg, hemodialysis, continuous renal
Case series suggest serum lactate, pH, or metformin concen- replacement therapy [CRRT], hemoperfusion, and plasma
tration did not have prognostic value in patients who develop exchange), either alone or in combination, have been used to
toxicity while on therapeutic metformin dosing. Lalau and treat both acute metformin overdoses and metformin toxicity in
Race reported a series of 49 patients who developed “lactic the therapeutic setting.128-145
acidosis” while on metformin therapy and found there was no If hemodialysis can be tolerated, it can reduce metformin
difference in median arterial lactate level between patients who body burden, clear lactate, and correct acid–base abnormalities
survived and died, and plasma metformin concentrations were with great efficiency. However, seriously ill patients often have
3 times higher in patients who survived.125 Several other stud- marginal hemodynamics or are hemodynamically unstable and
ies have reported similar lack of predictive value of arterial pH, may not tolerate standard hemodialysis measures. The different
serum metformin, and arterial lactate concentrations.120,126,127 CRRT modalities (eg, continuous veno-venous hemofiltration,
continuous veno-venous hemodialysis, and continuous veno-
venous hemodiafiltration) may be reasonable alternatives to
Differential Diagnosis hemodialysis, as they cause less adverse hemodynamic effects
Differential diagnosis of metformin toxicity (Table 1) than with hemodialysis. However, CRRT is intended to substi-
includes toxicological and nontoxicological etiologies of tute for impaired renal function over an extended period and
hyperlactatemia and metabolic acidosis.83,86 Additional con- applied for 24 hours a day. Continuous renal replacement ther-
siderations include severe systemic illnesses (eg, diabetic apy is not intended to and cannot substitute for hemodialysis as
Wang and Hoyte 9

a method to achieve rapid xenobiotic (eg, metformin) clearance mortality. This suggests administration of large quantities of
compared to other modalities. Metformin has a low molecular bicarbonate may prolong survival to allow treatment of the
weight and low protein binding and is freely soluble in water, underlying cause of “lactic acidosis.”156 In terms of biguanide
making it amenable to plasma clearance by CRRT. However, toxicity, a retrospective review of more than 300 cases of
metformin has a relatively high volume of distribution biguanide metabolic acidosis and hyperlactatemia, survival of
(3.1 L/kg) and it has been estimated it takes an average of patients who were administered bicarbonate was no better than
15 cumulative hours of intermittent hemodialysis to achieve patients who received supportive care.60 Given the limited
therapeutic metformin concentrations following an overdose, options when metabolic derangement result in a pH less than
and the maximum metformin clearance rate has been reported 7.20 in the presence of underlying cardiovascular disease or
to approach 2.4%/h during CRRT.122,134,137,140,141 evidence of hemodynamic compromise, it is reasonable to con-
sider sodium bicarbonate as a pH buffer while awaiting the
deployment of hemodialysis. Sodium bicarbonate may deliver
Alkalinization an unwanted sodium load to patients with marginal cardiac
The benefit of base therapy in the treatment of metabolic acido- reserve and may contribute to adverse cardiovascular events.
sis associated with metformin toxicity is unclear. Base therapy Tris-hydroxymethyl aminomethane (THAM) is an alterna-
for metabolic acidosis is recommended at an arterial pH vary- tive alkalinizing agent to sodium bicarbonate. In contrast to
ing from 6.9 to 7.2.146 It is reasonable to consider base therapy sodium bicarbonate, THAM is a proton acceptor by virtue of
when arterial pH is less than 7.20 in the presence of underlying its amine group and neutralizes acids without generating car-
cardiovascular disease or evidence of hemodynamic compro- bon dioxide. Tris-hydroxymethyl aminomethane readily dif-
mise, as lower values may impair cardiovascular function and fuses through the extracellular space and moves into the
is associated with increased mortality.147-150 Unless efforts are intracellular compartment. Thus, THAM is able to balance
directed at reversing the underlying causes for the acidosis, pH in the setting of acidemia due to metabolic acid accumula-
base therapy is futile. In acidemic states where cardiac output tion or carbon dioxide retention. It is excreted in its protonated
is inadequate to meet systemic oxygen requirements and is form at a rate exceeding creatinine clearance and thus it may be
unimproved by catecholamines, partial correction of arterial less effective in renal dysfunction. However, THAM is dialyz-
pH may be necessary in order to restore adequate hemody- able and renal dysfunction does not obviate its use. Tris-
namics. The goal is to improve cardiac contractility and restore hydroxymethyl aminomethane may lower the partial pressure
responsiveness of the myocardium and peripheral vessels to of carbon dioxide as effectively as sodium bicarbonate and
endogenous and infused catecholamines, not an arbitrary arter- improves cardiac contractility in parallel with improvement
ial pH per se. In clinical practice, when base is given, the aim is in acid–base balance. 157-159 Tris-hydroxymethyl amino-
to maintain pH *7.2.151-153 methane has been used in conjunction with renal replacement
Sodium bicarbonate is the most commonly used base. Man- therapy in an acute overdose of metformin.160
agement of “lactic acidosis” with the use of sodium bicarbonate
is controversial despite its regular use. The source of metabolic
acidosis and lactate during metformin toxicity is intracellular. Glucose and Insulin
An increase in serum lactate concentration occurs when lactate Hypoglycemia should be aggressively treated with intravenous
diffuses out of cells and enters the extracellular compartment. dextrose. Glucose and insulin therapy is reasonable as it pro-
Thus, the primary problem is not in the extracellular compart- vides a source of glucose, facilitates glucose utilization, sus-
ment and administration of sodium bicarbonate does not tains glycolysis, mitigates hypoglycemia, and attenuates
directly address the intracellular problem. Extracellular hydro- lipolysis. The benefit of glucose and insulin therapy is sug-
gen and bicarbonate ions do not readily diffuse across cell gested by clinical experience with their use in severe phenfor-
membranes into the intracellular compartment where the pur- min toxicity in which mortality appears to be significantly
ported benefits of bicarbonate therapy occur. Administered reduced compared with patients who were not treated with
bicarbonate reacts with acids to form water and carbon dioxide, glucose and insulin.4
which readily diffuses into the intracellular compartment and
drives the formation of hydrogen and bicarbonate ions; sodium
bicarbonate administration will cause intracellular acidosis
Metabolic Rescue
unless the partial pressure of carbon dioxide can be decreased Methylene blue (MB) may be considered as a metabolic rescue
by increased ventilation. Thus, sodium bicarbonate administra- for metformin toxicity. Treatment that could help bypass elec-
tion, which increases extracellular pH, would not be expected tron transport impediment at complex I and support electron
to readily correct intracellular acidosis and improve hemody- transport, mitochondrial membrane potential, and ATP produc-
namics or augment sensitivity to catecholamines.154 Most tion may be helpful. Methylene blue can serve as an alternative
patients treated with conventional doses of bicarbonate showed electron carrier by accepting electrons from NADH and is
no increase in cardiac output or decrease in morbidity or mor- reduced to leucomethylene blue, which can then deliver its
tality.155 However, those treated with large doses of bicarbo- electrons to either ubiquinone or cytochrome c, thus resuscitat-
nate and concomitant dialysis appeared to have a decrease in ing oxidative phosphorylation and the citric acid cycle and in
10 Journal of Intensive Care Medicine XX(X)

the process regenerating MB.161-164 Although this may be 4. Misbin RL. Phenformin-associated lactic acidosis: pathogenesis
mechanistically reasonable, the effectiveness of MB as an and treatment. Ann Intern Med. 1977;87:591-595.
adjunct treatment is unclear. However, there are at least 5 case 5. Bailey CJ. Biguanides and NIDDM. Diabetes Care. 1992;15(6):
reports of serious metformin toxicity in which MB was used as 755-771.
adjunct therapy with 80% survival.165-169 Methylene blue may 6. Chawnder JA, Gilbert P. Interaction of the biguanides chlorhex-
also address other pathophysiologic effects. Some evidence has idine and alexidine with phospholipid vesicles: evidence for sep-
shown metformin increases peripheral perfusion via activation arate modes of action. J Appl Bacteriol. 1989;66(3):253-258.
of endothelial nitric oxide synthase (eNOS).170 Perhaps, MB 7. Sheppard S. Moroxydine: the story of a mislaid antiviral. Acta
may improve responsiveness to vasopressors, in addition to Derm Venereol Suppl (Stockh). 1994;183:1-9.
attenuating metformin’s activation of the eNOS system. 8. Metformin. http://www.nlm.nih.gov/medlineplus/druginfo/meds/
a696005.html. Accessed March 15, 2018.
9. Lupin Pharmaceuticals. http://www.lupinpharmaceuticals.com/
Conclusion pdf/12/12/PIL%20Metformin%20HCl%20ER%20tabs_RTF.
Biguanides include various medications, disinfectants, and PDF. Published July, 2011. Accessed March 21, 2015.
experimental compounds based on the biguanidine molecule. 10. Scheen AJ. Clinical pharmacokinetics of metformin. Clin Phar-
Currently, the most commonly used biguanides is the antidia- macokinet. 1996;30(5):359-371.
betic pharmaceutical metformin. Adverse effects include gas- 11. Stanton RC. Metformin use in type 2 diabetes mellitus with CKD:
trointestinal upset, vitamin B12 deficiency, and hemolytic is it time to liberalize dosing recommendations? Am J Kidney Dis.
anemia. Severe metformin toxicity characterized by hyperlac- 2015;66(2):193-195.
tatemia and metabolic acidosis occurs during therapeutic dos- 12. Lipska KJ, Bailey CJ, Inzucchi SE. Use of metformin in the
ing in the setting of coexisting conditions such as renal failure setting of mild-to-moderate renal insufficiency. Diabetes Care
and following acute serious overdose. Although hyperlactate- 2011;34(5):1431-1437.
mia and metabolic acidosis are markers of metformin toxicity, 13. Lu HC, Parikh PP, Lorber DL. Phenformin-associated lactic
the degree of hyperlactatemia and severity of acidemia have acidosis due to imported phenformin. Diabetes Care. 1996;
not been shown to be of prognostic value. Regardless of the 19(12):1449-1450.
etiology of toxicity, treatment should include supportive care 14. Rosand J, Friedberg JW, Yang JM. Fatal phenformin-associated
and consideration for adjunct therapies such as gastrointestinal lactic acidosis. Ann Intern Med. 1997;127(2):170.
decontamination, glucose and insulin, alkalinization, extracor- 15. Krishnamurthy M, Sahouria JJ, Desai R, Caquiat J. Buformin-
poreal techniques to reduce metformin body burden, and meta- induced lactic acidosis-a symptom of modern healthcare malady.
bolic rescue. J Am Geriatr Soc. 2004;52(1):1785.
16. Fimognari FL, Pastoreli R, Incalzi RA. Phenformin-induced lactic
Acknowledgments acidosis in an older diabetic patient: a recurrent drama (phenfor-
The authors would like to acknowledge Dr Luke Yip for his significant min and lactic acidosis). Diabetes Care. 2006;29(4):950-951.
contributions to the pathophysiology section and discussion of methy- 17. Fimognari FL, Corsonello A, Pastorelli R, Incalzi RA. Older age
lene blue as a metabolic rescue agent. and phenformin therapy: a dangerous association. Intern Emerg
Med. 2008;3:401-403.
Declaration of Conflicting Interests 18. Ching CK, Lai CK, Poon WT, et al. Hazards posed by a banned
The author(s) declared no potential conflicts of interest with respect to drug-phenformin is still hanging around. Hong Kong Med J. 2008;
the research, authorship, and/or publication of this article. 14(1):50-54.
19. Ching CK, Lam YH, Chan AY, Mak TW. Adulteration of
Funding herbal antidiabetic products with undeclared pharmaceuticals:
The author(s) received no financial support for the research, author- a case series in Hong Kong. Br J Clin Pharmacol. 2012;73(5):
ship, and/or publication of this article. 795-800.
20. Alkalay D, Khemani L, Wagner WE, Bartlett MF. Pharmacoki-
ORCID iD netics of phenformin in man. J Clin Pharmacol. 1975;15(5-6):
George Sam Wang, MD http://orcid.org/0000-0002-2931-3508 446-448.
21. Inzucchi SE, lipsak KJ, Mayo H, Bailey CJ, McGuire DK. Met-
References formin in patients with type 2 diabetes and kidney disease: a
1. Bailey CJ, Turner RC. Metformin. N Engl J Med. 1996;334(9): systematic review. JAMA. 2014;3123(24):2668-2675.
574-579. 22. Periello G, Misericordia P, Volpi E, et al. Acute antihyperglyce-
2. Chan NN, Brain HP, Feher MD. Metformin-associated lactic mic mechanisms of metformin NIDDM. Evidence for suppression
acidosis: a rare or very rare clinical entity? Diabet Med. 1999; of lipid oxidation and hepatic glucose production. Diabetes. 1994;
16(4):273-281. 43(7):920-928.
3. Harrigan RA, Nathan MS, Beattie P. Oral agents for the treatment 23. Jackon RA, Hawa MI, Jaspan JB. Mechanism of metformin action
of type 2 diabetes mellitus: pharmacology, toxicity, and treat- in non-insulin-dependent diabetes. Diabetes. 1987;36(5):
ment. Ann Emerg Med. 2001;38(1):68-78. 632-640.
Wang and Hoyte 11

24. Babich MM, Pike I, Shiffman ML. Metformin-induce acute hepa- 44. Kirkiz S, Yarali N, Arman Bilir O, et al. Metformin-induced
titis. Am J Med. 1998;104(5):490-492. hemolytic anemia. Med Princ Pract. 2014:23(2):183-185.
25. Nammour FE, Fayad NF, Peikin SR. Metformin-induced chole- 45. Meir A, Kleinman Y, Rund D, et al. Metformin-induced hemoly-
static hepatitis. Endocr Pract. 2003;9(4):307-309. tic anemia in a patient with glucose-6-phosphate dehydrogenase
26. Kutoh E. Possible metformin-induce hepatoxicity. Am J Geriatr deficiency. Diabetes Care. 2003;26(3):956-957.
Pharmcother. 2005;3(4):270-273. 46. Packer CD, Hornick TR, Augustine SA. Fatal hemolytic anemia
27. Biyyani RS, Battula S, Erhardt CA, Korkor K. Metformin- associated with metformin: a case report. J Med Case Rep. 2008;
induced cholangiohepatitis. BMJ Case Rep. 2009;2009. 2:300.
28. Cone CJ, Bachyryca AM, Murata GH. Hepatotoxicity associated 47. Kashyap AS, Kashayap S. Haemolytic anaemia due to metformin.
with metformin therapy in treatment of type 2 diabetes mellitus Postgrad Med J. 2000;76(892):125-126.
with nonalcoholic fatty liver disease. Ann Pharmacother. 2010; 48. Jagia M, Taqi S, Hanafi M. Metformin poisoning: a complex
44(10):1655-1659. presentation. Indian J Anaesth. 2011;55(2):190-192.
29. Callaghan TS, Hadden DR, Tomkin GH. Megaloblastic anaemia 49. Sargen MR, Hoffstad OJ, Wiebe DJ, Margolis DJ. Geographic
due to vitamin B12 malabsorption associated with long-term met- variation in pharmacotherapy decision for U.S. Medicare enrol-
formin treatment. Br Med J. 1980;280(6225):1214-1215. lees with diabetes. J Diabetes Complications. 2012;26(4):
30. Bauman WA, Shaw S, Jayatilleke E, et al. Increased intake of 301-307.
calcium reverses vitamin B12 malabsorption induced by metfor- 50. Salpeter SR, Greber EG, Pasternak GA, et al. Risk of fatal and
min. Diabetes Care. 2000;23(9):1227-1231. nonfatal lactic acidosis with metformin use in type 2 diabetes
31. Ting RZ, Szeto CC, Chan MH, Ma KK, Chow KM. Risk factors mellitus. Arch Intern Med. 2003;163(21):2594-2602.
of vitamin B12 deficiency in patients receiving metformin. Arch 51. Salpeter SR, Greyber E, Pasternak GA, Salpeter EE. Risk of fatal
Intern Med 2006;166(18):1975-1979. and nonfatal lactic acidosis with metformin use in type 2 diabetes
32. Svare A. A patient presenting with symptomatic hypomagnese- mellitus (Review). Cochrane Database Syst Rev. 2010;(4):1-235.
mia caused by metformin-induced diarrhea: a case report. 52. Spiller HA, Quandrani DA. Toxic effects from metformin expo-
Cases J. 2009;2:156. sure. Ann Pharmacother. 2004;38(5):776-780.
33. Bouchoucha M, Uzzan B, Cohen R. Metformin and digestive 53. Bronstein AC, Spyker DA, Cantilena LR, Green JL, Rumack BH,
disorders. Diabetes Metab. 2011;37(2):90-96. Giffin SL. 2009 Annual Report of the American Association of
34. Mazokopakis EE, Starakis Ik. Recommendations for diagnosis Poison Control Center’s National Poison Data System (NPDS):
and management of metformin-induced vitamin B12 (Cbl) defi- 27th Annual Report. Clin Toxicol (Phila). 2003;48(10):979-1178.
ciency. Diabetes Res Clin Pract. 2012;97(3):359-367. 54. Bronstein AC, Spyker DA, Cantilena LR, Green JL, Rumack BH,
35. Pierce SA, Chung AH, Black KK. Evaluation of vitamin B12 Dart RC. 2010 Annual Report of the American Association of
monitoring in a veteran population on long-term, high-dose met- Poison Control Center’s National Poison Data System (NPDS):
formin therapy. Ann Pharmacother. 2012;46(11):1470-1476. 28th Annual Report. Clin Toxicol (Phila). 2011;49(10):910-941.
36. Iftikhar R, Kamran SM, Quadir A, et al. Prevalence of vitamin 55. Bronstein AC, Spyker DA, Cantilena LR, Rumack BH, Dart RC.
B12 deficiency in patients of type 2 diabetes mellitus on metfor- 2011 Annual Report of the American Association of Poison Con-
min: a case control study from Pakistan. Pan Afr Med J. 2013;16: trol Center’s National Poison Data System (NPDS): 29th Annual
67. Report. Clin Toxicol (Phila). 2012;50(10):911-1164.
37. Sato Y, Ouchi K, Funase Y, et al. Relationship between 56. Mowry JM, Spyker DA, Cantilena LR, Bailey JE, Ford M. 2012
metformin use, vitamin B12 deficiency, hyperhomocysteinemia, Annual Report of the American Association of Poison Control
and vascular complications in patients with type 2 diabetes. Center’s National Poison Data System (NPDS): 30th Annual
Endocr J. 2013;60(12):1275-1280. Report. Clin Toxicol (Phila). 2013;51(10):949-1229.
38. Jouela AJ, Pirttiaho H, Palva IP. Drug-induced malabsorption of 57. Mowry JM, Spyker DA, Cantilena LR, McMillan N, Ford M.
vitamin B12. VI. Malabsorption of vitamin B 12 during treatment 2013 Annual Report of the American Association of Poison Con-
with phenformin. Acta Med Scand. 1974;196(4):267-269. trol Center’s National Poison Data System (NPDS): 30th Annual
39. Adams JF, Clark JS, Ireland JT, et al. Malabsorption of vitamin Report. Clin Toxicol (Phila). 2014;52(10):1032-283.
B12 and intrinsic factor secretion during biguanide therapy. Dia- 58. Bergman U, Boman G, Wiholm BE. Epidemiology of adverse
betologia. 1983;42(1):16-18. drug reactions to phenformin and metformin. Br Med J. 1978;
40. Berger W. Incidence of severe side effects during therapy with sulfo- 2(6135):464-466.
nylureas and biguanides. Horm Metab Res Suppl. 1985;15:111-114. 59. Kreisberg RA, Wood BC. Drug and chemical-induced metabolic
41. Blum A, Ghaben W, Slonimsky G, et al. Metformin-induced acidosis. Clin Endocr Metab. 1983;12(2):391-411.
hemolytic anemia. Isr Med Assoc J. 2011;13(7):444-445. 60. Luft D, Schmulling RM, Eggstein M. Lactic acidosis in
42. Lin KD, Lin JD, Juang JH. Metformin-induced hemolysis with biguanide-treated diabetics: a review of 330 cases. Diabetologia.
jaundice. N Engl J Med 1998;339(25):1860-1861. 1978;14(2):75-87.
43. Ruggiero NA, Kish TD, Lee ML. Metformin-induced hemolytic 61. Davis EJ, Bremer J, Akerman KE. Thermodynamic aspects of
anemia in a patient with glucose-6-phosphate dehydrogenase translocation of reducing equivalents by mitochondria. J Biol
deficiency. Am J Ther. 2016;23(2):e575-e578. Chem. 1980;255(6):2277-2283.
12 Journal of Intensive Care Medicine XX(X)

62. LaNoue KF, Tischler ME. Electrogenic characteristics of the 81. Dennis SC, Gevers W, Opie LH. Protons in ischemia: where do
mitochondrial glutamate-aspartate antiporter. J Biol Chem. they come from; where do they go to? J Mol Cell Cardiol. 1991;
1974;249(23):7522-7528. 23:1077-1086.
63. Murphy E, Coll KE, Viale RO, Tischler ME, Williamson JR. 82. Noakes TD. 1996 J.B. Wolffe Memorial Lecture. Challenging
Kinetics and regulation of the glutamate-aspartate translocator beliefs: ex Africa semper aliquid novi. Med Sci Sports Exerc.
in rat liver mitochondria. J Biol Chem. 1979;254(17):8369-8376. 1997;29(5):571-590.
64. Mitchell P. Coupling of phosphorylation to electron and hydrogen 83. Protti A, Fortunato F, Monti M, et al. Metformin overdose, but not
transfer by a chemi-osmotic type of mechanism. Nature. 1961; lactic acidosis per se, inhibits oxygen consumption in pigs. Crit
191:144-148. Care. 2012;16(3):R75.
65. Robergs RA, Ghiasvand F, Parker D. Biochemistry of exercise- 84. Ball S, Woods HF, Alberti KG. Lacticacidosis, ketoacidosis,
induced metabolic acidosis. Am J Physiol Regul Integr Comp and hyperalaninaemia in a phenformintreated diabetic patient.
Physiol. 2004;287(3):R502-R516. Br Med J. 1974;4(5946):699-700.
66. Madiraju AK, Erion DM, Rahimi Y, et al. Metformin suppresses 85. Cohen RD, Ward JD, Brain AJ, et al. The relation between phen-
gluconeogenesis by inhibiting mitochondrial glycerophosphate formin therapy and lactic acidosis. Diabetologia. 1973;9(1):
dehydrogenase. Nature. 2014;510(7506):542-546. 43-46.
67. Bridges HR, Jones AJ, Pollak MN, Hirst J. Effects of metformin 86. Protti A, Russo R, Tagliabue P, et al. Oxygen consumption is
and other biguanides on oxidative phosphorylation in mitochon- depressed in patients with lactic acidosis due to biguanide intox-
dria. Biochem J. 2014;462(3):475-487. ication. Crit Care. 2010;14(1):R22.
68. El-Mir MY, Nogueira V, Fontaine E, Avéret N, Rigoulet M. 87. Al-Abri SA, Hayashi S, Thoren KL, Olson KR. Metformin
Dimethylbiguanide inhibits cell respiration via an indirect effect overdose-induced hypoglycemia in the absence of other antidia-
targeted on the respiratory chain complex I. J Biol Chem. 2000; betic drugs. Clin Toxicol (Phila). 2013;51(5):444-447.
275(1):223-228. 88. Kruger FA, Skillman TG, Hamwi GJ, Grubbs RC, Danforth N.
69. Owen MR, Doran E, Halestrap AP. Evidence that metformin The mechanism of action of hypoglycemic guanidine derivatives.
exerts its anti-diabetic effects through inhibition of complex 1 Diabetes. 1960;9:170-173.
of the mitochondrial respiratory chain. Biochem J. 2000;348(pt 89. Unger RH, Madison LL, Carter NW. Tolbutamide-phenformin in
3):607-614. ketoacidosis-resistant patients. JAMA. 1960;174:2132-2136.
70. Johnson HK, Waterhouse C. Lactic acidosis and phenformin. 90. Jangaard NO, Pereira JN, Pinson R. Metabolic effects of the
Arch Intern Med. 1968;122(4):367-370. biguanides and possible mechanism of action. Diabetes. 1968;
71. Mestman JH, Pocock DS, Kirchner A. Lactic acidosis with recov- 17(2):96-104.
ery in diabetes mellitus on phenformin therapy. Calif Med. 1969; 91. Fontaine E. Metformin and respiratory chain complex I: the last
111(3):181-185. piece of the puzzle? Biochem J. 2014;463(3):e3-e5.
72. Bengtsson K, Karlberg B, Lindgren S. Lactic acidosis in 92. Kruse JA. Metformin-associated lactic acidosis. J Emerg Med.
phenformin-treated diabetics. A clinical and laboratory study. 2001;20(3):267-272.
Acta Med Scand. 1972;191(3):203-208. 93. Bruijestens LA, Luin MV, Buscher-Jungerhans PM, Bosch FH.
73. Assan R, Heuclin C, Girard JR, LeMaire F, Attali JR. Reality of severe metformin-induced lactic acidosis in the
Phenformin-induced lactic acidosis in diabetic patients. Diabetes. absence of chronic renal impairment. Neth J Med. 2008;66(5):
1975;24(9):791-800. 185-190.
74. Dembo AJ, Marliss EB, Halperin ML. Insulin therapy in 94. Von Mach MA, Sauer O, Weilemann LS. Experiences of a poison
phenformin-associated lactic acidosis; a case report, biochemical center with metformin-associated lactic acidosis. Exp Clin Endo-
considerations and review of the literature. Diabetes. 1975;24(1): crinol Diabetes. 2004;112(4):187-190.
28-35. 95. Spiller HA, Weber JA, Winter ML, Klein-Schartz W, Hofman M.
75. Baur JA, Birnbaum MJ. Control of gluconeogenesis by metfor- Multicenter case series of pediatric metformin ingestion. Ann
min: does redox trump energy charge? Cell Metab. 2014;20(2): Pharmacother. 2000;34(12):1385-1388.
197-199. 96. Lacher M, Hermanns-Clausen M, Haeffner K, Brandis M, Pohl
76. Marliss EB, Aoki TT, Toews CJ, et al. Amino acid metabolism in M. Severe metformin intoxication with lactic acidosis in an ado-
lactic acidosis. Am J Med. 1972;52(4):474-481. lescent. Eur J Pediatr. 2015;164(6):362-365.
77. Gevers W. Generation of protons by metabolic processes in heart 97. Gjedde S, Christiansen A, Pedersen SB, Rungby J. Survival fol-
cells. J Mol Cell Cardiol. 1977;9(11):867-874. lowing a metformin overdose of 63 g: a case report. Pharmacol
78. Zilva JF. The origin of the acidosis in hyperlactataemia. Ann Clin Toxicol. 2003;93(2):98-99.
Biochem. 1978;15(1):40-43. 98. Dell’Anglo DM, Perino LJ, Todino JD, Algren A, Morgan BW.
79. Hochachka PW, Dunn JF. Metabolic arrest: the most effective Metformin overdose with a resultant serum pH of 6.59: survival
means of protecting tissues against hypoxia. Prog Clin Biol Res. without sequalae. J Emerg Med. 2010;39(1):e77-e80.
1983;136:297-309. 99. Kopec KT, Kowalski MJ. Metformin-associated lactic acidosis
80. Busa WB, Nuccitelli R. Metabolic regulation via intracellular pH. (MALA): case files of the Einstein Medical Center medical tox-
Am J Physiol. 1984;246(4 pt 2):R409-R438. icology fellowship. J Med Toxicol. 2013;9:61-66.
Wang and Hoyte 13

100. Ahmad S, Beckett M. Recovery from pH 6.38: lactic acidosis metformin-induced hyperlactatemia without metformin accu-
complicated by hypothermia. Emerg Med J. 2002;19(2): mulation. Diabetes Metab. 2014;40(3):220-223.
169-171. 119. Lalau JD, Lemaire-Hurtel AS, Lacroix C. Establishment of a
101. McNamara K, Isbister GK. Hyperlactatemia and clinical sever- database of metformin plasma concentrations and erythrocyte
ity of acute metformin overdoses. Intern Med J. 2015;45(4): levels in normal and emergency situations. Clin Drug Investig.
402-408. 2011;31(6):435-438.
102. Suchard JR, Grotsky TA. Fatal metformin overdose presenting 120. Vecchio S, Giampreti A, Petrolini VM, et al. Metformin accu-
with progressive hyperglycemia. West J Emerg Med. 2008;9(3): mulation: lactic acidosis and high plasma metformin levels in a
160-165. retrospective case series of 66 patients on chronic therapy. Clin
103. Forrester MB. Adult metformin ingestions reported to Texas Toxicol (Phila). 2014;52(2):129-135.
poison control centers, 2000-2006. Hum Exp Toxicol 2008; 121. Dell’Anglio DM, Perino LJ, Kazzi Z, et al. Acute metformin
27(7):575-583. overdose: examining serum pH, lactate level, and metformin
104. Quaile MP, Melich DH, Jordan HL, Nold JB. Toxicity and tox- concentrations in survives versus nonsurvivors: a systematic
icokinetics of metformin in rats. Toxicol Appl Pharmacol. 2010; review of the literature. Ann Emerg Med. 2009;54(6):818-823.
243(3):340-347. 122. Seidowsky A, Nseir S, Hodret N, Fourrier F. Metformin-
105. Misbin RI, Green L, Stadel BV, et al. Lactic acidosis in patients associated lactic acidosis: a prognostic and therapeutic study.
with diabetes treated with metformin. N Engl J Med. 1998; Crit Care Med. 2009;37(7):2191-2196.
338(4):256-256. 123. Peters N, Jay N, Barroud D, et al. Metformin-associated lactic
106. Misbin RI. The phantom of lactic acidosis due to metformin in acidosis in an intensive care unit. Crit Care. 2008;12(6):R149.
patients with diabetes. Diabetes Care. 2004;27(7):1791-1793. 124. Akinci B, Yener S, Bengi G, Yesil S. Alterations of coagulation in
107. Lalau JD, Race JM. Lactic acidosis in metformin therapy: metformin intoxication. Hormones (Athens). 2008;7(4):325-329.
searching for a link with metformin in reports of ‘metformin 125. Lalau DJ, Race RL. Lactic acidosis in metformin-treated
associated lactic acidosis.’ Diabetes Obes Metab. 2001;3(3): patients. Prognostic value of arterial lactate levels and plasma
195-201. metformin concentrations. Drug Saf. 1999;20(4):377-384.
108. Bodmer M, Meier C, Krahenburl S, Jick SS, Meier CR. Metfor- 126. Stades AME, Heikens JT, Erkelens DW, et al. Metformin and
min, sulfonylureas, or other antidiabetes drugs and the risk of lactic acidosis: cause or coincidence? A review of case reports.
lactic acidosis or hypoglycemia: a nested case-control analysis. J Int Med. 2004;255:179-187.
Diabetes Care. 2008;31(11):2086-2091. 127. Kajbaf F, Lalau JD. The prognostic value of blood pH and lactate
109. Stades AM, Heikens JT, Ertkelens DW, Hlleman F, Hoeskstra B. and metformin concentrations in severe metformin-associated lac-
Metformin and lactic acidosis: cause or coincidence? A review tic acidosis. BMC Pharmacol Toxicol. 2013;14:22-27.
of case reports. J Intern Med. 2004;255(2):179-187. 128. Guo PYF, Storsely LJ, Finkel SN. Severe lactic acidosis treated
110. Vecchio S, Protti A. Metformin-induced lactic acidosis: no one with prolonged hemodialysis: recovery after massive overdoses
left behind. Crit Care. 2011;15(1):107. of metformin. Semin Dial. 2006;19(1):80-83.
111. Perrone J, Phillips C, Gaieski D. Occult metformin toxicity in 129. Arroyo AM, Walroth TA, Mowry JB, Kao LW. The MALAdy of
three patients with profound lactic acidosis. J Emerg Med. 2011; metformin poisoning: is CVVG the cure? Am J Ther. 2010;
40(3):271-275. 17(1):96-100.
112. Shenoy C. Metformin-associated lactic acidosis precipitate by 130. Wen YK. Impact of acute kidney injury on metformin-
acute renal failure. Am J Med Sci. 2006;331(1):55-57. associated lactic acidosis. Int Urol Nephrol. 2009;41:967-972.
113. Vander T, Hallevy H, Ifergane G, Herishanu YO. Metformin- 131. Finkle SN. Should dialysis be offered in all cases of metformin-
induced encephalopathy without lactic acidosis. Diabet Med. associated lactic acidosis? Crit Care. 2009;13(1):1-2.
2004;21(2):194-195. 132. Lalau JD, Westeel PF, Debussche X, et al. Bicarbonate haemo-
114. Jung EY, Cho HS, Seo JW, et al. Metformin-induced encephalo- dialysis: an adequate treatment for lactic acidosis in diabetics
pathy without lactic acidosis in a patient with a contraindication treated by metformin. Intensive Care Med. 1987;13(6):383-387.
for metformin. Hemodial Int. 2009;13(2):172-175. 133. Lalau JD, Andrejak M, Morineier P, et al. Hemodialysis in the
115. Kang YJ, Bae EJ, Seo JW, et al. Two additional cases of treatment of lactic acidosis in diabetics treated by metformin: a
metformin-associated encephalopathy in patients with end- study of metformin elimination. Int J Clin Phamracol Ther Tox-
stage renal disease undergoing hemodialysis. Hemodial Int. icol. 1989;27(6):285-288.
2013;17(1):111-115. 134. Alivanis P, Giannikouris L, Paliuras C, et al. Metformin-
116. Fernandes GC, Koltermann T, Campos L, Vedolin L, Rieder CR. associated lactic acidosis treated with continuous renal replace-
Teaching NeuroImages: the lentiform fork sign: an MRI pattern of ment therapy. Clin Ther. 2006;28(3):396-400.
metformin-associated encephalopathy. Neurology. 2015;84(3):e15. 135. Arroyo AM, Walroth TA, Morwy JB, Kao LW. The MALAdy of
117. Zitzmann S, Reimann IR, Schmechel H. severe hypoglycemia in metformin poisoning: is CVVH the cure? Am J Ther. 2010;
an elderly patient treated with metformin. Int J Clin Pharmcol 17(1):96-100.
Ther. 2002;40:108-110. 136. Barrueto F, Meggs WJ, Barchman MJ. Clearance of metformin
118. Lalau JD, Azzoug ML, Kajbaf F, Briet C, Desailloud R. by hemofiltration in overdose. Clin Toxicol (Phila). 2002;40(2):
Metformin accumulation without hyperlactatemia and 177-180.
14 Journal of Intensive Care Medicine XX(X)

137. Dischtwald S, Weinbroum AA, Sorkine P, Ekstine MP, Daha E. 154. Cooper DJ, Walley KR, Wiggs BR, Russell JA. Bicarbonate
Metformin-associated lactic acidosis following acute kidney does not improve hemodynamics in critically ill patients who
injury. Efficacious treatment with continuous renal replacement have lactic acidosis. A prospective, controlled clinical study.
therapy. Diabet Med. 2012;29(2):245-250. Ann Intern Med. 1990;112(7):492-498.
138. Guo PY, Storsley LJ, Finkle SN. Severe lactic acidosis treated 155. Kraut JA, Kurtz I. Use of base in the treatment of severe acide-
with prolonged hemodialysis: recover after massive overdoses of mic states. Am J Kidney Dis. 2001;38(4):703-727.
metformin. Semin Dial. 2006;19(1):80-83. 156. Halperin ML, Cheema-Dhadli S, Halperin FA, Kamel KS. Ratio-
139. Heaney D, Majid A, Junor B. Bicarbonate haemodialysis as a nale for the use of sodium bicarbonate in a patient with lactic acido-
treatment of metformin overdose. Nephrol Dial Transplant. sis due to a poor cardiac output. Nephron. 1994;66(3):258-261.
1997;12(5):1046-1047. 157. Kallet RH, Jasmer RM, Luce JM, Lin LH, Marks JD. The treat-
140. Keller G, Cour M, Hernu R, et al. Management of metformin- ment of acidosis in acute lung injury with Tris-hydroxymethyl
associated lactic acidosis by continuous renal replacement ther- aminomethane (THAM). Am J Respir Crit Care Med. 2000;
apy. PLoS One. 2011;6(8):e23200. 161(4 pt 1):1149-1153.
141. Mujtaba M, Geara AS, Madhrira M, et al. Toxicokinetics of 158. Hoste EA, Colpaert K, Vanholder RC, et al. Sodium bicarbonate
metformin-associated lactic acidosis with continuous renal versus THAM in ICU patients with mild metabolic acidosis.
replacement therapy. Eur J Drug Metab Pharmacokinet. 2012; J Nephrol. 2005;18(3):303-307.
37(2):249-253. 159. Weber T, Tschernich H, Sitzwohl C, et al. Tromethamine buffer
142. Panzer U, Kluge S, Kreymann G, Wolf G. Combination of inter- modifies the depressant effect of permissive hypercapnia on myo-
mittent haemodialysis and high-volume continuous haemofiltra- cardial contractility in patients with acute respiratory distress syn-
tion for the treatment of severe metformin-induced lactic drome. Am J Respir Crit Care Med. 2000;162(4 pt 1):1361-1365.
acidosis. Nephrol Dial Transplant. 2004;19(8):2157-2158. 160. Lam N, Sekhon G, House AA. Metformin-associated lactic
143. Rifkin SI, McFarren C, Juvvadi R, Weinstein SS. Prolonged acidosis following intentional overdose successfully treated with
hemodialysis for severe metformin intoxication. Ren Fail. Tris-hydroxymethyl aminomethane and renal replacement ther-
2011;33(4):459-461. apy. Case Rep Neprhol. 2012;2012:671595.
144. Soyoral YU, Begenik H, Emre H, Aytemiz E, Ozturk M, Erkoc 161. Atamna H, Atamna W, Al-Eyd G, Shanower G, Dhahbi JM.
R. Dialysis therapy for lactic acidosis caused by metformin Combined activation of the energy and cellular-defense path-
intoxication presentation of two cases. Human Exp Toxicol. ways may explain the potent anti-senescence activity of methy-
2011;30(12):1995-1997. lene blue. Redox Biol. 2015;6:426-435.
145. Turkcuer I, Erdur B, Sari I, Yuksel A, Tura P, Yuksel S. Severe 162. Wen Y, Li W, Poteet EC, et al. Alternative mitochondrial elec-
metformin intoxication treated with prolonged haemodialyses tron transfer as a novel strategy for neuroprotection. J Biol
and plasma exchange. Eur J Emerg Med. 2009;16(1):11-13. Chem. 2011;286(18):16504-16515.
146. Kraut JA, Madias NE. Lactic acidosis. N Engl J Med. 2014; 163. Lin AL, Poteet E, Du F, et al. Methylene blue as a cerebral
371(24):2309-2319. metabolic and hemodynamic enhancer. PLoS One. 2012;7(10):
147. Sabatini S, Kurtzman NA. Bicarbonate therapy in severe meta- e46585.
bolic acidosis. J Am Soc Nephrol. 2008;20(4):692-695. 164. Yang SH, Li W, Sumien N, et al. Alternative mitochondrial
148. Wildenthal K, Mierzwiak DS, Myers RW, Mitchell JH. Effects electron transfer for the treatment of neurodegenerative diseases
of acute lactic acidosis on left ventricular performance. Am J and cancers: methylene blue connects the dots. Prog Neurobiol.
Physiol. 1968;214(6):1352-1359. 2017;157:273-291.
149. Schotola H, Toischer K, Popov AF, et al. Mild metabolic acido- 165. Livshits Z, Nelson LS, Hernandez SH, et al. Severe metformin
sis impairs the b-adrenergic response in isolated human failing toxicity: role of methylene blue and CVVHD as therapeutic
myocardium. Crit Care. 2012;16(4):R153. adjuncts. Clin Toxicol. 2010;48(6):611-612.
150. Jung B, Rimmele T, Le Goff C, et al. Severe metabolic or mixed 166. Höjer J, Westerbergh J, Edfeldt-Ugarph M, Johansson A. Methy-
acidemia on intensive care unit admission: incidence, prognosis lene blue stopped metformin-associated lactic acidosis and refrac-
and administration of buffer therapy. A prospective, multiple- tory vasodilatation. Lakartidningen. 2013;110(42):1865-1866.
center study. Crit Care. 2011;15(5):R238. 167. Heredia D, Mancl E, Sayegh B, Simpson K. Successful use of
151. Kimmoun A, Ducrocq N, Sennoun N, et al. Efficient extra- and methylene blue for hemodynamically unstable metformin toxi-
intracellular alkalinization improves cardiovascular functions in city. Chest. 2014;146(4 suppl 2):253A.
severe lactic acidosis induced by hemorrhagic shock. Anesthe- 168. Graham RE, Cartner M, Winearls J. A severe case of vasoplegic
siology. 2014;120(4):926-934. shock following metformin overdose successfully treated with
152. Huang YG, Wong KC, Yip WH, McJames SW, Pace NL. Car- methylene blue as a last line therapy. BMJ Case Rep. 2015;2015:
diovascular responses to graded doses of three catecholamines 1-5.
during lactic and hydrochloric acidosis in dogs. Br J Anaesth. 169. Plumb B, Parker A, Wong P. Feeling blue with metformin-
1995;74(5):583-590. associated lactic acidosis. BMJ Case Rep. 2013;2013:1-4.
153. Davies AO. Rapid desensitization and uncoupling of human 170. Mayer B, Brunner F, Schmidt K. Inhibition of nitric oxide synth-
beta-adrenergic receptors in an in vitro model of lactic acidosis. esis by methylene blue. Biochem Pharmacol. 1993;45(2):
J Clin Endocrinol Metab. 1984;59(3):398-405. 367-374.

You might also like