You are on page 1of 9

Clinica Chimica Acta 520 (2021) 34–42

Contents lists available at ScienceDirect

Clinica Chimica Acta


journal homepage: www.elsevier.com/locate/cca

Review

Acute aluminum phosphide poisoning: The menace of phosphine exposure


Deepak Yadav, Rajasri Bhattacharyya *, Dibyajyoti Banerjee *
Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India

A R T I C L E I N F O A B S T R A C T

Keywords: Aluminum phosphide (AlP) is a popular fumigant used widely for the safe storage of food grain. Although A1P is
Pesticides free from toxic residues, it releases phosphine which acts on mitochondrial components of almost all types of
Poisoning pests. Unfortunately A1P is also a common suicidal agent in developing countries with no known antidote. In
Antidote
addition, accidental exposure to phosphine may also occur. AlP poisoning affects cardiac and vascular tissue
Phosphine
Acetylcholinesterase
directly and can result in multiorgan system failure leading to death in severe cases. There is no specific
biomarker for diagnosing AlP poisoning and management depends on a high level of clinical suspicion. Although
acetylcholinesterase has been suggested as a surrogate biomarker of AlP exposure, there are opposing views. In
this review, we analyzed the relevant published material with emphasis on the need to recognize and explore the
use of plasma mitochondrial enzyme activity as a potential biomarker for AlP exposure.

1. Aluminum phosphide use for grain storage this phenomenon lies in its protection of stored grains.
Apart from toxicity to pests, AlP poses a threat to human life as it is a
Aluminum phosphide (AlP), commonly known as rice pill or wheat common poisoning agent.
pill, is widely used as a fumigant for storage of these grains. AlP is
available over the counter in some developing countries and is 2. Epidemiology of AlP poisoning
commonly used as an outdoor and indoor pesticide. It is also used as a
fumigant in other foodstuffs [1,2]. AlP is a common suicide pill. From the data at hand, it appears that
Highly toxic phosphine gas is released when it comes in contact with the world is facing an epidemic of AlP poisoning. Pesticide poisoning is a
water or with an acidic environment. The availability of phosphine is public health problem, particularly in the developing world. In partic­
directly proportional to humidity [3,4]. Phosphine is slow acting and ular, AlP poisoning is critical because 500 mg AlP tablets containing
effective at low concentrations if the exposure time is prolonged [5]. 56% of the chemical are lethal to human life. Exposure to 400–600 ppm
AlP is a popular fumigant for food storage for the following reasons: phosphine gas from any source is life-threatening [8]. Some highlights of
• Free of toxic residues or leaves little residue on grains or foodstuffs the epidemiological data are provided in Table 1.
• Does not affect seed viability. Apart from suicide attempts, accidental phosphine exposure is
• No harmful effects on the ecosystem, atmosphere or food grains prevalent among workers in grain stores and the chemical industry
and prevents significant crop losses [1,6–14]. [18,19]. It also occurs during grain transportation in ships [20]. In low-
Therefore, AlP will continue to play a role in grain storage and income countries, AlP fumigation is sometimes performed to eliminate
transport [15]. bed bugs. It may cause accidental phosphine exposure [21]. The lives of
Phosphine spreads quickly. It readily diffuses into the environment pet animals such as horses are sometimes in danger due to phosphine
through air space present between stored grains and acts on the respi­ exposure from AlP [22]. Phosphine is heavier than air, but it can travel a
ratory system of insects [16]. There is considerable current interest in certain distance after release. Therefore, proximity to gas once released
understanding the physical interaction of phosphine gas with stored is not always essential for unintentional phosphine exposure (both
grains and its pesticide action. Mathematical models on the subject are accidental and homicidal) [23]. It is believed that this phenomenon is a
proposed [17]. severe issue related to AlP-induced phosphine release. There is potential
Phosphine is highly toxic to aerobically respiring organisms. It does for accidental phosphine exposure from grain storehouses in the case of
not affect anaerobic or metabolically dormant plants. The advantage of phosphine leakage. A similar kind of incident has been reported in flats

* Corresponding authors.
E-mail addresses: bdr.rajasri@yahoo.in (R. Bhattacharyya), dibyajyoti5200@yahoo.co.in (D. Banerjee).

https://doi.org/10.1016/j.cca.2021.05.026
Received 13 October 2020; Received in revised form 19 May 2021; Accepted 24 May 2021
Available online 30 May 2021
0009-8981/© 2021 Elsevier B.V. All rights reserved.
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

Table 1 phosphine gas is highly toxic to insects, burrowing pests, and other
Some facts and figures showing epidemiology of acute AlP poisoning. forms of animal life [29]. In this context, there is concern regarding the
Continent Country Year Cases emergence of phosphine-resistant pests. This issue has been aggravated
primarily due to the lack of alternative fumigants [30]. Fortunately,
Europe Denmark – Poisoning is rare [42]
Germany 1983–2003 188 [42] phosphine resistance does not confer cross-resistance to other fumigants
France – Poisoning is rare [42] [31]. Therefore, in pests with strong phosphine resistance, coadminis­
United 1997–2003 93 [42] tration of other fumigants is recommended [32]. This is indeed a
Kingdom dangerous trend, as it can result in the reintroduction of non-ecofriendly
Asia Iran The death rate is high [114]
fumigants. Therefore, the time is ripe for intense research into alterna­

Sri Lanka – The death rate is high [114]
Saudi 2006–2015 68 [115] tive fumigants that are equally as efficient as phosphine but less toxic.
Arabia 2000–2007 471 [42] The concept of bio-fumigation is in its infancy [33,34]. We feel that it is
India 2018 35,862 total poisoning suicide cases a promising concept, but a great deal of research is required to identify
[116] and majority of poisoning cases
an approach that can compete with AlP at this time. Nevertheless, we
due to aluminum phosphide [117]
should continue learning about phosphine toxicity, as another effective
fumigant is not presently at hand.
Apart from toxicity to pests, AlP poses a threat to human life, and the
Africa Morocco – The death rate is high [114] exact mechanism described above occurs in the case of accidental or
(– Data not available). suicidal exposure to phosphine.

where AlP fumigation was performed in the basement for storage [24]. 3.2. Mechanism of toxicity in human beings

3. Mechanisms of toxicity The exact mechanism of action of phosphine in humans that causes
lethality is yet to be determined. Nevertheless, phosphine blocks cellular
3.1. Mechanism of toxicity in pests and rodents respiration at the mitochondrial level. It leads to the formation of highly
reactive hydroxyl radicals and other reactive oxygen species (ROS) and
The mechanism of phosphine toxicity has been investigated in pests causes severe oxidative stress. Phosphine also inhibits oxygen uptake,
and rodents in detail. Phosphine inhibits enzymes of the electron which causes tissue injury. There may be direct adrenal and cardiac
transport chain (ETC) in insect and rodent mitochondria. It disrupts myocyte toxicity and consequent decreased blood pressure. Further­
energy metabolism and mitochondrial function. Most in vitro studies more, phosphine is associated with less perfusion to vital organs. There
point towards the inhibition of cytochrome C oxidase and other ETC is a decrease in overall ATP (adenosine triphosphate) production. Dur­
enzymes [25]. Phosphine acts at complex IV of the ETC to inhibit elec­ ing severe poisoning, multiorgan system failure occurs [35]. Phosphine
tron flow [26]. Respiration is inhibited by phosphine in insect and rat reduces ferric iron to ferrous iron and releases iron from its protein-
liver mitochondria [26,27]. Catalase activity and gene expression are bound form. This phenomenon has the potential to aggravate injury
also reduced. Furthermore, phosphine inhibits glucose-6-phosphate due to oxidative stress [36]. All these mechanisms account for the
dehydrogenase activity and reduces the glutathione concentration toxicity of phosphine.
[28]. Cellular hypoxia is induced, which leads to the formation of highly Notably, in in vitro studies, phosphine was shown to be an ETC poi­
reactive hydroxyl radicals (Fig. 1a). For the reasons mentioned above, son. In in vivo studies, direct evidence of ETC inhibition was not

Fig. 1a. Toxicity by oxidative stress: Aluminum


phosphide in the presence of moisture and/or
gastric hydrochloric acid release phosphine gas.
Phosphine inhibits the catalase, cytochrome c oxi­
dase (ETC) and glucose-6-phosphate dehydrogenase
(G6PD) activity. G6PD inhibition reduces gluta­
thione concentration. All these generate ROS. It
disrupts the mitochondrial function so, there is a
decrease in overall ATP production. Phosphine de­
creases oxygen uptake. This phenomenon leads to
hypoxia. The injury in AlP poisoning is a combina­
torial effect of hypoxia and oxidative stress that may
lead to multiorgan failure.

35
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

Fig. 1b. Toxicity by reducing property of PH3: Phosphine reduces the protein-bound Fe+ 3 to Fe2 and releases the protein-bound iron. This phenomenon, along with
+

ROS, accelerates oxidative damage. It breaks the disulfide bonds of the proteins. For example, breakage of the disulfide bond of insulin may cause altered insulin
function and hyperglycemia.

observed. However, phosphine inhibits several enzymes, such as cata­ authors recommend investigation along these lines of research because
lase, peroxidase, and glycerol-phosphate dehydrogenase. Additionally, apart from oxidative stress, destruction of the structure of vital proteins
it reduces the glutathione concentration. Therefore, the toxicity of may be a possible mechanism underlying the lethality of phosphine
phosphine is mostly due to oxidative stress-induced cellular injury [25]. exposure. The major mechanisms of AlP toxicity are pictorially repre­
Apart from the inhibition of antioxidant enzymes and production of sented in Figs. 1a & 1b.
ROS due to inhibition of ETC, phosphine has been shown to react with
hydrogen peroxide and generate ROS. As stated earlier, phosphine re­ 4. Clinical features of AlP poisoning
duces iron. It can aggravate the generation of lethal ROS from hydrogen
peroxide (such as hydroxyl radicals) in the presence of phosphine. In Poisoning may be due to direct AlP ingestion or inhalation of phos­
vitro, phosphine causes peroxidation of lipids, which supports the above phine released from AlP [42]. After ingestion, AlP releases harmful
findings [36]. Since hydrogen peroxide is abundantly present in the phosphine gas in the stomach. Gastric HCl favors this release. Phosphine
human body, it has been reported that the chemical reaction of phos­ is rapidly absorbed throughout the gastrointestinal tract (GIT). Acci­
phine with hydrogen peroxide in the presence of iron in vivo is a major dental phosphine exposure is also lethal to humans, as the lungs rapidly
mechanism of oxidative tissue injury in humans in the case of AlP absorb it.
poisoning [37]. Focused research in this direction is recommended, and Phosphine mainly affects the cardiac and vascular tissues. Specif­
if the above hypothesis is proven to be true, iron chelators can attenuate ically, phosphine is directly cardiotoxic; it induces cellular respiration
phosphine toxicity. inhibition and depresses the aerobic metabolism of heart muscles [43].
The reducing property of phosphine can affect the human body in This leads to congestive heart failure, myocarditis, conduction abnor­
numerous ways. Phosphine can reduce disulfide bonds [38]. The malities, hypotension, pericarditis and subendocardial infarction [44].
reduction of disulfide bonds in several proteins will alter their structure. The signs and symptoms of AlP are nonspecific and depend on the
Insulin is a peptide that has disulfide bonds, and insulin receptor also has dose, method of exposure and time since exposure to the poison. After
disulfide bonds [39]. If phosphine reduces the disulfide bonds of insulin accidental exposure to AlP, patients feel breathlessness, tightness in the
or its receptor, glucose homeostasis will be abnormal [40]. Hypergly­ chest and headache [45]. The release of lethal phosphine gas from the
cemia is reported following phosphine exposure, but it is due to the AlP primarily affects most of our body’s vital organs, including the
reduction of the disulfide bond of insulin or its receptor [41]. The heart, lungs, GIT, and kidneys [43,45]. Other clinical features of acute

36
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

AlP exposure include nausea, vomiting, jaundice, diarrhea, ataxia, while dealing with AlP poisoning are as follows:
paraesthesia, tremor, muscle weakness and diplopia [1]. • Inhibition of phosphine absorption through GIT and enhancement
In the case of mild ingestion of AlP, acute features generally devel­ of phosphine excretion through the lungs and kidneys
oped within 1–2 h of poisoning [46]. In moderate to severe cases of oral • Prevention of the release of phosphine from AlP
poisoning, almost all patients develop acute features immediately after • Attenuation of organ toxicity
ingestion, including vomiting, nausea, sweating, hypotension, retro­ • Supportive measures are necessary as soon as possible [68].
sternal burning, tachycardia, shock and coma [47]. In the case of shock, Gastric lavage with magnesium sulfate (MgSO4) or potassium per­
plasma renin activity is often observed to be elevated. This correlates manganate (KMnO4) (1:10,000) is widely used [44]. Some studies have
with the dose of the poison consumed [48]. also highlighted the use of sodium bicarbonate (NaHCO3) and coconut
The unusual complications of acute AlP exposure include atrial oil for gastric lavage [69]. The combination of KMnO4, NaHCO3 and
infarction, hepatic toxicity, skeletal muscle damage, ascites and renal MgSO4 in the lavage formulation oxidizes and neutralizes gastric HCl.
failure [49]. Consistent with expectations, a case report study showed This inhibits the release of phosphine from AlP. Coconut oil forms a
that there was an increase in cardiac enzyme markers such as creatine protective layer over the gastric mucosa, preventing the absorption of
kinase-MB and troponin T [50,51]. phosphine [68]. Activated charcoal and antacids adsorb and reduce the
Patients show respiratory features such as cough, cyanosis, dyspnea, absorption of phosphine from GIT, respectively [1]. Liquid paraffin is
pulmonary edema and respiratory failure [45]. Elevation of blood also used for the same purpose [70]. Melatonin administration has been
cortisol with corresponding changes in adrenal gland histopathology is suggested as a potential approach for managing cardiotoxicity, as it
seen [52]. In cases of mild poisoning, patients usually show slow re­ scavenges ROS, accelerates ATP production and prevents apoptosis by
covery, but death is the rule in severe poisoning [53]. inhibiting cytochrome c release [71]. Goharbari et al., 2017 suggested
AlP poisoning can have exceptional atypical presentations, and pri­ the oral administration of liothyronine post gastric lavage as a promising
mary care physicians should have a high suspicion of phosphine expo­ adjuvant therapy for AlP toxicity. This treatment enhances systolic
sure in appropriate cases. This high suspicion is necessary as there is no blood pressure, catalase activity, and total thiol groups and improves
established biomarker of AlP poisoning. There can be a wide range of total antioxidant capacity [72]. The most critical factor for the suc­
complications of AlP poisoning. For example, a patient may have pre­ cessful treatment of AlP toxicity is the resuscitation of shock. Various
dominantly pleural effusion. Others may present as acute renal failure. measures have been suggested for this, such as administering normal
Some other patients may develop pancreatitis. saline intravenously to keep central venous pressure at approximately
The clinical features profoundly differ in these cases [49]. Among 12–14 cm of water. Low-dose dopamine and hydrocortisone have been
them, hemolysis and methemoglobinemia may complicate the course of recommended to keep systolic blood pressure above 90 mm Hg and
AlP poisoning [54]. Severe hypoglycemia may be observed occasionally combat shock [70]. The SPO2 level is maintained by delivering 100%
[55]. oxygen through a face mask, which further helps in managing ARDS. In
Polyserositis occurred in a case series. The patients developed pleural a case study of a 21-year-old man with AlP poisoning, a combination of
effusions, pericardial effusion and ascites. Mitochondrial damage in the glucagon and digoxin with antioxidant agents successfully treated the
endothelium leads to capillary leakage syndrome, which accounts for patient [73].
this condition’s possible mechanism [56]. The delayed complication of As stated above, there is no specific antidote available for AlP
polyserositis has been associated with or without acute respiratory poisoning; however, various substances have been proposed to achieve a
distress syndrome (ARDS) [57]. better outcome and supportive care [74–82]. The effects of these mea­
Other atypical symptoms of AlP poisoning reported from time to time sures vary from patient to patient. The outcome mainly depends upon
may include drowsiness, cold body, hypotonicity, pale skin, abdominal the route of exposure, duration of exposure, amount of poison, avail­
pain and loose motion [58,59]. A hypotensive state with reduced SPO2 ability of supportive measures and time interval between consumption
levels is observed. ECG findings can be associated with ventricular and resuscitation.
ectopic and atrial fibrillation following AlP exposure [60]. Intravascular
hemolysis occurs in patients with glucose-6-phosphate dehydrogenase 6. Diagnosis:
enzyme deficiency [61]. Methemoglobinemia, acute pancreatitis,
esophagobronchial fistula, restlessness, tachypnea, altered sensorium, There is no biomarker recommended for the diagnosis of AlP
peripheral cyanosis, and cold sweaty skin may predominate in some poisoning. Some basic principles can be used as a diagnostic tool to some
cases of AlP poisoning [58,62–63]. extent. The diagnosis of AlP poisoning is made mainly by clinical
Spontaneous self-ignition is also a rarely observed physical phe­ suspicion.
nomenon in the case of AlP poisoning. This may occur due to the
contribution of released phosphine coupled with heat and negative 1. Clinical history obtained from the patient or attendants is often
pressure generated by suction [64]. Dramatic hot vomiting is reported helpful in disclosing the substance used for suicide [47].
after potassium permanganate gastric lavage following AlP ingestion 2. Some symptoms may rarely be present following acute AlP exposure.
[65]. These include vomitus smelling of decaying garlic or fish, shock,
hypotension and abnormalities in cardiac rate [47].
5. Management of AlP poisoning: 3. The silver nitrate test is not entirely reliable but is proposed to detect
phosphine either from the stomach content or directly from the pa­
To date, there is no specific antidote for AlP poisoning, and in the tient’s breath. Exposure to phosphine converts freshly prepared sil­
absence of an antidote, the only option is supportive treatment [66]. ver nitrate-dipped filter paper to a blackish color precipitate. There
With time, various experimental and clinical investigations have pro­ are chances of false negatives in patients receiving oxygen, which
posed substances that can counteract AlP toxicity. However, active converts phosphine into phosphorus pentoxide, and false-positive
research in the field is ongoing [35]. results due to hydrogen sulfide in the air [47]. For phosphine
The current treatment aims to provide supportive care to patients detection in gastric samples, the silver nitrate test is generally used as
until phosphine is excreted from the GIT, kidneys and lungs. In the case a screening test. This conclusion is confirmed by other sophisticated
of accidental exposure to phosphine, the first step is to bring the patient tests, such as gas chromatographic phosphine detection [83]. The
into fresh air, whereas in the case of ingestion, decontamination of the gastric lavage content in AlP poisoning may be heated to obtain the
gut is the priority [67]. phosphine liberated for detection by the silver nitrate test [15]. Apart
The basic principles proposed by researchers to be kept in mind from silver nitrate, silver diethyldithiocarbamate is used for

37
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

phosphine detection. This approach is even used for quantitative abnormality is also common in these cases. Several laboratory-based
detection of phosphine by spectrophotometry. Nevertheless, silver assessments have been reported in AlP cases where leucopenia is an
diethyldithiocarbamate has not yet been examined in hospital indicator of severe toxicity. Metabolic acidosis is seen in moderate
emergencies to detect phosphine from gastric content (or breath of toxicity. In these cases, serum SGOT (serum glutamic oxaloacetic
the patient) [84]. In this context, it is pertinent to mention that transaminase) and SGPT (serum glutamate-pyruvate transaminase) ac­
mercuric chloride impregnated strips are used to detect the presence tivity may be elevated [93]. Another study reported a direct relationship
of phosphine in the air. However, their use in clinical cases of acute between plasma renin level and mortality and the dose of AlP. Plasma
AlP poisoning has never been reported [85]. renin levels are significantly elevated, while cortisol levels are decreased
4. The availability of phosphine detector tubes and bulbs can also be in AlP toxicity [94].
used to diagnose phosphine exposure [1]. Several detector tubes are
available, mostly to detect phosphine in the environment [86]. 7. Status of acetylcholinesterase (AChE) activity in AlP
Currently, a wireless sensor approach integrated with a cloud com­ poisoning
puter is a promising technology for environmental monitoring or air
sampling of phosphine [86]. There is absolutely no experience with AlP exposure is believed by some researchers to affect AChE activity
phosphine detection using these methods in cases of AlP poisoning. as one of its primary mechanisms of toxicity [95]. In the last few de­
We believe that these technologies require urgent trials in a primary cades, much research has been conducted to examine the effect of AlP/
health care center setting to devise a point of care (POC) instrument phosphine on the level and activity of AChE. Most of the data reported
for phosphine detection for the early diagnosis of AlP. suggest that AlP inhibits various mitochondrial enzymes, but a signifi­
5. The most recent advancement is the development of gas chroma­ cant reduction in AChE activity has also been observed in serum.
tography with a nitrogen–phosphorous detector, which can detect However, some studies oppose these findings. There is no effect on
even minute amounts of phosphine [1]. In severe AlP poisoning, erythrocyte cholinesterase activity in accidental phosphine inhalation
postmortem phosphine concentrations have been detected in various cases [42]. Therefore, this controversy can be approached by consid­
tissues using gas chromatography coupled with mass spectrometry. ering two components of AlP, aluminum and phosphine, and examining
The injector temperature is approximately 180 ◦ C, and a m/z ratio of the effect of these two components on AChE activity. Nevertheless, some
34 is used for such analysis [87]. Unique chemical strategies have clinical and experimental reports suggest that both phosphine and
been applied for trace amounts of phosphine detection in postmor­ aluminum inhibit AChE [96,97].
tem samples. Phosphine is oxidized to phosphite and hypophosphite An in vitro study conducted by Mohammad J et al. in 1989 reported
in a biological system. This is reduced back to phosphine as an that the levels of AChE in mouse and human serum were significantly
analytical strategy and detected by headspace GC–MS. However, reduced after exposure to phosphine gas. This reduction was also found
antemortem use of such techniques is not yet reported from a bio­ to be correlated with the dose and exposure time [98]. Other studies
logical sample [88]. support these results by stating that AlP poisoning causes the inhibition
6. Serial blood phosphine estimation is reported in antemortem cases of of cholinesterase in rats, and repression in the treatment group was as
acute AlP poisoning [89]. Postmortem analysis of AlP poisoning high as 47% and 40% compared to the control group [95,99].
samples has revealed that blood phosphine may not be detectable. An exciting finding was pointed out in a review article by Alfred et al.
However, the blood of AlP poisoning cases may contain a high in 2016, showing that phosphine exposure decreases serum cholines­
amount of aluminum [90]. Much antemortem experience with terase activity. However, brain AChE activity was not reduced in pa­
aluminum estimation in biological samples is not reported in the tients after metal phosphide ingestion [94].
context of AlP poisoning. We believe that this is a promising The baseline value of AChE has yet to be determined. AChE activity
approach for screening AlP poisoning, and focused research should shows significant intra- and interindividual variation. Therefore, in
be carried out to address this issue. organophosphorus (OP) poisoning, AChE reactivation following oxime
treatment is considered a parameter. Reactivation induced by oximes
Phosphine is more frequently detected either in the gastric content or predicts OP exposure. AlP inhibits AChE activity in vitro. However, the in
in the breath but generally not in blood or urine samples. In these vitro inhibition is not reactivatable by oximes [100].
samples, phosphine is easily and quickly oxidized to phosphite and Therefore, we believe that AChE activity cannot currently be used as
hypophosphite in vivo [65,91]. Nevertheless, urinary phosphite and a biomarker for phosphine exposure.
hypophosphite may be estimated by chromatographic techniques [91], Phosphine can inhibit AChE [94]. This is also the case with AlP and
but such an approach has yet to be explored in a clinical case of acute aluminum ions [100,101]. Other studies from time to time have also
AlP poisoning. supported this fact and stated that aluminum inhibits brain AChE ac­
Analytical assays are more frequently conducted with postmortem tivity [102,103]. Therefore, as a solution to the controversy, the effects
samples, and in the emergency department of hospitals, such tests (such of aluminum and phosphine on serum AChE activity need to be assessed
as urine hypophosphite detection, etc.) are generally not done. In pri­ separately to achieve any conclusion about the reason for AChE inhi­
mary health care platforms, gas chromatography is usually not avail­ bition in AlP-exposed patients [104]. We believe that AChE is an
able. Therefore, there is an urgent need to determine a biomarker for essential target for intense research in the context of AlP poisoning. The
poisoning that can be detected in the practical realities of a primary behavior of the biomarker may not mimic OP compound exposure.
health center. Additionally, it may not be reactivatable by oximes [100]. Following OP
In search of a serum-based biomarker for patients who have ingested compound exposure, there are cholinergic symptoms. Both erythrocyte
AlP, Kariman et al., 2012 analyzed antioxidant capacity, total plasma cholinesterase and brain cholinesterase are inhibited [105]. However,
thiol concentration and lipid peroxidation by chromatography and with AlP exposure, the clinical picture suggests that there is peripheral
compared them with healthy controls. The results of the study were cholinesterase inhibition. There may not be cholinergic symptoms, and
similar to those of previously reported animal studies. There was a sig­ brain cholinesterase activity may not be reduced [106]. All of these
nificant increase in lipid peroxidation and a reduction in total thiol observations point towards the fact that aluminum ions are the culprit
groups and total antioxidant capacity in the AlP ingested group for AChE inhibition. It is important to emphasize that aluminum ions are
compared to the control. This confirmed that AlP significantly enhances not expected to cross the blood–brain barrier to inhibit brain cholines­
oxidative stress in the body [92]. terase activity. However, focused research is necessary in this area as
There may be acute cardiotoxicity following AlP ingestion. These nothing can be concluded with certainty.
patients often develop hypomagnesemia and hypomagnocytia. ECG

38
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

Table 2 lethal human toxicity. Additionally, AlP poisoning is a public health


A table summarizing the key points of acute AlP poisoning. problem. Therefore, apart from phosphine exposure biomarker research,
Issues Particulars we urgently need to find better alternatives to AlP for grain stores.
Active research in this direction is highly recommended and, indeed,
I. Major toxic effects of AlP i)Congestive heart failure [44]
poisoning at organ system ii)Hypotension [44] essential, as phosphine resistant pests have emerged. To meet these
level iii) Pericarditis [44] demands, work is ongoing to develop indicators of pest phosphine
iv) Subendocardial infraction [44] resistance [109]. The mechanism of phosphine resistance in pests is also
v) Tachycardia [47] an area of current research. Phosphine resistance in some pests in vitro is
vi) Shock [47]
vii) Coma [47]
a function of the antioxidant enzyme response. The increased antioxi­
viii) Abdominal pain, diarrhea, Jaundice, dant enzyme activity is likely to scavenge ROS and cause phosphine
vomiting, nausea and diplopia [1] resistance. However, all of this speculation requires more detailed
ix) Affects mainly lungs, gastrointestinal tract research in the field [110].
and kidneys [45]
The critical issues concerning acute AlP poisoning are summarized in
AlP liberates phosphine. It causes toxicity
II. Chemical reactions AlP + 3H2O → Al (OH)3 + PH3 [1] Table 2. Considering current research on the development of alternative
liberating PH3 from AlP AlP + 3HCl → AlCl3 + PH3 [1] fumigants, we believe that AlP will be used for decades to come
[111–113]. Therefore, biomarker research for AlP exposure should be a
III. Chemical reactivity of PH3 1.PH3 → forms complex with metal ion co- prioritized. We recommend focused research on mitochondrial enzyme
factors → Inhibits cytochrome c oxidase,
catalase etc. → produces ROS → oxidative stress
activities in biological fluids following AlP exposure.
→ lipid peroxidation
2. PH3 + 2 O2 → H3PO4, PO3− 4 → Non toxic
Molecular effects Declaration of Competing Interest
a) Inhibition of cytochrome c oxidase [25]
b) Cellular injury due to lipid peroxidation [35] The authors declare that they have no known competing financial
c) Decrease in level of catalase and glutathione
interests or personal relationships that could have appeared to influence
concentration [25]
d) Reduction in activity of SOD [28] the work reported in this paper.
e) Cellular hypoxia [29]
f) Release of Fe from membrane bound
transferrin receptors [36] Acknowledgement

DY acknowledges PGIMER, Chandigarh for providing fellowship.


8. Plasma mitochondrial enzymes: The proposed biomarker of DB and RB acknowledges PGIMER Chandigarh (Sanction No. 71/2-
AlP exposure Edu-16/1915) for providing financial assistance.
The authors acknowledge the language editing and formatting ser­
There is current interest in developing a POC test for the detection of vices of Dr. Sheemona Chowdhary, Research Associate, Department of
AlP poisoning. In this context, we believe that mitochondrial enzymes Experimental Medicine and Biotechnology, PGIMER, Chandigarh
should be tested for their screening potential for poisoning. The toxicity 160012 and Elsevier Web shop at the stage of finalizing the revised
of AlP is due to liberated phosphine, a mitochondrial poison [94]. manuscript.
Phosphine exposure results in swollen and dysmorphic mitochondria in
vivo [107]. Therefore, mitochondrial enzymes are expected to be leaked
References
in the extracellular site. These biomarkers should be screened for the
development of acute AlP overdose. After AlP exposure, mitochondrial [1] M. Gurjar, A. Azim, A. Baronia, K. Sharma, Managing aluminum phosphide
enzyme activities are enhanced following isolation of mitochondria poisonings, J. Emerg. Trauma Shock. 4 (2011) 378, https://doi.org/10.4103/
[94]. Therefore, phosphine may not inhibit mitochondrial enzyme ac­ 0974-2700.83868.
[2] A. Formato, D. Naviglio, G.P. Pucillo, G. Nota, Improved fumigation process for
tivities, so elevated mitochondrial enzymes in plasma/serum following stored foodstuffs by using phosphine in sealed chambers, J. Agric. Food Chem. 60
AlP exposure are expected to be detected in terms of elevated enzyme (2012) 331–338, https://doi.org/10.1021/jf204323s.
activity. [3] E. Sankarganesh, A.G. Girish, A.R. Sujeetha, A. Mariadoss, Influence of relative
humidity on phosphine concentration during aluminium phosphide (AIP)
There may be an elevation of mitochondrial enzymes in the plasma fumigation¬ in pigeon pea, Int. J. Chem. Stud. 8 (2020) 2082–2084, https://doi.
or urine samples of AlP patients. If that is the case, then this elevation org/10.22271/chemi.2020.v8.i1ae.8572.
will serve as a biomarker of AlP intoxication. In many parts of the globe, [4] H. J. Banks, Influence of water and temperature on release of phosphine from
aluminium phosphide-containing formulations, Journal of stored products
such poisoning is epidemic. However, there is no effective biomarker for research. 27 (1991) 41-56. https://doi.org/10.1016/0022-474X(91)90031-7.
screening such cases. We believe that mitochondrial enzyme activity in (accessed Apr. 2021).
plasma should be urgently investigated to develop such a biomarker [5] Manual of fumigation for insect control - Chemicals used as fumigants (cont.) -
Phosphine. http://www.fao.org/3/x5042e/x5042e0a.htm (accessed Oct. 2020).
[108]. It is important to emphasize that the mitochondrial enzymes/ [6] Y. Singh, S. Joshi, V. Satyawali, A. Gupta, Acute aluminium phosphide poisoning,
proteins that are exclusively present in mitochondria and not present in what is new? Egypt J. Int. Med. 26 (2014) 99, https://doi.org/10.4103/1110-
any other organelle and the cytosol should qualify for such investiga­ 7782.145298.
[7] Y. Zhao, S. Abbar, T.W. Phillips, M.W. Schilling, Phosphine fumigation and
tion. We list below a few such proteins/enzymes that fulfill the above
residues in dry-cured ham in commercial applications, Meat Sci. 107 (2015)
criteria and should be investigated for this purpose. 57–63, https://doi.org/10.1016/j.meatsci.2015.04.008.
1. Succinate dehydrogenase [8] A.A. Moghadamnia, An update on toxicology of aluminum phosphide, DARU J.
2. Citrate synthase Pharm. Sci. 20 (2012) 25, https://doi.org/10.1186/2008-2231-20-25. PMID:
23351193; PMCID: PMC3555759.
3. Malate dehydrogenase − 2 [9] J.B. Ristaino, W. Thomas, Agriculture, methyl bromide, and the ozone hole: can
4. Superoxide dismutase − 2 we fill the gaps? Plant Dis. 81 (1997) 964–977, https://doi.org/10.1094/
5. Coproporphyrinogen oxidase PDIS.1997.81.9.964.
[10] S. Arora, J. Stanley, C. Srivastava, Phosphine: an eco-friendly alternative for
management of wheat storage insects, Ecology (2020), https://doi.org/10.1101/
9. Summary and conclusion 2020.03.05.978486.
[11] T. Barreau, S. Hoshiko, R. Kreutzer, S. Smorodinsky, J. Talarico, Sulfuryl fluoride
poisonings in structural fumigation, a highly regulated industry—potential causes
Workers in grain storehouses are at increased risk of phosphine and solutions, IJERPH. 16 (2019) 2026, https://doi.org/10.3390/
exposure, and phosphine leakage from grain storehouses can cause ijerph16112026.

39
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

[12] A.E.R. Friedemann, L. Andernach, H. Jungnickel, D.W. Borchmann, D. Baltaci, [37] D. Banerjee, U.K. Madhusoodanan, S. Nayak, J. Jacob, Urinary hydrogen
P. Laux, H. Schulz, A. Luch, Phosphine fumigation – time dependent changes in peroxide: a probable marker of oxidative stress in malignancy, Clin. Chim. Acta
the volatile profile of table grapes, J. Hazard. Mater. 393 (2020), 122480, 334 (2003) 205–209, https://doi.org/10.1016/S0009-8981(03)00236-5.
https://doi.org/10.1016/j.jhazmat.2020.122480. [38] D.J. Cline, S.E. Redding, S.G. Brohawn, J.N. Psathas, J.P. Schneider, C. Thorpe,
[13] S. Rajendran, Status of fumigation in stored grains in India, Ind. J. f Entomol. 78 New water-soluble phosphines as reductants of peptide and protein disulfide
(2016) 28, https://doi.org/10.5958/0974-8172.2016.00022.5. bonds: reactivity and membrane permeability †, Biochemistry 43 (2004)
[14] Pesticide residues in food, (n.d.). https://www.who.int/news-room/fact-sheets/ 15195–15203, https://doi.org/10.1021/bi048329a.
detail/pesticide-residues-in-food (accessed October 13, 2020). [39] L.G. Sparrow, N.M. McKern, J.J. Gorman, P.M. Strike, C.P. Robinson, J.
[15] K.K. Garg, Review of aluminum phosphide poisoning, Int. J. Med. Sci. Public D. Bentley, C.W. Ward, The disulfide bonds in the c-terminal domains of the
Health 9 (2020) 392–400, https://doi.org/10.5455/ human insulin receptor ectodomain, J. Biol. Chem. 272 (1997) 29460–29467,
ijmsph.2020.07118202020072020. https://doi.org/10.1074/jbc.272.47.29460.
[16] Untitled, (n.d.). http://www.excelcropcare.com/product/international/ [40] S.-G. Chang, K.-D. Choi, S.-H. Jang, H.-C. Shin, Role of disulfide bonds in the
formulation/fumigents-and-rodenticide/celphos.php (accessed October 13, structure and activity of human insulin, Mol. Cells. 16 (2003) 323–330. PMID:
2020). 14744022.
[17] B.M. Plumier, M. Schramm, Y. Ren, D.E. Maier, Modeling post-fumigation [41] O. Mehrpour, M. Abdollahi, M.D. Sharifi, Oxidative stress and hyperglycemia in
desorption of phosphine in bulk stored grain, J. Stored Prod. Res. 85 (2020), aluminum phosphide poisoning, J. Res. Med. Sci. 19 (2014) 196. PMID:
101548, https://doi.org/10.1016/j.jspr.2019.101548. 24778678.
[18] A. Etemadi-Aleagha, M. Akhgari, F.S. Iravani, Aluminum Phosphide Poisoning- [42] O. Mehrpour, M. Jafarzadeh, M. Abdollahi, A systematic review of aluminium
Related Deaths in Tehran, Iran, 2006 to 2013:, Medicine. 94 (2015) e1637. phosphide poisoning, Arch. Ind. Hygiene Toxicol. 63 (2012) 61–73, https://doi.
https://doi.org/10.1097/MD.0000000000001637. org/10.2478/10004-1254-63-2012-2182.
[19] D.L. Sudakin, Occupational exposure to aluminium phosphide and phosphine [43] S.B. Lall, K. Sinha, S. Mittra, S.D. Seth, An experimental study on cardiotoxicity of
gas? A suspected case report and review of the literature, Hum Exp. Toxicol. 24 aluminium phosphide, Indian J. Exp. Biol. 35 (1997) 1060–1064. PMID:
(2005) 27–33, https://doi.org/10.1191/0960327105ht496oa. 9475040.
[20] R. Wilson, F.H. Lovejoy, R.J. Jaeger, P.L. Landrigan, Acute phosphine poisoning [44] A. Bansal, V.K. Agrawal, R.K. Singh, B.L. Kumawat, P. Mahajan, Aluminum
aboard a grain freighter. Epidemiologic, clinical, and pathological findings, phosphide poisoning: possible role of supportive measures in the absence of
JAMA. 244 (1980) 148–150. PMID: 7382074, doi:10.1001/ specific antidote, Indian, J. Crit. Care Med. 19 (2015) 109–112, https://doi.org/
jama.1980.03310020024020. 10.4103/0972-5229.151019.
[21] B. Afzal, E. Siddiqui, G.I. Kazi, I.Q. Khan, M. Daniyal, M.A.R. Khan, Phosphine [45] S. Gupta, S.K. Ahlawat, Aluminum phosphide poisoning – a review, J. Toxicol.
inhalation, case series of domestic unintentional poisoning commonly used as Clin. Toxicol. 33 (1995) 19–24, https://doi.org/10.3109/15563659509020211.
anti-bed bug chemical from a low income country, Int J Community Med Public [46] A. Goel, P. Aggarwal, Pesticide poisoning, Natl. Med. J. India. 20 (2007)
Health. 5 (2018) 3643. https://doi.org/10.18203/2394-6040.ijcmph20183113. 182–191. PMID: 18085124.
[22] A.L. Nagy, C. Cortinovis, L.J. Spicer, M.C. Perego, F. Caloni, Long-established and [47] A. Bhalla, S. Singh, Aluminum phosphide poisoning, J. Mahatma Gandhi Inst.
emerging pesticide poisoning in horses, Equine Vet Educ. 31 (2019) 496–500, Med. Sci. 20 (2015) 15, https://doi.org/10.4103/0971-9903.151721.
https://doi.org/10.1111/eve.12887. [48] S.N. Chugh, H.R. Singhal, L. Mehta, K. Chugh, V. Shankar, K.C. Malhotra, Plasma
[23] B. Krishna, C. D’Silva, Rodenticide poisoning, Indian J. Crit. Care Med. 23 (2019), renin activity in shock due to aluminium phosphide poisoning, J. Assoc.
https://doi.org/10.5005/jp-journals-10071-23318. Physicians India. 38 (1990) 398–399. PMID: 2200784.
[24] N. Al-Kandary, S. Al-Waheeb, Patterns of accidental deaths in Kuwait: a [49] B. Mostafazadeh, Aluminium phosphide poisoning, Toxicity Drug Test. (2012)
retrospective descriptive study from 2003–2009, BMC Public Health. 15 (2015) 345–361, https://doi.org/10.5772/30350.
302, https://doi.org/10.1186/s12889-015-1630-8. [50] P. Bansal, S. Giri, R. Bansal, L. Tomar, Survival in a case of aluminum phosphide
[25] N.S. Nath, I. Bhattacharya, A.G. Tuck, D.I. Schlipalius, P.R. Ebert, Mechanisms of poisoning with severe myocardial toxicity, Indian, J. Health Sci. Biomed. Res. 10
phosphine toxicity, J. Toxicol. 2011 (2011) 1–9, https://doi.org/10.1155/2011/ (2017) 343, https://doi.org/10.4103/kleuhsj.ijhs_2_17.
494168. [51] R. Katira, G.P. Elhence, M.L. Mehrotra, S.S. Srivastava, A. Mitra, R. Agarwala,
[26] N.R. Price, S.J. Dance, Some biochemical aspects of phosphine action and A. Ram, A study of aluminum phosphide (AIP) poisoning with special reference to
resistance in three species of stored product beetles, Comparative Biochemistry electrocardiographic changes, J. Assoc. Physicians India. 38 (1990) 471–473.
and Physiology Part C: Comparative Pharmacology. 76 (1983) 277–281. PMID: PMID: 2292551.
6140108 https://doi.org/10.1016/0742-8413(83)90078-6. [52] S.N. Chugh, S. Ram, A. Sharma, B.B. Arora, A.S. Saini, K.C. Malhotra,
[27] H. Nakakita, Y. Katsumata, T. Ozawa, The effect of phosphine on respiration of Adrenocortical involvement in aluminium phosphide poisoning, Indian J. Med.
rat liver mitochondria, J. Biochem. 69 (1971) 589–593, https://doi.org/10.1093/ Res. 90 (1989) 289–294. PMID: 2620956.
oxfordjournals.jbchem.a129501. [53] P. Kumar, P.M. Babu, P. Nair, null Rajeswari, S.V. Ravi, K. Sivakumar,
[28] H. Haghi Aminjan, S.R. Abtahi, E. Hazrati, M. Chamanara, M. Jalili, B. Paknejad, null Sanbakasree, M. Raveendran, A rare survival in celphos poisoning, J. Assoc.
Targeting of oxidative stress and inflammation through ROS/NF-kappaB pathway Physicians India. 66 (2018) 68–69. PMID: 30347959.
in phosphine-induced hepatotoxicity mitigation, Life Sci. 232 (2019), 116607, [54] K. Soltaninejad, L.S. Nelson, N. Khodakarim, Z. Dadvar, S. Shadnia, Unusual
https://doi.org/10.1016/j.lfs.2019.116607. complication of aluminum phosphide poisoning: Development of hemolysis and
[29] FSS | Fumigation Service & Supply Inc, fumigation, fumigant, aluminum methemoglobinemia and its successful treatment, Indian, J. Crit. Care Med. 15
phosphide, fumigation equipment, fumigations, foggers, furniture fumigation, (2011) 117–119, https://doi.org/10.4103/0972-5229.83021.
wood fumigation, grain fumigation, fumigation supplies. https://www. [55] O. Mehrpour, A. Aghabiklooei, M. Abdollahi, S. Singh, Severe hypoglycemia
fumigationzone.com/files/hb/Weevil-Cide%20Pellets%20or%20Tablets-EPA% following acute aluminum phosphide (rice tablet) poisoning; a case report and
20%2370506-13,14,15%20Label.pdf (accessed Oct. 2020). review of the literature, Acta Med. Iran. 50 (2012) 568–571. PMID: 23109032.
[30] M.K. Nayak, G.J. Daglish, T.W. Phillips, P.R. Ebert, Resistance to the fumigant [56] A. Bhalla, S. Mahi, N. Sharma, S. Singh, Polyserositis: an unusual complication of
phosphine and its management in insect pests of stored products: a global aluminum phosphide poisoning, APJMT. 1 (2012). https://doi.org/10.
perspective, Annu. Rev. Entomol. 65 (2020) 333–350, https://doi.org/10.1146/ 22038/apjmt.2012.34.
annurev-ento-011019-025047. [57] S. Koreti, Y.S. Verma, N. Prasad, G. Patel, N. Rajput, Aluminium phosphide
[31] R. Jagadeesan, M.K. Nayak, Phosphine resistance does not confer cross-resistance poisoning in children-challenges in diagnosis and management, (2014). http://
to sulfuryl fluoride in four major stored grain insect pests: lack of cross-resistance saspublisher.com/wp-content/uploads/2014/08/SAJB-28505-509.pdf (accessed
to SO 2 F 2 in PH 3 -resistant insects, Pest. Manage. Sci. 73 (2017) 1391–1401, October 13, 2020).
https://doi.org/10.1002/ps.4468. [58] S. Mirakbari, Hot charcoal vomitus in aluminum phosphide poisoning – a case
[32] R. Jagadeesan, V.T. Singarayan, K. Chandra, P.R. Ebert, M.K. Nayak, Potential of report of internal thermal reaction in aluminum phosphide poisoning and review
Co-Fumigation with Phosphine (PH3) and Sulfuryl Fluoride (SO2F2) for the of literature, Indian J. Anaesth. 59 (2015) 433, https://doi.org/10.4103/0019-
Management of Strongly Phosphine-Resistant Insect Pests of Stored Grain, 5049.160952.
J. Econ. Entomol. (2018), https://doi.org/10.1093/jee/toy269. [59] N.A. Nik Muhamad, R. Hawari, H. Shafie, A case report of aluminium phosphide
[33] M. Deb, D. Kumar, Bioactivity and efficacy of essential oils extracted from poisoning, Med. J. Malaysia. 71 (2016) 213–214. PMID: 27770125.
Artemisia annua against Tribolium casteneum (Herbst. 1797) (Coleoptera: [60] A.T. Khlaif, Aluminium phosphide poisoning-case study, Global J. adv. res. 4
Tenebrionidae): An eco-friendly approach, Ecotoxicology and Environmental (2017) 256–260 (accessed October 13, 2020), http://www.gjar.org/publish
Safety. 189 (2020) 109988. https://doi.org/10.1016/j.ecoenv.2019.109988. paper/vol4issue9/d747r51.pdf.
[34] Y. Rajashekar, A. Raghavendra, N. Bakthavatsalam, Acetylcholinesterase [61] P. Aggarwal, R. Handa, N. Wig, A. Biswas, R. Saxena, J.P. Wali, Intravascular
Inhibition by Biofumigant (Coumaran) from Leaves of Lantana camara in Stored hemolysis in aluminium phosphide poisoning, Am. J. Emergency Med. 17 (1999)
Grain and Household Insect Pests, Biomed Res. Int. 2014 (2014) 1–6, https://doi. 488–489, https://doi.org/10.1016/S0735-6757(99)90255-3.
org/10.1155/2014/187019. [62] N. Kumar, A. Mishra, A. Anand, AN unusual survival in a case of celphos
[35] A. Karimani, A.H. Mohammadpour, M.R. Zirak, R. Rezaee, B. Megarbane, poisoning, Natl J. Med. Res. 2 (2017) 518–519 (accessed October 13, 2020),
A. Tsatsakis, G. Karimi, Antidotes for aluminum phosphide poisoning – an update, https://www.bibliomed.org/mnsfulltext/78/78-1348086667.pdf?1586285831.
Toxicol. Rep. 5 (2018) 1053–1059, https://doi.org/10.1016/j. [63] A. Kumar, C.V. Singh, S. Dayal, V.K. Gupta, S. Kumar, Acute severe suicidal
toxrep.2018.10.009. poisoning by celphos powder: a rare case report from rural India, J. Indian Acad.
[36] G.B. Quistad, S.E. Sparks, J.E. Casida, Chemical model for phosphine-induced Forensic Sci. 35 (2013) 275–278.
lipid peroxidation, Pest Manage. Sci. 56 (2000) 779–783, https://doi.org/ [64] A. Wahab, M.U. Rabbani, S. Wahab, R.A. Khan, Spontaneous self-ignition in a
10.1002/1526-4998(200009)56:9<779::AID-PS207>3.0.CO;2-U. case of acute aluminium phosphide poisoning, Am. J. Emergency Med. 27 (752)
(2009) e5–752.e6, https://doi.org/10.1016/j.ajem.2008.09.045.

40
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

[65] J. Yadav, B.K. Athawal, B.P. Dubey, V.K. Yadav, Spontaneous ignition in case of J. Legal Med. 131 (2017) 1001–1007, https://doi.org/10.1007/s00414-017-
celphos poisoning, Am. J. Forensic Med. Pathol. 28 (2007) 353–355, https://doi. 1562-1.
org/10.1097/PAF.0b013e31815b4bed. [89] S.N. Chugh, R. Pal, V. Singh, S. Seth, Serial blood phosphine levels in acute
[66] S. Hakimoglu, I. Dikey, A. Sari, L. Kekec, K. Tuzcu, M. Karcioglu, Successful aluminium phosphide poisoning, J. Assoc. Phys. India. 44 (1996) 184–185. PMID:
management of aluminium phosphide poisoning resulting in cardiac arrest, Turk. 9251315.
J. Anaesth. Reanim. 43 (2015) 288–290, https://doi.org/10.5152/ [90] F. Anger, F. Paysant, F. Brousse, I.L. Normand, P. Develay, Y. Gaillard, A. Baert,
TJAR.2015.75010. M.A.L. Gueut, G. Pepin, J.P. Anger, Fatal aluminum phosphide poisoning, J. Anal.
[67] N. Sinha, Aluminium phosphide poisoning, Indian J. Medical Specialities. 9 Toxicol. 24 (2000) 90–92, https://doi.org/10.1093/jat/24.2.90. PMID:
(2018) 167–170, https://doi.org/10.1016/j.injms.2018.06.006. 10732945.
[68] S.S. Bajwa, S. Bajwa Kaur, J. Kaur, K. Singh, A. Panda, Management of celphos [91] G.S. Bumbrah, K. Krishan, T. Kanchan, M. Sharma, G.S. Sodhi, Phosphide
poisoning with a novel intervention: a ray of hope in the darkest of clouds, poisoning: a review of literature, PMID: 21763089, Forensic Sci. Int. 214 (2012)
Anesth. Essays Res. 4 (2010) 20, https://doi.org/10.4103/0259-1162.69301. 1–6, https://doi.org/10.1016/j.forsciint.2011.06.018.
[69] S. Shadnia, M. Rahimi, A. Pajoumand, M.-H. Rasouli, M. Abdollahi, Successful [92] H. Kariman, K. Heydari, M. Fakhri, A. Shahrami, A.A. Dolatabadi, H.
treatment of acute aluminium phosphide poisoning: possible benefit of coconut A. Mohammadi, M. Gharibi, Aluminium phosphide poisoning and oxidative
oil, Hum. Exp. Toxicol. 24 (2005) 215–218, https://doi.org/10.1191/ stress: serum biomarker assessment, J. Med. Toxicol. 8 (2012) 281–284, https://
0960327105ht513oa. doi.org/10.1007/s13181-012-0219-1.
[70] D. Sharma, L.C. Mittal, S.R. Meena, C.P. Meena, G. Yadav, Aluminum Phosphide [93] S.N. Chugh, K.L. Jaggal, A. Sharma, B. Arora, K.C. Malhotra, Magnesium levels in
poisoning, Indian medical Gazette. (2014) 333-339. http://medind.nic.in/ice/ acute cardiotoxicity due to aluminium phosphide poisoning, Indian J. Med. Res.
t14/i9/icet14i9p333.pdf https://imsear.searo.who.int/handle/123456789/ 94 (1991) 437–439. PMID: 1774096.
157656 (accessed Oct. 2020). [94] A.M. Sciuto, B.J. Wong, M.E. Martens, H. Hoard-Fruchey, M.W. Perkins,
[71] M.H. Asghari, M. Abdollahi, M.R. de Oliveira, S.M. Nabavi, A review of the Phosphine toxicity: a story of disrupted mitochondrial metabolism: the toxicology
protective role of melatonin during phosphine-induced cardiotoxicity: focus on of phosphine poisoning, Ann. N.Y. Acad. Sci. 1374 (2016) 41–51, https://doi.org/
mitochondrial dysfunction, oxidative stress and apoptosis, J. Pharm. Pharmacol. 10.1111/nyas.13081.
69 (2017) 236–243, https://doi.org/10.1111/jphp.12682. [95] S. Mittra, S.S. Peshin, S.B. Lall, Cholinesterase inhibition by aluminium phosphide
[72] M. Goharbari, F. Taghaddosinejad, M. Arefi, M. Sharifzadeh, M. Mojtahedzadeh, poisoning in rats and effects of atropine and pralidoxime chloride, Acta
S. Nikfar, M. Baeeri, M. Rahimifard, M. Abdollahi, Therapeutic effects of oral Pharmacol Sin. 22 (2001) 37–39. PMID: 11730559.
liothyronine on aluminum phosphide poisoning as an adjuvant therapy: a clinical [96] W.T. Potter, V.F. Garry, J.T. Kelly, R. Tarone, J. Griffith, R.L. Nelson, Radiometric
trial, Hum. Exp. Toxicol. 37 (2018) 107–117, https://doi.org/10.1177/ assay of red cell and plasma cholinesterase in pesticide appliers from minnesota,
0960327117694074. Toxicol. Appl. Pharmacol. 119 (1993) 150–155, https://doi.org/10.1006/
[73] Z. Oghabian, O. Mehrpour, Treatment of aluminium phosphide poisoning with a taap.1993.1054.
combination of intravenous glucagon, digoxin and antioxidant agents, SQUMJ. [97] J.K. Marquis, A.J. Lerrick, Noncompetitive inhibition by aluminum, scandium
16 (2016) e352–e355. https://doi.org/10.18295/squmj.2016.16.03.015. and yttrium of acetylcholinesterase from Electrophorus electricus, Biochem.
[74] M. Soltani, S.F. Shetab-Boushehri, H. Mohammadi, S.V. Shetab-Boushehri, Pharmacol. 31 (1982) 1437–1440, https://doi.org/10.1016/0006-2952(82)
Proposing boric acid as an antidote for aluminium phosphide poisoning by 90040-5.
investigation of the chemical reaction between boric acid and phosphine, J. Med. [98] M.J. Al-Azzawi, Z.S. Al-Hakkak, B.W. Al-Adhami, In vitro inhibitory effects of
Hypotheses Ideas 7 (2013) 21–24, https://doi.org/10.1016/j.jmhi.2012.11.001. phosphine on human and mouse serum cholinesterase, Toxicol. Environ. Chem.
[75] N. Fakhraei, R. Hashemibakhsh, S.M. Rezayat, A.H. Abdolghafari, M.R. Jaafari, 29 (1990) 53–56, https://doi.org/10.1080/02772249009357617.
F. Mumtaz, S.E. Mousavi, On the benefit of nanocurcumin on aluminium [99] H. Saidi, S. Shojaie, Effect of sweet almond oil on survival rate and plasma
phosphide-induced cardiotoxicity in a rat model, Nanomed Res J. 4 (2019). htt cholinesterase activity of aluminum phosphide-intoxicated rats, Hum. Exp.
ps://doi.org/10.22034/nmrj.2019.02.008. Toxicol. 31 (2012) 518–522, https://doi.org/10.1177/0960327111407229.
[76] A. Ranjbar, L. Gholami, H. Ghasemi, N. Kheiripour, EEffects of nano-curcumin [100] S. Chowdhary, R. Bhattacharyya, D. Banerjee, A novel fluorescence based assay
and curcumin on the oxidant and antioxidant system of the liver mitochondria in for the detection of organophosphorus pesticide exposed cholinesterase activity
aluminum phosphide-induced experimental toxicity, Nanomedicine J. 7 (2020) using 1-naphthyl acetate, Biochimie 160 (2019) 100–112, https://doi.org/
58–64. https://doi.org/10.22038/nmj.2020.07.07. 10.1016/j.biochi.2019.02.014.
[77] S.M. Marashi, M. Majidi, M. Sadeghian, M. Jafarzadeh, S. Mohammadi, Z. Nasri- [101] J. Patočka, J. Bajgar, Aluminium activation and inhibition of human brain
Nasrabadi, Protective role of coenzyme Q10 as a means of alleviating the toxicity acetylcholinesterase in vitro, Inorg. Chim. Acta. 135 (1987) 161–163, https://doi.
of aluminum phosphide: an evidence-based review, Tzu Chi Medical J. 27 (2015) org/10.1016/S0020-1693(00)83282-X.
7–9, https://doi.org/10.1016/j.tcmj.2014.12.002. [102] P. Zatta, M. Ibn-Lkhayat-Idrissi, P. Zambenedetti, M. Kilyen, T. Kiss, In vivo and in
[78] M. Goswami, M. Bindal, P. Sen, S.K. Gupta, R. Avasthi, B.K. Ram, Fat and oil vitro effects of aluminum on the activity of mouse brain acetylcholinesterase,
inhibit phosphine release from aluminium phosphide-its clinical implication, Brain Res. Bull. 59 (2002) 41–45, https://doi.org/10.1016/S0361-9230(02)
Indian J. Exp. Biol. 32 (1994) 647–649. PMID: 7814045. 00836-5.
[79] A. Sharma, A. Sharma, A. Acharya, D. Aryal, B.G. Rajbanshi, P.R. Bhattarai, [103] R.R. Kaizer, M.C. Corrêa, R.M. Spanevello, V.M. Morsch, C.M. Mazzanti, J.
A. Regmi, A. Ghimire, Extracorporeal membrane oxygenation in aluminum F. Gonçalves, M.R.C. Schetinger, Acetylcholinesterase activation and enhanced
phosphide poisoning in Nepal: a case report, J. Med. Case Reports. 12 (2018) 311, lipid peroxidation after long-term exposure to low levels of aluminum on different
https://doi.org/10.1186/s13256-018-1864-z. mouse brain regions, J. Inorg. Biochem. 99 (2005) 1865–1870, https://doi.org/
[80] N. Rahimi, A.H. Abdolghaffari, A. Partoazar, N. Javadian, T. Dehpour, A.R. Mani, 10.1016/j.jinorgbio.2005.06.015.
A.R. Dehpour, Fresh red blood cells transfusion protects against aluminum [104] R.R. Kaizer, M.C. Corrêa, L.R.S. Gris, C.S. da Rosa, D. Bohrer, V.M. Morsch, M.R.
phosphide-induced metabolic acidosis and mortality in rats, PLoS ONE 13 (2018), C. Schetinger, Effect of long-term exposure to aluminum on the
e0193991, https://doi.org/10.1371/journal.pone.0193991. acetylcholinesterase activity in the central nervous system and erythrocytes,
[81] J. Ahmadi, S. Joukar, H. Anani, S. Karami-Mohajeri, Dihydroxyacetone as a Neurochem Res. 33 (2008) 2294–2301, https://doi.org/10.1007/s11064-008-
definitive treatment for aluminium phosphide poisoning in rats, Arch. Ind. 9725-6.
Hygiene Toxicol. 69 (2018) 169–177, https://doi.org/10.2478/aiht-2018-69- [105] S. Chowdhary, R. Bhattacharyya, D. Banerjee, Acute organophosphorus
3106. poisoning, Clin. Chim. Acta. 431 (2014) 66–76, https://doi.org/10.1016/j.
[82] A.H. Abdolghaffari, A. Baghaei, R. Solgi, M. Gooshe, M. Baeeri, M. Navaei-Nigjeh, cca.2014.01.024.
S. Hassani, A. Jafari, S.M. Rezayat, A.R. Dehpour, S.E. Mehr, M. Abdollahi, [106] A.T. Proudfoot, Aluminium and zinc phosphide poisoning, Clinical Toxicology. 47
Molecular and biochemical evidences on the protective effects of triiodothyronine (2009) 89–100, https://doi.org/10.1080/15563650802520675.
against phosphine-induced cardiac and mitochondrial toxicity, Life Sci. 139 [107] R. Anand, P. Kumari, A. Kaushal, A. Bal, W.Y. Wani, A. Sunkaria, R. Dua, S. Singh,
(2015) 30–39, https://doi.org/10.1016/j.lfs.2015.07.026. A. Bhalla, K.D. Gill, Effect of acute aluminum phosphide exposure on rats—a
[83] P. Parhizgar, R. Forouzanfar, S.K. Hadeiy, N. Zamani, H.H. Moghaddam, Sudden biochemical and histological correlation, Toxicol. Lett. 215 (2012) 62–69,
cardiac arrest in an asymptomatic zinc phosphide-poisoned patient: a case report, https://doi.org/10.1016/j.toxlet.2012.09.020.
Cardiovasc. Toxicol. 20 (2020) 525–530, https://doi.org/10.1007/s12012-020- [108] D. Yadav, R. Bhattacharyya, D.Banerjee, Lessons to be Learnt from Mitochondria
09578-2. PMID: 32451765. in Acute Aluminium Phosphate Poisoning. Acta Scientific Medical sciences
[84] R. Dechant, G. Sanders, R. Graul, Determination of phosphine in air, Am. Ind. (ASMS) 2019 ISSN:2582-0931. https://actascientific.com/ASMS/pdf/ASMS-SI-
Hyg. Assoc. J. (2007) 75–79, https://doi.org/10.1080/00028896609342796. 01-0001.pdf.
[85] M. Muthu, S.K. Parpia, H.A. Parpia, Detector for phosphine at permissible levels [109] C.G. Athanassiou, N.G. Kavallieratos, D.L. Brabec, P. Agrafioti, M. Sakka, J.
in air, J. Agric. Food Chem. 21 (1973) 184–186, https://doi.org/10.1021/ F. Campbell, Using immobilization as a quick diagnostic indicator for resistance
jf60186a010. PMID: 4688903. to phosphine, J. Stored Prod. Res. 82 (2019) 17–26, https://doi.org/10.1016/j.
[86] J.G. Leesch, Accuracy of different sampling pumps and detector tube jspr.2019.01.004.
combinations to determine phosphine concentrations, J. Econ. Entomol. 75 [110] S.K. Yadav, C. Srivastava, S. Sabtharishi, Phosphine resistance and antioxidant
(1982) 899–905, https://doi.org/10.1093/jee/75.5.899. enzyme activity in Trogoderma granarium Everts, J. Stored Prod. Res. 87 (2020),
[87] H. Yan, H. Chen, Z. Li, M. Shen, X. Zhuo, H. Wu, P. Xiang, Phosphine analysis in 101636, https://doi.org/10.1016/j.jspr.2020.101636.
postmortem specimens following inhalation of phosphine: fatal aluminum [111] P. Agrafioti, C.G. Athanassiou, Bh. Subramanyam, Efficacy of heat treatment on
phosphide poisoning in children, J. Anal. Toxicol. 42 (2018) 330–336, https:// phosphine resistant and susceptible populations of stored product insects,
doi.org/10.1093/jat/bky005. J. Stored Prod. Res. 81 (2019) 100–106, https://doi.org/10.1016/j.
[88] H. Yan, P. Xiang, S. Zhang, et al., Diagnosis of aluminum phosphide poisoning jspr.2018.11.007.
using a new analytical approach: forensic application to a lethal intoxication, Int.

41
D. Yadav et al. Clinica Chimica Acta 520 (2021) 34–42

[112] Lemic, Jembrek, Bažok, Pajač Živković, Ozone Effectiveness on Wheat Weevil and a flowchart to treat it, Arch. Industrial Hygiene Toxicol. 67 (2016) 183–193,
Suppression: Preliminary Research, Insects. 10 (2019) 357. https://doi.org/ https://doi.org/10.1515/aiht-2016-67-2784.
10.3390/insects10100357. [115] S. Alnasser, S.M. Hussain, T.S. Kirdi, A. Ahmed, Aluminum phosphide poisoning
[113] C.D. Singano, B.M. Mvumi, T.E. Stathers, H. Machekano, C. Nyamukondiwa, in Saudi Arabia over a nine-year period, Annals of Saudi Medicine. 38 (2018)
What does global warming mean for stored-grain protection? Options for 277–283, https://doi.org/10.5144/0256-4947.2018.277.
Prostephanus truncatus (Horn) control at increased temperatures, J. Stored Prod. [116] CHAPTER – 2 SUICIDES IN INDIA, (n.d.). https://ncrb.gov.in/sites/default/files/
Res. 85 (2020), 101532, https://doi.org/10.1016/j.jspr.2019.101532. chapter-2-suicides-2018.pdf (accessed October 13, 2020).
[114] B. Hashemi-Domeneh, N. Zamani, H. Hassanian-Moghaddam, M. Rahimi, [117] M.C. Meena, S. Mittal, Y. Rani, Fatal aluminium phosphide poisoning,
S. Shadnia, P. Erfantalab, A. Ostadi, A review of aluminium phosphide poisoning Interdisciplinary Toxicology. 8 (2015) 65–67, https://doi.org/10.1515/intox-
2015-0010.

42

You might also like