MedPath

Atropine Advanced Drug Monograph

Published:Jul 28, 2025

Generic Name

Atropine

Brand Names

Atnaa, Atropen, Busulfex, Donnatal, Duodote, Enlon-plus, Isopto Atropine, Lomotil, Minims Atropine Sulphate, Motofen, Phenohytro

Drug Type

Small Molecule

Chemical Formula

C17H23NO3

CAS Number

51-55-8

Associated Conditions

Amblyopia, Atrioventricular Heart-block, Bradycardia, Bronchospasm, Crying, Detrusor Hyperreflexia, Excessive bronchial secretion, Hypertonic uterine contraction, Hypertonicity of the small intestine, Ocular Inflammation, Parkinsonism, Peptic Ulcer, Poisoning by parasympathomimetics (cholinergics), Poisoning caused by mushrooms, Poisoning caused by organophosphate anticholinesterase nerve agents, Poisoning caused by organophosphorus pesticides, Pylorospasm, Rhinorrhoea, Sinus Bradycardia, Spasms, Toxic effect of organophosphate and carbamate, Hypermobility of the colon, Laughing, Life-threatening muscarinic side effects, Muscarinic side effects, Severe muscarinic side effects

Atropine (DB00572): A Comprehensive Pharmacological and Clinical Monograph

Section 1: Introduction and Chemical Profile

1.1 Overview and Historical Context

Atropine is a tropane alkaloid and a potent, non-selective anticholinergic agent that functions as a competitive antagonist at muscarinic acetylcholine receptors. It is classified as a small molecule drug with the DrugBank ID DB00572.[1] As a cornerstone of modern pharmacology, its applications span numerous medical specialties, including emergency medicine, cardiology, anesthesiology, and ophthalmology.[2] Its profound importance in global health is underscored by its inclusion in the World Health Organization (WHO) List of Essential Medicines, a compilation of the most efficacious, safe, and cost-effective medications required for a basic health system.[1]

The history of Atropine is a compelling narrative that traces the journey of a natural product from ancient folk remedy to a scientifically validated and indispensable therapeutic agent. The physiological effects of plants containing Atropine have been recognized for millennia. As far back as the last century B.C., Cleopatra reportedly used extracts from the Egyptian henbane plant, which contains tropane alkaloids, to dilate her pupils for cosmetic allure.[4] This practice continued into the Renaissance, when women used the juice from the berries of the

Atropa belladonna plant for the same purpose. This cosmetic application gave the plant its name, which translates to "beautiful lady" in Italian.[4]

The transition from herbal remedy to purified pharmaceutical began in 1831, when the German pharmacist Heinrich Mein successfully isolated pure Atropine from its plant source.[5] This milestone paved the way for its characterization and eventual synthesis. The complete chemical synthesis of Atropine was first achieved in 1901 by the German chemist Richard Willstätter, a landmark achievement in organic chemistry.[6] By the 1910s, Atropine had been integrated into clinical practice, notably as a preanesthetic medication to reduce the production of saliva and other respiratory secretions that could complicate airway management during surgery.[5] While the advent of newer anesthetics has somewhat reduced this specific application, Atropine's role in medicine has only expanded, solidifying its status as a critical tool for treating life-threatening bradycardia and as a primary antidote for chemical poisoning.[9] This historical progression, from the empirical observation of a plant's mydriatic effect to the isolation and synthesis of a molecule whose mechanism could be harnessed for diverse, life-saving applications, exemplifies a classic pathway in drug discovery.

1.2 Chemical Identity and Physicochemical Properties

Atropine is a naturally occurring tropane alkaloid extracted from plants of the Solanaceae family, including Atropa belladonna (deadly nightshade), Datura stramonium (Jimson weed), and Hyoscyamus niger (henbane).[8] Its formal chemical name is α-(hydroxymethyl)-benzeneacetic acid, (3-endo)-8-methyl-8-azabicyclo[3.2.1]oct-3-yl ester.[12]

A crucial aspect of its chemical identity is its stereochemistry. Atropine is a racemic mixture, meaning it consists of equal parts of two enantiomers: d-hyoscyamine and l-hyoscyamine.[1] The pharmacological activity of the drug, however, is attributed almost entirely to the levo-isomer, l-hyoscyamine.[1] This means that when a dose of racemic Atropine is administered, 50% of the drug mass is pharmacologically active, while the other 50% (the d-isomer) is largely inert. This stereochemical reality has direct consequences for the drug's metabolism and disposition in the body, as the two isomers are handled differently, a topic that will be explored in the pharmacokinetics section. Because it is a racemic mixture, Atropine itself is optically inactive.[15]

For clinical use, Atropine is most commonly formulated as Atropine Sulfate, a salt that exhibits significantly greater solubility in water compared to the atropine base, facilitating its preparation for injection and ophthalmic use.[1] The key chemical and physical properties of Atropine are summarized in Table 1.1.

Table 1.1: Chemical and Physical Properties of Atropine

PropertyValue / DescriptionSource(s)
Chemical Nameα-(hydroxymethyl)-benzeneacetic acid, (3-endo)-8-methyl-8-azabicyclo[3.2.1]oct-3-yl ester12
SynonymsDL-Hyoscyamine, Tropine tropate11
DrugBank IDDB005721
CAS Number51-55-812
Molecular FormulaC17​H23​NO3​1
Average Molecular Weight289.37 Da1
AppearanceWhite crystalline powder; long, orthorhombic prisms from acetone; rhombic needles from dilute ethyl alcohol3
Melting Point114–118.5 °C3
Solubility (Atropine Base)Soluble in ethanol, chloroform; slightly soluble in water (1.6 g/L at 18 °C) and ether8
Solubility (Atropine Sulfate)Very soluble in water (1 g in 0.4 mL); soluble in alcohol15
pKa9.7–9.88
StabilityLight sensitive; should be stored in airtight containers protected from light8

Section 2: Pharmacodynamics: Mechanism of Action

2.1 Core Mechanism: Muscarinic Acetylcholine Receptor Antagonism

The pharmacodynamic effects of Atropine are mediated through a single, well-defined mechanism: it is a competitive, reversible, and non-selective antagonist of muscarinic acetylcholine receptors (mAChRs).[1] Acetylcholine (ACh) is the primary neurotransmitter of the parasympathetic nervous system, which governs the body's "rest and digest" functions. Atropine functions by physically binding to mAChRs on the cell surface of various effector organs, thereby preventing ACh from binding and initiating its physiological response.[14]

This antagonism is described as "competitive" because Atropine and ACh compete for the same binding site on the receptor. It is "reversible," meaning Atropine binds non-covalently and can dissociate from the receptor, allowing normal function to resume once the drug is cleared. This reversibility also means the blockade is "surmountable"; if the concentration of ACh at the receptor site is increased sufficiently, ACh can outcompete Atropine for binding and overcome the blockade. This principle is the basis for the use of anticholinesterase agents (which prevent the breakdown of ACh) like physostigmine as an antidote for Atropine poisoning.[9] By inhibiting the parasympathetic nervous system, Atropine allows the effects of the opposing sympathetic nervous system ("fight or flight") to become dominant, leading to its characteristic physiological effects.[9]

2.2 Receptor Subtype Specificity and Downstream Effects

The breadth of Atropine's therapeutic applications and its extensive side-effect profile are both direct consequences of its non-selectivity. It exhibits high binding affinity for all five known subtypes of muscarinic receptors (M1, M2, M3, M4, and M5), with a half-maximal inhibitory concentration (IC50​) of approximately 2.5 nM.[1] This non-selective binding allows a single drug to influence a wide array of organ systems, which is highly advantageous in emergencies like systemic poisoning but also guarantees off-target effects during more localized therapy. This pharmacological "promiscuity" is a classic trade-off, contrasting with the modern goal of developing highly selective drugs.[23]

  • M1 Receptors (CNS, Salivary Glands): M1 receptors are prevalent in the central nervous system (CNS) and salivary glands. Atropine's blockade of these receptors contributes to the reduction of salivation (antisialagogue effect). At higher doses, this action in the CNS can lead to cognitive effects, including confusion, restlessness, and delirium.[24]
  • M2 Receptors (Heart, Presynaptic Autoreceptors): The M2 receptor is the primary target for Atropine's profound cardiac effects.
  • Postsynaptic Blockade in the Heart: The sinoatrial (SA) and atrioventricular (AV) nodes of the heart are richly innervated by the vagus nerve, which releases ACh. ACh binds to postsynaptic M2 receptors, which are coupled to inhibitory G-proteins (Gi​). This activation decreases intracellular cyclic AMP (cAMP) and opens potassium channels, which hyperpolarizes the cardiac cells. The collective effect is a slowing of the heart rate (negative chronotropy) and a reduction in AV conduction velocity (negative dromotropy).[20] Atropine competitively blocks these M2 receptors, effectively removing the vagal "brake" on the heart. This allows the underlying sympathetic tone to predominate, resulting in an increased SA node firing rate and enhanced AV conduction, which clinically manifests as tachycardia.[20]
  • Presynaptic Blockade and Paradoxical Bradycardia: A critical clinical nuance of Atropine's action is the phenomenon of paradoxical bradycardia, a transient slowing of the heart rate that can occur at low doses (less than 0.5 mg) or with slow intravenous administration.[9] The leading hypothesis for this effect involves the blockade of presynaptic M2 autoreceptors located on the vagal nerve terminals themselves. These autoreceptors normally function as a negative feedback mechanism, inhibiting further ACh release when activated. At low concentrations, Atropine may preferentially block these sensitive presynaptic receptors first. This blockade removes the inhibitory feedback ("disinhibition"), causing a temporary surge in ACh release into the synaptic cleft. This transiently higher concentration of ACh can then overcome the still-incomplete blockade at the postsynaptic receptors, resulting in a brief period of increased vagal effect and heart rate slowing, before the rising drug concentration achieves a dominant postsynaptic blockade and induces tachycardia.[9] This mechanism underscores the clinical importance of administering an adequate dose (e.g., 1 mg in adults) as a rapid IV push to quickly saturate all receptor sites and ensure the desired therapeutic effect.[26]
  • M3 Receptors (Smooth Muscle, Glands, Eye): The blockade of M3 receptors, which are coupled to Gq-proteins (Gq​), is responsible for many of Atropine's other well-known effects.[16]
  • Smooth Muscle: M3 receptor activation normally causes smooth muscle contraction. Atropine's blockade leads to relaxation of bronchial smooth muscle (bronchodilation), gastrointestinal smooth muscle (decreased motility, leading to constipation), and the detrusor muscle of the bladder (urinary retention).[3]
  • Exocrine Glands: M3 receptor activation stimulates gland secretion. Atropine's blockade inhibits salivary, lacrimal, bronchial, and sweat glands. This leads to the characteristic side effects of dry mouth (xerostomia), dry eyes, thickened bronchial secretions, and impaired sweating (anhidrosis).[9]
  • Eye: In the eye, M3 receptors mediate contraction of the pupillary sphincter muscle (causing pupil constriction, or miosis) and the ciliary muscle (allowing for accommodation, or focusing on near objects). Atropine blocks these receptors, preventing pupillary constriction and allowing the unopposed radial dilator muscle to cause mydriasis (pupil dilation). The paralysis of the ciliary muscle is known as cycloplegia.[9]
  • M4/M5 Receptors (CNS): These subtypes are also found primarily in the CNS. Their blockade by Atropine, which readily crosses the blood-brain barrier, contributes to the central effects of the drug, particularly at higher or toxic doses, which can manifest as excitation, disorientation, hallucinations, and delirium.[9]

2.3 Comparative Pharmacology

Understanding Atropine's properties is enhanced by comparing it to other anticholinergic agents.

  • Atropine vs. Scopolamine: Both are tertiary amine tropane alkaloids that cross the blood-brain barrier. However, their profiles differ. Atropine exerts a more potent and prolonged effect on the heart, intestines, and bronchial muscle. In contrast, scopolamine has a stronger effect on the iris, ciliary body, and certain secretory glands.[1] Clinically, scopolamine is known for producing more pronounced CNS effects, such as sedation and amnesia, due to its greater lipid solubility and easier passage into the CNS.[29]
  • Atropine vs. Glycopyrrolate: This comparison highlights a key structural difference with significant clinical implications. Glycopyrrolate is a quaternary ammonium compound, whereas Atropine is a tertiary amine. The charged quaternary structure of glycopyrrolate prevents it from readily crossing the blood-brain barrier.[29] This makes glycopyrrolate the preferred agent when purely peripheral anticholinergic effects are desired (e.g., reducing secretions during anesthesia) without the risk of central side effects like confusion or delirium, which is a particularly important consideration in elderly or susceptible patients.[29] Glycopyrrolate also has a longer intravenous duration of action (2–4 hours) compared to Atropine (approximately 30 minutes).[29]

Section 3: Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)

The clinical use of Atropine is guided by its pharmacokinetic profile, which describes its movement into, through, and out of the body. Its ADME properties determine the onset and duration of its effects and inform appropriate dosing strategies.

3.1 Absorption and Bioavailability

Atropine is absorbed rapidly and effectively following parenteral administration (intramuscular, intravenous, subcutaneous).[30] After an intramuscular (IM) injection, peak plasma concentrations are achieved in approximately 30 minutes, with levels comparable to those seen after intravenous (IV) administration.[18] When delivered via an IM autoinjector for emergency use, the time to maximum plasma concentration (

Tmax​) is even faster, occurring in as little as 3 minutes.[30]

Oral administration of Atropine as a single agent is not practiced; it is only available in oral combination products, such as with diphenoxylate for the treatment of diarrhea.[1]

A clinically significant route of absorption is ophthalmic. Despite being a topical application, Atropine eye drops lead to substantial systemic absorption through the conjunctiva and nasolacrimal drainage system.[18] The systemic bioavailability of the pharmacologically active l-hyoscyamine enantiomer from a 1% ophthalmic solution is surprisingly high, averaging about 64% with a wide range of 19% to 95%.[18] This significant systemic uptake from a supposedly "local" treatment explains why systemic anticholinergic side effects can occur, especially in vulnerable populations like children, and underscores the rationale for clinical practices aimed at minimizing absorption, such as using the lowest effective concentration and nasolacrimal occlusion.[23]

3.2 Distribution

Following absorption, Atropine is widely distributed throughout the body's tissues and fluids.[30] This extensive distribution is reflected in its large apparent volume of distribution (

Vd​), with reported values ranging from 1.0–1.7 L/kg to over 200 L, indicating that the drug does not remain confined to the bloodstream but partitions extensively into tissues.[18]

Plasma protein binding is relatively low and has been reported with some variability, with values cited between 14–22% and around 44%.[14] It binds primarily to alpha-1-acid glycoprotein (AAG).[31] As a tertiary amine, Atropine is lipid-soluble and readily crosses the blood-brain barrier, which accounts for its central nervous system effects.[9] It also efficiently crosses the placenta, with fetal blood concentrations observed to be equal to or greater than maternal concentrations within minutes of administration.[30] Traces of the drug are also secreted into breast milk.[30]

3.3 Metabolism

Atropine is extensively metabolized in the liver. The primary metabolic pathways are enzymatic hydrolysis, mediated by the enzyme atropine esterase, and oxidation via microsomal mono-oxygenase enzymes, part of the Cytochrome P450 system.[18] The major identified metabolites are noratropine (accounting for ~24% of a dose), atropine-N-oxide (~15%), tropine (~2%), and tropic acid.[18]

The metabolism is stereoselective. The pharmacologically active l-hyoscyamine enantiomer is preferentially metabolized, while the inactive d-hyoscyamine enantiomer is largely excreted from the body unchanged.[9]

A critical drug-disease interaction occurs in the context of organophosphate poisoning. Organophosphate compounds are known to inhibit the very microsomal enzymes that are responsible for Atropine's metabolism.[18] This inhibition leads to a decrease in Atropine clearance, resulting in higher and more sustained plasma concentrations of the antidote. In this unique clinical scenario, the poison paradoxically enhances the bioavailability and duration of action of its own antidote, a beneficial interaction that contributes to Atropine's efficacy in treating such poisonings.

3.4 Elimination

Atropine is eliminated from the body with a plasma concentration that follows a biexponential decay, indicating a rapid initial distribution phase followed by a slower elimination phase.[31] The elimination half-life (

t1/2​) is variable, with reported ranges of 2–4 hours for general parenteral use, a mean of 3.0 hours for IV administration, and a broader range of 3–10 hours in some studies.[9] The half-life following ocular administration is approximately 2.5 hours.[18]

Excretion occurs primarily through the kidneys. A substantial portion of the administered dose, reported as between 13% and 60%, is excreted unchanged in the urine, with the remainder appearing as metabolites.[9] The pharmacokinetics of Atropine have been noted to be nonlinear, especially with parenteral dosing.[18] Minor gender differences in pharmacokinetics have also been observed, with females exhibiting approximately 15% higher peak concentrations (

Cmax​) and area-under-the-curve (AUC) values and a slightly shorter half-life compared to males.[14]

Table 3.1: Key Pharmacokinetic Parameters of Atropine

ParameterValue / Range (Route, if specified)Source(s)
Bioavailability~64% (range 19–95%) (Ocular)18
Tmax​ (Time to Peak)~3 min (IM Autoinjector); ~28 min (Ocular); ~30 min (IM)18
Plasma Half-life (t1/2​)2–4 hours (General); 3.0 ± 0.9 hours (IV); ~2.5 hours (Ocular)9
Volume of Distribution (Vd​)1.0–1.7 L/kg18
Plasma Protein Binding14–44%14
MetabolismHepatic (hydrolysis via atropine esterase, oxidation via CYP enzymes); Major metabolites: noratropine, atropine-N-oxide, tropic acid18
ExcretionPrimarily renal; 13–60% excreted unchanged in urine9

Section 4: Clinical Applications and Therapeutic Use

Atropine's potent and broad anticholinergic effects make it an indispensable drug in a variety of clinical settings, from routine procedures to life-threatening emergencies. Its uses are well-established in cardiology, toxicology, anesthesiology, and ophthalmology.

4.1 Emergency Medicine and Cardiology

  • Symptomatic Bradycardia: Atropine is the first-line pharmacological agent for the management of hemodynamically unstable sinus bradycardia, where a slow heart rate leads to symptoms like hypotension, altered mental status, or signs of shock.[18] It is also effective in treating certain types of atrioventricular (AV) block, specifically Mobitz Type I (Wenckebach) second-degree AV block and third-degree (complete) heart block where the escape rhythm originates high in the conduction system (e.g., in the AV node).[9] Conversely, it is generally considered ineffective and not recommended for high-degree AV blocks located lower in the conduction system, such as Mobitz Type II second-degree block or third-degree block with a wide QRS complex (indicating a ventricular escape rhythm), as the vagus nerve has little influence on these lower pacemakers.[9]
  • Cardiac Arrest (Historical Use): For many years, Atropine was a standard component of Advanced Cardiac Life Support (ACLS) protocols for cardiac arrest rhythms like asystole and pulseless electrical activity (PEA). However, due to a lack of robust evidence demonstrating any survival benefit, it was removed from these international guidelines in 2010 and is no longer routinely recommended for this indication.[9]

4.2 Toxicology: The Primary Antidote

Atropine plays a critical, life-saving role as an antidote for poisoning by substances that cause excessive cholinergic stimulation through the inhibition of the enzyme acetylcholinesterase. This includes organophosphate and carbamate insecticides, as well as chemical warfare nerve agents such as sarin (GB), soman (GD), and VX.[1] It is also the treatment for poisoning by certain species of mushrooms that contain the toxin muscarine.[10]

In these poisonings, the accumulation of massive amounts of acetylcholine leads to a state of cholinergic crisis. Atropine works by competitively blocking the muscarinic receptors, shielding them from overstimulation. It effectively reverses the classic muscarinic "SLUDGE" symptoms: Salivation, Lacrimation, Urination, Defecation, Gastrointestinal distress, and Emesis, as well as bradycardia and life-threatening bronchoconstriction and hypersecretion of bronchial mucus.[19] It is crucial to recognize that Atropine only antagonizes the muscarinic effects. It has no effect on the nicotinic symptoms of poisoning, such as muscle fasciculations, weakness, and eventual respiratory muscle paralysis. For this reason, it is often administered concurrently with a cholinesterase reactivator, such as pralidoxime chloride (2-PAM), which works to restore the function of the acetylcholinesterase enzyme and reverse the nicotinic effects.[14] Dosing in poisoning is aggressive and must be titrated to clinical effect, with the primary goal being the drying of respiratory secretions to ensure a patent airway ("atropinization"). The required doses can be very large and may far exceed the standard maximum doses used for cardiac indications.[18]

4.3 Anesthesiology

In the perioperative setting, Atropine is used for several purposes:

  • Preanesthetic Medication: It can be administered preoperatively as an antisialagogue to reduce salivary and bronchial secretions, which can help maintain a clear airway during general anesthesia.[9]
  • Intraoperative Bradycardia: It is used to prevent or treat bradycardia that can be triggered by vagal reflexes during surgical manipulation (e.g., traction on visceral organs) or by the administration of certain anesthetic drugs like succinylcholine.[14]
  • Reversal of Neuromuscular Blockade: At the end of surgery, anticholinesterase agents like neostigmine are used to reverse the effects of non-depolarizing neuromuscular blocking agents. These reversal agents increase the amount of ACh at all cholinergic synapses, leading to undesirable muscarinic side effects like severe bradycardia and salivation. Atropine (or glycopyrrolate) is co-administered to competitively block these muscarinic effects, while allowing the desired nicotinic effect—the restoration of skeletal muscle function at the neuromuscular junction—to occur unimpeded.[34]

4.4 Ophthalmology

Topical ophthalmic Atropine is used for both diagnostic and therapeutic purposes:

  • Mydriasis and Cycloplegia: It is a potent agent for dilating the pupil (mydriasis) and paralyzing the ciliary muscle to inhibit accommodation (cycloplegia). This is necessary for conducting a thorough examination of the retina and for performing an accurate cycloplegic refraction to determine a child's full refractive error.[1] Due to its very long duration of action (effects can last 7–14 days), shorter-acting agents like tropicamide are more commonly preferred for routine diagnostic examinations.[9]
  • Uveitis and Iritis: In inflammatory conditions of the eye like uveitis, Atropine is used therapeutically to reduce pain caused by ciliary muscle spasm and to keep the pupil dilated, which helps prevent the formation of posterior synechiae (adhesions between the iris and the anterior surface of the lens).[5]
  • Amblyopia Treatment: Atropine is an established therapy for amblyopia ("lazy eye"). It is used as a "penalization" agent, where drops are instilled into the child's healthy eye. The resulting mydriasis and cycloplegia blur the vision in the good eye, forcing the child's brain to rely on and thereby strengthen the neural pathways of the amblyopic eye. Evidence suggests that atropine penalization is as effective as the traditional treatment of patching the good eye.[1]

4.5 Other Therapeutic Uses

  • Gastrointestinal Disorders: Atropine is a component of the combination drug Lomotil (atropine/diphenoxylate), which is used to treat diarrhea. Atropine contributes a minor antimotility effect, but its primary role is as an abuse deterrent. It is included in subtherapeutic amounts that are too low to cause significant effects at the recommended dose but produce unpleasant anticholinergic side effects if the product is taken in large quantities in an attempt to abuse the opioid diphenoxylate.[18]
  • Reduction of Secretions: Beyond anesthesia, Atropine can be used to treat conditions of excessive salivation (sialorrhea or ptyalism) and is frequently used in palliative and hospice care to reduce the accumulation of oral and pharyngeal secretions, managing the distressing symptom known as the "death rattle".[9]
  • Myopia Control: A significant and rapidly growing off-label use is the administration of low-concentration ophthalmic Atropine (0.01%–0.05%) to slow the rate of myopia progression in children. This emerging application will be discussed in detail in Section 6.[23]

Table 4.1: Dosing Regimens for Major Indications

The following table synthesizes dosing information from multiple clinical sources to provide a practical reference for the administration of Atropine across its primary indications. Dosages must always be tailored to the individual patient and clinical situation.

IndicationPatient PopulationRouteDose & FrequencyMaximum DoseKey Clinical NotesSource(s)
Symptomatic BradycardiaAdultIV / IO1 mg rapid push; repeat every 3–5 minutes as needed.Total 3 mgDoses <0.5 mg may cause paradoxical bradycardia. Ineffective for Mobitz II or 3rd-degree block with low escape rhythm.9
Pediatric (Child)IV / IO0.02 mg/kg; repeat every 5 minutes as needed. Minimum single dose: 0.1 mg.Single dose: 0.5 mg; Total cumulative: 1 mgRapid IV push required.18
Pediatric (Adolescent)IV / IO0.02 mg/kg; repeat every 5 minutes as needed. Minimum single dose: 0.1 mg.Single dose: 1 mg; Total cumulative: 2 mgRapid IV push required.18
Organophosphate / Nerve Agent PoisoningAdultIM / IVInitial: 2–3 mg (up to 6 mg in severe cases). Repeat every 5–60 minutes.No absolute max; titrate to effect. Doses up to 50 mg in 24h may be needed.Titrate to drying of bronchial and salivary secretions (atropinization). Often used with pralidoxime.26
PediatricIM / IV / IO0.02–0.05 mg/kg. Repeat every 5–20 minutes as needed.No absolute max; titrate to effect.Titrate to drying of secretions.26
Preanesthetic AntisialagogueAdultIV / IM / SC0.4–1 mg, given 30–60 minutes before anesthesia.1 mgTo reduce respiratory tract secretions.34
Pediatric (>5 kg)IV / IM / SC0.01–0.02 mg/kg, given 30–60 minutes before anesthesia.0.4 mgMinimum dose 0.1 mg.43
Ophthalmic - Uveitis / IritisAdult & PediatricOphthalmic (1% solution)1–2 drops applied to the conjunctival sac up to 3-4 times daily.-To relieve ciliary spasm and prevent synechiae.33
Ophthalmic - Cycloplegic RefractionAdult & Pediatric (>3 yrs)Ophthalmic (1% solution)1 drop applied 40 minutes prior to exam; may be repeated up to twice daily.2 drops/dayLong duration of action.32
Pediatric (3 mo–3 yrs)Ophthalmic (1% solution)1 drop applied 40 minutes prior to exam.1 drop/dayUse with caution due to risk of systemic effects.32

Section 5: Safety Profile, Adverse Effects, and Contraindications

The safety profile of Atropine is intrinsically linked to its non-selective antagonism of muscarinic receptors. Its adverse effects are, for the most part, predictable pharmacological extensions of its mechanism of action. A thorough understanding of this profile is essential for its safe clinical use, allowing clinicians to anticipate side effects, identify at-risk patients, and manage toxicity.

5.1 Adverse Drug Reactions

The adverse effects of Atropine are dose-dependent and can affect nearly every organ system with parasympathetic innervation.[8]

  • Common and Predictable Effects: The most frequently encountered side effects are direct results of M3 receptor blockade. These include dry mouth (xerostomia), blurred vision due to cycloplegia, sensitivity to light (photophobia) due to mydriasis, decreased sweating (anhidrosis), constipation from reduced gut motility, and difficulty with urination.[9]
  • Central Nervous System (CNS) Effects: Because Atropine crosses the blood-brain barrier, CNS effects are common, particularly with higher doses. These can range from mild effects like dizziness, restlessness, and confusion to severe toxicity manifesting as agitation, disorientation, delirium, and vivid hallucinations ("atropine psychosis"). In cases of severe overdose, this can progress to coma.[9] Elderly patients are known to be particularly susceptible to the CNS-altering effects of anticholinergic drugs.[9]
  • Cardiovascular Effects: The primary cardiovascular effect is sinus tachycardia. However, palpitations and arrhythmias, including premature ventricular contractions and, more rarely, atrial or ventricular fibrillation, can occur, especially in patients with pre-existing cardiac disease.[9] A significant clinical concern is that the increase in heart rate also increases myocardial oxygen demand. In a patient with active myocardial ischemia or a recent myocardial infarction, this atropine-induced tachycardia can worsen the ischemia, potentially extending the size of the infarct.[26]
  • Ocular Effects: The mydriasis and cycloplegia from ophthalmic use can be long-lasting, persisting for up to two weeks, causing prolonged photophobia and difficulty with near vision.[9] The most serious ocular risk is the precipitation of an acute angle-closure glaucoma attack. By causing the pupil to dilate, the iris tissue can bunch up at the periphery and physically block the trabecular meshwork, which is the drainage angle for aqueous humor. In an individual with anatomically narrow angles, this can cause a rapid and dangerous rise in intraocular pressure.[9]
  • Other Systemic Effects: Blockade of muscarinic receptors in the urinary tract can cause urinary retention, a major concern for patients with benign prostatic hyperplasia (BPH).[9] In the GI tract, it can convert a partial pyloric stenosis into a complete obstruction.[37] In the respiratory system, the inhibition of bronchial secretions can cause them to become thick and tenacious, leading to the formation of viscid mucous plugs that can obstruct airways, a particular risk for patients with chronic lung disease.[37] The inhibition of sweating (anhidrosis) impairs the body's ability to dissipate heat, which can lead to hyperthermia ("atropine fever"), especially in hot environments, with exercise, or in children, who have a less developed thermoregulatory system.[19]

Table 5.1: Adverse Reactions to Atropine by System Organ Class and Severity

The following table provides a structured overview of Atropine's adverse reactions, categorized by system, severity, and typical onset, based on extensive clinical data. The incidence for most effects is not known but is generally dose-dependent.

System Organ ClassSeverityAdverse ReactionOnsetSource(s)
CardiacSevereAsystole, Ventricular Fibrillation, Myocardial InfarctionRapid / Delayed37
ModerateSinus Tachycardia, Palpitations, Atrial Fibrillation, PVCsRapid / Early37
MildBradycardia (paradoxical, low-dose)Rapid9
Central Nervous SystemSevereComa, Seizures, Respiratory ArrestEarly / Delayed21
ModerateDelirium, Hallucinations, Confusion, Ataxia, AmnesiaEarly / Delayed9
MildDizziness, Restlessness, Headache, Drowsiness, TremorEarly9
OcularSevereAcute Angle-Closure Glaucoma, Visual ImpairmentEarly37
ModeratePhotophobia, Cycloplegia (loss of accommodation)Early9
MildMydriasis (dilated pupils), Blurred Vision, Dry EyesEarly44
GastrointestinalSevereIleus (paralytic)Delayed44
ModerateDysphagia (difficulty swallowing), ConstipationDelayed9
MildXerostomia (dry mouth), NauseaEarly9
GenitourinaryModerateUrinary RetentionEarly9
MildUrinary Urgency / IncontinenceEarly44
Dermatologic / SystemicSevereAnaphylactic Shock, Stevens-Johnson SyndromeRapid / Delayed44
ModerateAnhidrosis (impaired sweating), DehydrationDelayed19
MildFlushing, Dry Skin, RashRapid / Early26

5.2 Contraindications and Precautions

The contraindications for Atropine are logically derived from its physiological effects, targeting patient populations where these effects would exacerbate an underlying pathology.

  • Absolute Contraindication: A history of hypersensitivity or anaphylactic reaction to Atropine is an absolute contraindication.[39]
  • Relative Contraindications and Precautions: Atropine should be used with extreme caution or is relatively contraindicated in patients with:
  • Narrow-Angle Glaucoma: Due to the high risk of precipitating an acute attack.[9]
  • Cardiovascular Disease: Particularly in patients with acute myocardial ischemia or severe coronary artery disease, due to the risk of tachycardia worsening oxygen demand.[19] It is also contraindicated in specific high-degree AV blocks where it is known to be ineffective.[26] It is generally ineffective in heart transplant recipients due to cardiac denervation.[26]
  • Gastrointestinal and Genitourinary Obstruction: This includes patients with benign prostatic hyperplasia (BPH) due to the risk of inducing acute urinary retention, and patients with partial pyloric stenosis, where it may cause a complete obstruction.[19]
  • Other Conditions: Patients with chronic lung disease (risk of mucous plugging), individuals with a high fever, especially children (risk of inducing hyperthermia), and patients with Down syndrome, who exhibit increased sensitivity to the effects of Atropine.[26]

5.3 Significant Drug Interactions

  • Pharmacodynamic Synergism: The anticholinergic effects of Atropine are additive with other medications that possess anticholinergic properties. This includes tricyclic antidepressants, first-generation H1-antihistamines, phenothiazine antipsychotics, quinidine, and amantadine. Co-administration significantly increases the risk and severity of side effects like dry mouth, urinary retention, and confusion.[26]
  • Pharmaceutical Incompatibilities: Atropine sulfate for injection is chemically incompatible with alkaline solutions and should not be mixed in the same syringe with sodium bicarbonate. It has also been reported to form a precipitate when mixed with drugs such as noradrenaline bitartrate and pentobarbital sodium.[15]
  • Monoamine Oxidase Inhibitors (MAOIs): Co-administration with MAOIs is generally not recommended due to a potential risk of precipitating a hypertensive crisis.[46]

5.4 Overdose and Management

Atropine overdose results in an exaggerated manifestation of its anticholinergic effects, classically described by the toxidrome mnemonic: "Hot as a hare (hyperthermia), blind as a bat (mydriasis, cycloplegia), dry as a bone (anhidrosis, dry mucous membranes), red as a beet (flushing), and mad as a hatter (delirium, hallucinations)".[9] Severe intoxication can lead to circulatory collapse, respiratory depression, paralysis, and coma.[21] The fatal dose is not well defined but may be as low as 10 mg in children.[21]

Management involves supportive care, including cooling measures for hyperthermia and respiratory support. The specific antidote is physostigmine, a reversible acetylcholinesterase inhibitor. Unlike other anticholinesterases (e.g., neostigmine), physostigmine is a tertiary amine and can cross the blood-brain barrier, allowing it to reverse both the central and peripheral symptoms of atropine toxicity. It is administered via slow intravenous injection. Because physostigmine has a short half-life, repeated doses may be necessary.[9]

Section 6: Emerging Research and Future Directions

While Atropine is one of the oldest drugs in the modern pharmacopeia, it remains a subject of vibrant and impactful research. Two areas, in particular, highlight its evolving role: its repurposing for pediatric myopia control and the discovery of a novel, non-cholinergic mechanism of action.

6.1 Myopia Control in Children

Perhaps the most significant emerging application for Atropine is its off-label use in low concentrations to slow the progression of myopia (nearsightedness) in children. This represents a major therapeutic paradigm shift, repurposing a classic drug to address a chronic condition with a growing global prevalence.

  • Dose-Dependent Efficacy and Safety: Early studies showed that 1% Atropine was highly effective at halting myopia progression, but its use was limited by significant side effects, including profound photophobia and loss of near vision.[27] The breakthrough came from the hypothesis that lower doses might retain a useful degree of efficacy while minimizing these adverse effects. Subsequent large-scale clinical trials have confirmed this. Research has established a clear dose-dependent response: higher concentrations (e.g., 0.05%) demonstrate greater efficacy in slowing both the change in refractive error and the underlying axial elongation of the eye compared to lower concentrations (e.g., 0.01%).[23] However, these higher concentrations are also associated with a greater incidence of side effects. Based on current evidence, concentrations in the range of 0.025% to 0.05% appear to offer the most favorable balance between efficacy and tolerability.[23]
  • Mechanism of Action in Myopia: The precise mechanism by which Atropine slows eye growth is not fully understood and is an area of intense investigation. It is now widely believed that the effect is not primarily mediated by the paralysis of accommodation (cycloplegia). Instead, research points towards a more complex mechanism involving direct action on muscarinic receptors located in the retina and/or the sclera (the eye's outer wall). This interaction is thought to influence the biochemical signaling cascades that control scleral remodeling and growth. One leading theory suggests that Atropine may modulate choroidal thickness, which in turn influences the growth signals sent to the sclera.[27]
  • Pivotal Clinical Trials: The evidence base for this application has been built by several landmark randomized controlled trials. The LAMP (Low-Concentration Atropine for Myopia Progression) study was instrumental in establishing the dose-response relationship and demonstrating the superior efficacy of 0.05% Atropine over lower doses over several years.[50] The CHAMP (Childhood Atropine for Myopia Progression) trial investigated a novel, preservative-free, and stable formulation of low-dose Atropine (NVK002). This is a critical step towards obtaining regulatory approval and providing a standardized, commercially manufactured product, as current clinical use relies on variable preparations from compounding pharmacies.[52] Other trials, such as MTS1 and CHAMP-UK, continue to investigate the long-term efficacy and safety of low-dose atropine treatment.[53]

6.2 Novel Mechanisms: Atropine-Induced Autophagy

Recent and potentially transformative research has uncovered a completely new biological activity for Atropine: the induction of autophagy. Autophagy is a fundamental cellular process of "self-eating," where cells degrade and recycle damaged organelles and proteins to maintain homeostasis. Remarkably, this effect appears to be entirely independent of Atropine's well-known function as a muscarinic receptor antagonist.[55]

  • Evidence for a Non-Muscarinic Mechanism: The evidence for this novel mechanism is compelling. Studies have shown that Atropine can induce the formation of autophagosomes (the hallmark of autophagy) in cell lines that do not express muscarinic receptors, such as human kidney epithelial cells. Furthermore, in vivo experiments in mice demonstrated that Atropine induced autophagy in tissues that lack mAChRs (like the liver and heart) but did not induce it in tissues that are rich in them (like the hippocampus and smooth muscle). When the mAChR gene was experimentally silenced in cardiac muscle cells, Atropine then gained the ability to induce autophagy, confirming that the receptor's presence was inhibitory to this specific effect.[55]
  • Proposed Molecular Pathway: While the full pathway is still being elucidated, initial investigations point towards the involvement of the mTORC1 signaling pathway, a master regulator of cell growth and autophagy. RNA sequencing of cells treated with Atropine showed a significant increase in the expression of the Rag GTPase gene. Rag GTPases are key components in the signaling cascade that regulates mTORC1 activity, providing a potential molecular link between Atropine and the induction of autophagy.[55]
  • Potential Future Implications: This discovery fundamentally challenges the century-old understanding of Atropine as a purely anticholinergic drug. It suggests that Atropine may be a more complex, multi-target agent than previously appreciated. The ability to modulate autophagy has immense therapeutic potential across a vast spectrum of human diseases, including neurodegenerative disorders (e.g., Parkinson's, Alzheimer's), cancer, and metabolic diseases. While this research is still in its nascent stages, it opens up entirely new and unforeseen avenues for the therapeutic application of one of medicine's oldest and most familiar drugs, serving as a powerful reminder that even well-established compounds can hold undiscovered biological secrets.

Section 7: Conclusion and Clinical Synopsis

Atropine (DB00572) stands as a paradigm of a classic pharmacological agent, possessing a dual identity as both a time-tested, indispensable medication and a subject of dynamic, cutting-edge research. Its enduring legacy is built upon its potent, non-selective antagonism of muscarinic acetylcholine receptors, a mechanism that has been harnessed for over a century to manage a wide range of medical conditions.

In contemporary clinical practice, Atropine remains a critical tool. Its role in emergency medicine is paramount, serving as the first-line agent for treating hemodynamically unstable bradycardia and, most crucially, as the primary life-saving antidote for poisoning by organophosphate insecticides and chemical nerve agents. Its utility in anesthesiology for controlling secretions and managing vagal reflexes, and in ophthalmology for diagnostics and the treatment of uveitis and amblyopia, are fundamental applications that continue to save sight and improve surgical outcomes.

The very non-selectivity that grants Atropine its broad therapeutic power is also its principal liability. The drug's safety profile is a direct and predictable consequence of its mechanism, producing a wide array of anticholinergic side effects that demand careful patient selection, cautious dosing, and vigilant monitoring. The contraindications for its use are not arbitrary but are logically derived from a deep understanding of its physiological effects on at-risk organ systems, such as in patients with narrow-angle glaucoma or obstructive uropathy.

Simultaneously, Atropine is at the forefront of therapeutic innovation. Its repurposing as a low-dose topical agent for controlling the progression of myopia is revolutionizing pediatric eye care, offering the first effective pharmacological intervention for a condition of growing global concern. This application exemplifies how dose optimization can unlock entirely new therapeutic windows for established drugs. Furthermore, the recent and unexpected discovery of a novel, non-muscarinic mechanism—the induction of autophagy—has opened a new chapter in Atropine's long history. This finding suggests that the complete story of this ancient drug may still be unfolding, holding the potential for future applications in fields far beyond its traditional scope.

In conclusion, Atropine is a testament to the enduring value of foundational pharmacology. Its well-defined primary mechanism explains its established, critical roles in medicine, while ongoing scientific inquiry continues to reveal new layers of biological complexity and novel therapeutic horizons, ensuring its relevance for generations to come.

Works cited

  1. Atropine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 28, 2025, https://go.drugbank.com/drugs/DB00572
  2. The Role of Atropine in Modern Medicine: Indications, Administration, and Clinical Outcomes | Request PDF - ResearchGate, accessed July 28, 2025, https://www.researchgate.net/publication/380290679_The_Role_of_Atropine_in_Modern_Medicine_Indications_Administration_and_Clinical_Outcomes
  3. Atropine - PubChem, accessed July 28, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Atropine
  4. The Source-Synthesis- History and Use of Atropine, accessed July 28, 2025, https://eajem.com/pdf/bb2eeae3-0e60-42a4-acea-81e4a349912c/articles/jaem.2014.1120141/EAJEM-13-2-En.pdf
  5. Atropine | EBSCO Research Starters, accessed July 28, 2025, https://www.ebsco.com/research-starters/health-and-medicine/atropine
  6. www.woodlibrarymuseum.org, accessed July 28, 2025, https://www.woodlibrarymuseum.org/museum/atropine/#:~:text=Pure%20atropine%20was%20first%20produced,more%20easily%20than%20does%20atropine.
  7. Atropine - Wood Library-Museum of Anesthesiology, accessed July 28, 2025, https://www.woodlibrarymuseum.org/museum/atropine/
  8. Atropine | 51-55-8 - ChemicalBook, accessed July 28, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB2163178.htm
  9. Atropine - Wikipedia, accessed July 28, 2025, https://en.wikipedia.org/wiki/Atropine
  10. Atropine Injection: Uses & Side Effects - Cleveland Clinic, accessed July 28, 2025, https://my.clevelandclinic.org/health/drugs/19824-atropine-injection
  11. Atropine (DL-Hyoscyamine, Tropine tropate, CAS Number: 51-55-8) | Cayman Chemical, accessed July 28, 2025, https://www.caymanchem.com/product/12008/atropine
  12. Atropine (DL-Hyoscyamine, Tropine tropate, CAS Number: 51-55-8) | Cayman Chemical, accessed July 28, 2025, https://www.caymanchem.com/product/32997/atropine
  13. en.wikipedia.org, accessed July 28, 2025, https://en.wikipedia.org/wiki/Atropine#:~:text=Chemistry,-Learn%20more&text=Atropine%2C%20a%20tropane%20alkaloid%2C%20is,It%20is%20an%20antimuscarinic%20agent.
  14. Atropine Injection, 2 mg - accessdata.fda.gov, accessed July 28, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/212319s000lbl.pdf
  15. Atropine (International Programme on Chemical Safety Evaluation, 2002) - InChem.org, accessed July 28, 2025, https://www.inchem.org/documents/antidote/antidote/atropine.htm
  16. Atropine, mAChR antagonist (CAS 51-55-8) (ab145582) - Abcam, accessed July 28, 2025, https://www.abcam.com/en-us/products/biochemicals/atropine-machr-antagonist-ab145582
  17. CAS Number 51-55-8 | Atropine - Spectrum Chemical, accessed July 28, 2025, https://www.spectrumchemical.com/cas/51-55-8
  18. Atropine - StatPearls - NCBI Bookshelf, accessed July 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK470551/
  19. Atropine (intramuscular route) - Side effects & dosage - Mayo Clinic, accessed July 28, 2025, https://www.mayoclinic.org/drugs-supplements/atropine-intramuscular-route/description/drg-20506387
  20. Atropine (Muscarinic Receptor Antagonist) - CV Pharmacology, accessed July 28, 2025, https://cvpharmacology.com/antiarrhy/atropine
  21. Atropine: Package Insert / Prescribing Information - Drugs.com, accessed July 28, 2025, https://www.drugs.com/pro/atropine.html
  22. Muscarinic Receptor Agonists and Antagonists - MDPI, accessed July 28, 2025, https://www.mdpi.com/1420-3049/6/3/142
  23. What is atropine? Parents' guide - My Kids Vision, accessed July 28, 2025, https://www.mykidsvision.org/knowledge-centre/all-about-atropine
  24. Muscarinic Antagonists - StatPearls - NCBI Bookshelf, accessed July 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK557541/
  25. Physiology, Muscarinic Receptor - StatPearls - NCBI Bookshelf, accessed July 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK555909/
  26. www.summahealth.org, accessed July 28, 2025, https://www.summahealth.org/-/media/project/summahealth/website/page-content/ems/ems-protocols/drug-index/atropine.pdf
  27. Role of Atropine in the control of Myopia Progression- A Review - PMC, accessed July 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9522992/
  28. Drugs Interfering with Muscarinic Acetylcholine Receptors and Their Effects on Place Navigation - Frontiers, accessed July 28, 2025, https://www.frontiersin.org/journals/psychiatry/articles/10.3389/fpsyt.2017.00215/full
  29. Glycopyrrolate - StatPearls - NCBI Bookshelf, accessed July 28, 2025, https://www.ncbi.nlm.nih.gov/books/NBK526035/
  30. Atropine Sulfate - Medical Countermeasures Database - CHEMM, accessed July 28, 2025, https://chemm.hhs.gov/countermeasure_atropine-sulfate.htm
  31. 21-146 Atropine Sulfate Clinical Pharmacology Biopharmaceutics Review - accessdata.fda.gov, accessed July 28, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2001/21-146_Atropine%20Sulfate_biopharmr.pdf
  32. ISOPTO ATROPINE - accessdata.fda.gov, accessed July 28, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/208151lbl.pdf
  33. Atropine Ophthalmic Dosage Guide + Max Dose, Adjustments ..., accessed July 28, 2025, https://www.drugs.com/dosage/atropine-ophthalmic.html
  34. Atropine - WikiAnesthesia, accessed July 28, 2025, https://wikianesthesia.org/wiki/Atropine
  35. Atropine Monograph for Professionals - Drugs.com, accessed July 28, 2025, https://www.drugs.com/monograph/atropine.html
  36. Atropine injection Uses, Side Effects & Warnings - Drugs.com, accessed July 28, 2025, https://www.drugs.com/mtm/atropine-injection.html
  37. Atropine (Atropine): Side Effects, Uses, Dosage, Interactions, Warnings - Rx List, accessed July 28, 2025, https://www.rxlist.com/atropine-drug.htm
  38. AtroPen (atropine IV/IM) dosing, indications, interactions, adverse effects, and more, accessed July 28, 2025, https://reference.medscape.com/drug/atropen-atropine-iv-im-343093
  39. ATROPINE injectable - MSF Medical Guidelines, accessed July 28, 2025, https://medicalguidelines.msf.org/en/viewport/EssDr/english/atropine-injectable-16682477.html
  40. IsoptoAtropine (atropine ophthalmic) dosing, indications, interactions, adverse effects, and more - Medscape Reference, accessed July 28, 2025, https://reference.medscape.com/drug/atropine-ophthalmic-343612
  41. Atropine/diphenoxylate Reviews & Ratings - Drugs.com, accessed July 28, 2025, https://www.drugs.com/comments/atropine-diphenoxylate/
  42. www.researchgate.net, accessed July 28, 2025, https://www.researchgate.net/publication/380290679_The_Role_of_Atropine_in_Modern_Medicine_Indications_Administration_and_Clinical_Outcomes#:~:text=Atropine%20is%20currently%20the%20most,compared%20to%20higher%20concentration%20preparation.
  43. Atropine: Side Effects, Uses, Dosage, Interactions, Warnings - Rx List, accessed July 28, 2025, https://www.rxlist.com/atropine/generic-drug.htm
  44. atropine - Drug Summary - PDR.Net, accessed July 28, 2025, https://www.pdr.net/drug-summary/Atropine-Sulfate-Injection-atropine-sulfate-684
  45. isopto® atropine, accessed July 28, 2025, https://pdf.hres.ca/dpd_pm/00044074.PDF
  46. atropine sulfate monohydrate solution/ drops - DailyMed, accessed July 28, 2025, https://www.dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=bfdb85b2-86d0-4f32-a8fa-272af48a191c
  47. Atropine Ophthalmic: MedlinePlus Drug Information, accessed July 28, 2025, https://medlineplus.gov/druginfo/meds/a682487.html
  48. Atreza, Atropine PO (atropine) dosing, indications, interactions ..., accessed July 28, 2025, https://reference.medscape.com/drug/atreza-atropine-po-342061
  49. Atropine eye drops for myopia control in children - My Kids Vision, accessed July 28, 2025, https://www.mykidsvision.org/knowledge-centre/atropine-eye-drops-for-myopia-control-in-children
  50. LAMP Study: Atropine 0.05% Effective For Myopia Control at Five Years, accessed July 28, 2025, https://www.reviewofoptometry.com/news/article/lamp-study-atropine-005-effective-for-myopia-control-at-five-years
  51. Study Details | The Effects of Low Dose Atropine on Choroidal Thickness | ClinicalTrials.gov, accessed July 28, 2025, https://www.clinicaltrials.gov/study/NCT03544827
  52. The latest research on atropine: efficacy, ideal candidates, and safety | Myopia Profile, accessed July 28, 2025, https://www.myopiaprofile.com/articles/latest-research-atropine-efficacy-safety-NVK002
  53. Study Details | Low-Dose Atropine for Treatment of Myopia - ClinicalTrials.gov, accessed July 28, 2025, https://clinicaltrials.gov/study/NCT03334253
  54. Low-dose Atropine Eye Drops to Reduce Progression of Myopia in Children in the United Kingdom | ClinicalTrials.gov, accessed July 28, 2025, https://clinicaltrials.gov/study/NCT03690089
  55. Atropine can induce autophagy independent of the M3 ... - TMR, accessed July 28, 2025, https://www.tmrjournals.com/public/articlePDF/20240326/f22d11e48177a5a980712a48dc48e8d0.pdf

Published at: July 28, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.