Small Molecule
C18H26ClN3
54-05-7
Discoid Lupus Erythematosus (DLE), Extraintestinal Amebiasis, Malaria, Polymorphic Light Eruption (PLE), Porphyria Cutanea Tarda, Rheumatoid Arthritis, Sarcoidosis, Acute, uncomplicated Malaria
Chloroquine is a synthetic 4-aminoquinoline derivative that has occupied a multifaceted and often paradoxical role in medicine for over 70 years. Initially developed in the 1930s and widely adopted after World War II, it became a cornerstone of global malaria prophylaxis and treatment due to its high efficacy, low cost, and convenient dosing. However, its dominance was curtailed by the inexorable spread of parasite resistance, which has rendered it largely ineffective against the most lethal malaria parasite, Plasmodium falciparum, in most parts of the world. Serendipitous clinical observations during its widespread use led to its repurposing as a disease-modifying antirheumatic drug (DMARD). Today, its most significant clinical applications are in the management of autoimmune conditions such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), where it exerts immunomodulatory effects by interfering with antigen presentation and inflammatory signaling. This therapeutic utility is tempered by a significant safety profile, characterized by the risk of irreversible retinopathy and potentially fatal cardiotoxicity, particularly with the long-term, high-dose regimens required for autoimmune diseases. The recent, intense but ultimately unsuccessful investigation of Chloroquine for the treatment of COVID-19 served as a modern case study in the complexities of drug repurposing, underscoring the critical need for rigorous clinical evidence to validate preclinical hypotheses. This report provides a comprehensive monograph on Chloroquine, detailing its chemical properties, pharmacological mechanisms, clinical applications, and complex safety considerations.
A thorough understanding of Chloroquine's pharmacological behavior begins with its fundamental chemical and physical characteristics.
Chloroquine is a small molecule drug belonging to the aminoquinoline class.[1] It is identified across scientific and regulatory databases by its DrugBank ID, DB00608, and its Chemical Abstracts Service (CAS) Number, 54-05-7.[1] Its systematic International Union of Pure and Applied Chemistry (IUPAC) name is N4-(7-chloro-4-quinolinyl)-N1,N1-diethyl-1,4-pentanediamine.[1] The drug is known by numerous synonyms and international names, including Chloraquine, Chlorochin, Chloroquina, and the common brand name Aralen.[1]
The molecular formula of Chloroquine is C18H26ClN3.[1] Structurally, it is a 4-aminoquinoline, composed of a quinoline bicyclic aromatic core that is substituted at position 7 with a chlorine atom and at position 4 with a [5-(diethylamino)pentan-2-yl]amino side chain.[2] This specific arrangement of a lipophilic aromatic ring system and a basic side chain is fundamental to its biological activity. Its structure is precisely defined by identifiers such as the SMILES string CCN(CC)CCCC(C)NC1=C2C=CC(=CC2=NC=C1)Cl.[3]
Chloroquine is a weak base, a property that is central to its mechanism of action and pharmacokinetic profile.[5] It presents as a white to light yellow crystalline solid with a bitter taste.[3] Its physicochemical properties are summarized in Table 1.
Table 1: Chemical and Physical Properties of Chloroquine
Property | Value | Source(s) |
---|---|---|
Generic Name | Chloroquine | 1 |
DrugBank ID | DB00608 | 1 |
CAS Number | 54-05-7 | 3 |
Drug Type | Small Molecule | 1 |
Chemical Class | 4-aminoquinoline | 6 |
Molecular Formula | C18H26ClN3 | 1 |
Average Molecular Weight | 319.872 g/mol | 1 |
IUPAC Name | N4-(7-chloro-4-quinolinyl)-N1,N1-diethyl-1,4-pentanediamine | 1 |
Physical Appearance | White to light yellow solid | 3 |
Melting Point | 87°C | 3 |
XLogP | 4.27 | 4 |
pKa (Strongest Basic) | 10.32 | 9 |
The physicochemical nature of Chloroquine is the primary determinant of its biological fate and action. Its lipophilic quinoline ring structure allows it to readily diffuse across biological membranes, including the membranes of host red blood cells and the intraerythrocytic malaria parasite.[6] Once inside the cell, its character as a weak base becomes paramount. The drug has a pKa of 8.5 for its quinoline nitrogen, meaning it is largely protonated at physiological pH.[6] However, when it enters highly acidic intracellular compartments—such as the parasite's food vacuole (pH ≈ 4.7) or a host cell's lysosome (pH ≈ 4.6)—the equilibrium shifts dramatically, and the drug becomes almost entirely protonated.[6] This charged, protonated form is significantly less membrane-permeable, effectively trapping the molecule within the acidic organelle. This phenomenon, known as "ion trapping," leads to a massive accumulation of Chloroquine in these specific compartments.[6] This accumulation is not a side effect but the very basis of its therapeutic activity, concentrating the drug at its site of antimalarial action and simultaneously enabling its immunomodulatory effects by altering the internal environment of host immune cells.
For clinical use, Chloroquine is typically formulated as a salt, most commonly chloroquine phosphate or chloroquine sulfate, for oral administration in tablet form.[8] It is critical to distinguish between the weight of the salt and the weight of the active Chloroquine base for accurate dosing. For instance, a 250 mg tablet of chloroquine phosphate is equivalent to 150 mg of Chloroquine base, while a 500 mg tablet contains 300 mg of base.[14]
The history of Chloroquine is a compelling narrative of 20th-century pharmaceutical development, marked by wartime necessity, serendipitous clinical observation, and the persistent challenge of microbial evolution.
The story begins with the natural antimalarial quinine, isolated from the bark of the South American Cinchona tree in the 19th century.[5] The quest for more reliable and scalable synthetic alternatives drove research in the early 20th century. This effort culminated in 1934 when Hans Andersag, working at the Bayer laboratories in Germany, first synthesized the 4-aminoquinoline compound he named Resochin.[6] However, initial assessments deemed the compound too toxic for human use, and it was consequently shelved for nearly a decade.[19]
The trajectory of the drug was irrevocably altered by the events of World War II. The need for effective antimalarials to protect Allied troops was a strategic imperative. A related formulation, sontoquine, was captured from German forces in North Africa and subsequently analyzed by American researchers.[19] This prompted a re-evaluation of the original compound, Resochin, which was found to be both safe and highly effective. Renamed Chloroquine, it underwent extensive clinical trials and was granted FDA approval on October 31, 1949, becoming commercially available shortly thereafter.[1]
In the post-war era, Chloroquine became the global drug of choice for both the treatment and prophylaxis of malaria, playing a central role in the World Health Organization's ambitious malaria eradication campaign.[1] During this period of widespread use, a pivotal, serendipitous discovery was made. Soldiers taking the drug for malaria prophylaxis reported unexpected improvements in concurrent inflammatory conditions, such as skin rashes and arthritis.[11] These astute clinical observations led to formal trials in the 1950s, which confirmed its efficacy in autoimmune diseases like systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), establishing its second major therapeutic role.[1]
In response to concerns about Chloroquine's long-term toxicity, particularly to the retina, a hydroxylated analogue, hydroxychloroquine, was developed and introduced in 1955.[11] This derivative offered a similar therapeutic profile but with a significantly better safety record, and it has since largely superseded Chloroquine for the chronic treatment of autoimmune diseases.[14]
Chloroquine's "golden age" as the premier antimalarial was ultimately brought to an end by the predictable evolutionary response of its target. Widespread resistance in P. falciparum was first documented in the late 1950s and spread globally over the subsequent decades, severely limiting its utility.[1] This crisis spurred the development of newer antimalarials, including mefloquine and the now-standard artemisinin-based combination therapies (ACTs).[1] Despite the challenge of resistance, Chloroquine's importance is recognized by its continued inclusion on the World Health Organization's List of Essential Medicines for its remaining indications.[4]
Chloroquine exhibits distinct mechanisms of action in its different therapeutic contexts. However, these diverse effects are all downstream consequences of a single, unifying pharmacological principle: its accumulation within acidic intracellular organelles, a property known as lysosomotropism.
The antimalarial action of Chloroquine targets a critical metabolic process of the Plasmodium parasite during its asexual lifecycle stage within human red blood cells.[6]
In autoimmune diseases like SLE and RA, Chloroquine's therapeutic effect stems from its interference with the cellular processes that drive the aberrant immune response. This mechanism is also a direct result of its lysosomotropic properties, but in this case, the target is the host's own immune cells.[12]
The same fundamental properties of Chloroquine have led to investigations of its antiviral activity. The ability to raise the pH within endosomes can interfere with the entry of many viruses that rely on a low pH environment to trigger the fusion of their envelope with the endosomal membrane.[1] Specifically for SARS-CoV-2, it was also proposed that Chloroquine could inhibit the terminal glycosylation of the Angiotensin-Converting Enzyme 2 (ACE2) receptor, potentially reducing the binding efficiency of the viral spike protein.[1] While these mechanisms provided a plausible
in vitro rationale for its use in COVID-19, they failed to translate into clinical benefit. Large-scale clinical trials found no evidence of efficacy and suggested potential harm, leading the U.S. Food and Drug Administration (FDA) to revoke its Emergency Use Authorization (EUA) on June 15, 2020.[1]
The clinical use and toxicity profile of Chloroquine are governed by its unique and extreme pharmacokinetic properties, particularly its vast distribution and slow elimination. A summary of these parameters is provided in Table 2.
Following oral administration, Chloroquine is rapidly and almost completely absorbed from the gastrointestinal tract.[5] Its bioavailability is high, though it can be variable, with reported ranges of 52-102% for oral solutions and 67-114% for oral tablets.[1] Peak plasma concentrations are typically reached within 0.5 to 12 hours after a dose, reflecting some inter-individual variability in absorption rates.[1]
The distribution of Chloroquine is the most defining feature of its pharmacokinetics. It possesses an exceptionally large apparent volume of distribution (Vd), reported to be between 200 and 800 L/kg.[1] This value, which vastly exceeds total body water, indicates that the vast majority of the drug in the body is not in the plasma but is extensively sequestered in various tissues.[5] This tissue binding is substantial, with Chloroquine having a particular affinity for melanin-containing cells in the skin and, most critically, the retinal pigment epithelium of the eye.[5] This specific accumulation is directly responsible for the risk of irreversible retinopathy with chronic use. In the plasma, Chloroquine is 46-79% bound to proteins, including albumin and alpha-1-acid glycoprotein.[1]
Chloroquine undergoes partial metabolism in the liver, primarily through N-dealkylation reactions catalyzed by the cytochrome P450 (CYP) enzyme system.[6] The main enzymes responsible for this biotransformation are CYP2C8 and CYP3A4/5, with lesser contributions from CYP2D6 and CYP1A1.[1] The principal metabolite, N-desethylchloroquine, retains some antimalarial activity and can be further dealkylated to inactive metabolites.[1] Notably, Chloroquine itself is an inhibitor of CYP2D6, which creates a potential for drug-drug interactions with other medications metabolized by this pathway.[44]
The primary route of elimination for Chloroquine and its metabolites is via the kidneys.[1] A substantial portion of the drug, approximately 50%, is excreted in the urine as the unchanged parent compound, with about 10% excreted as desethylchloroquine.[1] The elimination process is extremely slow, a direct consequence of the extensive tissue sequestration and large volume of distribution. Because only the drug present in the plasma is available for renal clearance, the slow release from tissue reservoirs results in a remarkably long terminal elimination half-life, typically ranging from 20 to 60 days.[1] This protracted elimination means that the drug can be detected in the body for months after discontinuation of therapy.[44]
This extreme pharmacokinetic profile can be viewed as a double-edged sword. The long half-life is highly advantageous for malaria prophylaxis, as it allows for a convenient and effective once-weekly dosing regimen that promotes adherence.[15] However, this same property becomes a significant liability in the context of chronic, daily dosing for autoimmune diseases. The slow elimination and extensive tissue binding lead to progressive drug accumulation over months and years. This cumulative burden, especially in sensitive tissues like the retina and myocardium, is the direct cause of its most severe, irreversible toxicities. Thus, the very characteristic that makes it an excellent prophylactic agent is also what makes it a hazardous long-term therapy, necessitating careful risk management and diligent patient monitoring.
Table 2: Summary of Key Pharmacokinetic Parameters for Chloroquine
Parameter | Value | Source(s) |
---|---|---|
Bioavailability (Oral) | 67–114% (tablets) | 1 |
Tmax (Oral) | 0.5–12 hours | 1 |
Volume of Distribution (Vd) | 200–800 L/kg | 1 |
Plasma Protein Binding | 46–79% | 1 |
Primary Metabolic Pathways | Hepatic N-dealkylation (CYP2C8, CYP3A4/5) | 1 |
Primary Active Metabolite | N-desethylchloroquine | 1 |
Route of Elimination | Renal | 1 |
Percent Excreted Unchanged | ~50% in urine | 1 |
Terminal Half-life | 20–60 days | 1 |
Total Plasma Clearance | 0.35–1 L/h/kg | 1 |
Chloroquine's clinical utility has evolved significantly over time, reflecting both its broad biological activity and the changing landscape of infectious and autoimmune diseases.
The primary FDA-approved indications for Chloroquine are rooted in its anti-parasitic activity.[7]
The most significant modern use of Chloroquine and its analogue hydroxychloroquine is as a disease-modifying antirheumatic drug (DMARD) for a variety of autoimmune conditions.[1]
This evolution has led to a clinical paradox: Chloroquine's primary, original indication for malaria is now largely obsolete in practice due to resistance, while its most important and impactful contemporary uses in rheumatology are often technically off-label and are precisely the applications that carry the highest risk of long-term cumulative toxicity. This highlights a significant disconnect between the drug's regulatory history and its modern clinical reality.
Chloroquine's broad biological activities have led to its investigation for various other conditions, including viral infections like HIV and Zika virus.[1] Most prominently, the onset of the COVID-19 pandemic in early 2020 saw a surge of intense global interest in Chloroquine. Based on promising
in vitro data suggesting it could inhibit SARS-CoV-2 replication, the FDA issued an Emergency Use Authorization (EUA) on March 28, 2020, to allow its use in certain hospitalized patients.[38] However, subsequent large, randomized clinical trials failed to demonstrate any clinical benefit and, in some cases, suggested an increased risk of harm, particularly serious cardiac events.[39] Consequently, the FDA revoked the EUA on June 15, 2020, a decision echoed by warnings from the European Medicines Agency (EMA).[9] This episode serves as a powerful modern lesson on the importance of robust clinical evidence and the potential dangers of extrapolating preclinical findings directly to patient care.
The decline of Chloroquine as a first-line antimalarial is a classic and sobering example of the impact of antimicrobial resistance on global public health.
The first documented cases of Chloroquine-resistant P. falciparum emerged nearly simultaneously but independently in two distinct geographic foci in the late 1950s: on the Thai-Cambodia border in Southeast Asia and in Colombia, South America.[8] From these origins, resistance spread inexorably throughout the 1960s and 1970s across Asia and South America. Sub-Saharan Africa, the region with the highest malaria burden, was spared until the late 1970s, when resistance was first detected in Kenya and Tanzania. Within a decade, it had swept across the continent.[6] More recently, resistance has also been reported in
P. vivax.[21]
The primary molecular mechanism of Chloroquine resistance in P. falciparum has been definitively linked to mutations in a gene known as the P. falciparum chloroquine resistance transporter (pfcrt).[21] This gene encodes a protein located on the membrane of the parasite's digestive vacuole. The mutated PfCRT protein is thought to function as a transporter that actively pumps protonated Chloroquine out of the vacuole.[10] This efflux mechanism prevents the drug from accumulating to the high concentrations necessary to inhibit heme polymerization, allowing the parasite to survive and replicate despite the presence of the drug.
The global spread of Chloroquine resistance had a devastating impact, leading to a dramatic resurgence in malaria-related illness and death, particularly among children in Africa.[21] It was a major contributing factor to the failure of the mid-20th century global malaria eradication campaign. This public health crisis forced a fundamental shift in global malaria treatment policy, moving away from inexpensive Chloroquine monotherapy to significantly more expensive but highly effective artemisinin-based combination therapies (ACTs).[1] The cost difference is stark: a course of Chloroquine costs approximately $0.10, whereas a course of ACTs can cost $1.00 to $2.40 or more.[61] This price differential represents a major and ongoing economic challenge for malaria control programs in low-income countries.
While effective for its indications, Chloroquine has a significant and complex safety profile that requires careful patient selection, dosing, and monitoring to mitigate risks.
The most frequently reported adverse effects are generally mild and include gastrointestinal disturbances such as nausea, vomiting, diarrhea, and abdominal cramps.[49] Headaches are also common.[49] A particularly notable side effect is pruritus (itching), which occurs with very high frequency in individuals of African descent.[63] Other systemic effects can include skin rashes, pigmentary changes of the skin and hair, and hair loss.[63] Neuropsychiatric effects, while less common, can be severe and may include confusion, anxiety, paranoia, hallucinations, and suicidal ideation.[50]
The most feared long-term complication of Chloroquine therapy is irreversible retinopathy.
Cardiac toxicity is another serious, potentially fatal complication associated with both chronic use and acute overdose.
Due to its metabolism and effects on cardiac rhythm, Chloroquine is subject to numerous drug-drug interactions, as summarized in Table 3.
Table 3: Major Drug Interactions with Chloroquine
Interacting Drug/Class | Effect of Interaction | Clinical Management/Recommendation | Source(s) |
---|---|---|---|
Antacids, Kaolin | Decreased Chloroquine absorption | Separate administration by at least 4 hours | 6 |
Cimetidine | Increased Chloroquine plasma levels (inhibits metabolism) | Concomitant use should be avoided | 6 |
Ampicillin | Decreased ampicillin bioavailability | Separate administration by at least 2 hours | 6 |
Cyclosporine | Increased cyclosporine serum levels | Monitor cyclosporine levels closely; may require Chloroquine discontinuation | 6 |
Mefloquine | Increased risk of convulsions | Co-administration may increase seizure risk | 6 |
QT-prolonging agents (e.g., amiodarone, moxifloxacin, azithromycin) | Increased risk of life-threatening ventricular arrhythmias | Use with extreme caution or avoid; ECG monitoring is required | 51 |
CYP2D6 Substrates | Inhibition of CYP2D6 can increase levels of co-administered drugs | Use with caution; may require dose reduction of the CYP2D6 substrate | 82 |
Accurate dosing of Chloroquine is paramount to ensure efficacy while minimizing the risk of toxicity. Dosages are often expressed in terms of the salt form (chloroquine phosphate) but should be calculated based on the equivalent amount of Chloroquine base.
For conditions like RA and SLE, the goal is to use the lowest effective dose to control symptoms. A typical adult dose is 250 mg of chloroquine phosphate (150 mg base) daily.[85] However, to mitigate the risk of retinal toxicity, the daily dose should not exceed 2.3 mg/kg based on the patient's actual body weight. For many patients, this necessitates intermittent dosing (e.g., taking the medication only on certain days of the week) rather than daily administration.[14]
Table 4: Summary of Dosage and Administration by Indication
Indication | Population | Dosage (as Base) | Duration/Notes | Source(s) |
---|---|---|---|---|
Malaria Prophylaxis | Adult | 300 mg once weekly | Start 1-2 weeks pre-travel, continue 4 weeks post-travel | 15 |
Pediatric | 5 mg/kg once weekly (max 300 mg) | Same as adult schedule | 15 | |
Malaria Treatment (Uncomplicated) | Adult | 600 mg initial, then 300 mg at 6, 24, & 48 hrs | Total course over 3 days (1.5 g base total) | 15 |
Pediatric | 10 mg/kg initial, then 5 mg/kg at 6, 24, & 36 hrs | Total course over 3 days (25 mg/kg base total) | 15 | |
Extraintestinal Amebiasis | Adult | 600 mg daily for 2 days, then 300 mg daily | For at least 2-3 weeks, with an intestinal amebicide | 16 |
Rheumatoid Arthritis / SLE | Adult | 150 mg daily (or intermittent) | Long-term use. Do not exceed 2.3 mg/kg/day to minimize retinopathy risk. | 14 |
Chloroquine's place in modern medicine is best understood by comparing it to its principal alternatives in both infectious disease and rheumatology.
The decision to use Chloroquine for malaria prophylaxis is now highly dependent on geography due to resistance. Table 5 compares it with other common prophylactic agents. Chloroquine's main advantages are its extremely low cost and convenient weekly dosing schedule. However, its efficacy is limited to a few regions of the world. Atovaquone/proguanil and doxycycline are effective against resistant strains but are more expensive and require daily dosing. Mefloquine is also effective and dosed weekly but is associated with a significant risk of neuropsychiatric side effects.
Table 5: Comparative Analysis of Antimalarials for Prophylaxis
Agent | Efficacy vs. Resistant P. falciparum | Dosing Schedule | Cost (Relative) | Key Adverse Effects | Use in Pregnancy/Pediatrics |
---|---|---|---|---|---|
Chloroquine | None | Weekly | Very Low | Pruritus, GI upset, long-term retinopathy risk | Considered safe in pregnancy; weight-based dosing for children |
Atovaquone/Proguanil | High | Daily | High | GI upset (generally well-tolerated) | Not recommended in pregnancy; pediatric formulation available |
Doxycycline | High | Daily | Low | Photosensitivity, GI upset, vaginal candidiasis | Contraindicated in pregnancy and children <8 years |
Mefloquine | High | Weekly | Moderate | Neuropsychiatric effects (vivid dreams, anxiety, psychosis), dizziness | May be used in 2nd/3rd trimesters; use with caution in children |
In rheumatology, Chloroquine (and more commonly, hydroxychloroquine) is considered a "milder" conventional synthetic DMARD (csDMARD). Table 6 compares it to other common DMARDs for rheumatoid arthritis. Methotrexate is the undisputed first-line "anchor" drug for moderate-to-severe RA due to its superior efficacy in preventing joint damage. Chloroquine is often used for milder disease or in combination with other DMARDs. Its favorable safety profile regarding myelosuppression and hepatotoxicity (compared to methotrexate) and its relative safety in pregnancy are key advantages. However, the unique and serious risks of long-term retinopathy and cardiotoxicity require diligent monitoring, and its efficacy is less robust than that of methotrexate or biologic agents.
Table 6: Comparative Analysis of Chloroquine with Other DMARDs for RA
Agent | Relative Efficacy | Onset of Action | Primary Long-Term Toxicities | Monitoring Requirements | Cost (Relative) | Use in Pregnancy |
---|---|---|---|---|---|---|
Chloroquine/HCQ | Mild to Moderate | Slow (2-6 months) | Retinopathy, Cardiomyopathy | Annual ophthalmology exams | Low | Generally considered safe |
Methotrexate | High | Intermediate (1-2 months) | Hepatotoxicity, Myelosuppression, Pulmonary fibrosis | Frequent blood tests (liver, CBC) | Low | Contraindicated (teratogenic) |
Sulfasalazine | Moderate | Intermediate (1-3 months) | GI upset, Rash, Myelosuppression | Frequent blood tests (liver, CBC) | Low | Generally considered safe |
Biologics (TNF inhibitors) | Very High | Rapid (weeks) | Serious infections, Malignancy risk | Tuberculosis screening, infection monitoring | Very High | Varies by agent |
Chloroquine holds a unique and paradoxical position in the history of modern medicine. It is a drug that rose from a wartime imperative to become a pillar of global public health, fundamentally altering the fight against malaria. Its journey encapsulates the power of synthetic chemistry to improve upon nature, the critical role of serendipity in discovering new therapeutic applications, and the sobering reality of microbial evolution in the face of selective pressure.
The spread of resistance has largely relegated Chloroquine from its primary role as a first-line antimalarial to a niche agent for use in the few remaining areas of susceptibility. Simultaneously, its second life as a cornerstone therapy for systemic lupus erythematosus and a useful agent in rheumatoid arthritis has ensured its continued clinical relevance. This dual identity is governed by a single, elegant pharmacological principle—lysosomotropism—which is both the source of its efficacy and the root of its most dangerous toxicities. The very pharmacokinetic properties of extensive tissue accumulation and slow elimination that make it a convenient weekly prophylactic create the preconditions for irreversible retinal and cardiac damage with chronic use.
The recent intense but failed investigation into its use for COVID-19 provided a stark, public lesson on the gap between in vitro plausibility and in vivo reality, reinforcing the primacy of rigorous clinical evidence. Ultimately, Chloroquine's legacy is one of complexity. It is a powerful, inexpensive, and historically transformative medication whose study offers enduring lessons in pharmacology, infectious disease, and immunology. It remains a valuable tool in the therapeutic arsenal, but one that demands a profound respect for its intricate benefit-risk profile and a commitment to careful, evidence-based patient management.
Published at: August 2, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.