- 1Shanxi Key Laboratory of Aging Mechanism Research and Translational Applications, Changzhi Medical College, Changzhi, China
- 2School of Basic Medical Sciences, Changzhi Medical College, Changzhi, China
1 Introduction
Hydroxychloroquine (HCQ), an economically accessible pharmaceutical with multifaceted therapeutic effects, is utilized in the management of both autoimmune and infectious diseases. Within the realm of rheumatology, HCQ is a fundamental treatment for systemic lupus erythematosus (SLE) (1), where it functions by inhibiting Toll-like receptor (TLR7/9)-mediated nucleic acid recognition, elevation of lysosomal pH and the reduction of type I interferon secretion (2). In the context of rheumatoid arthritis (RA), HCQ plays a role in the suppression of B-cell activation and the production of proinflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), with its combination with methotrexate leading to significant improvements in disease activity (3). Furthermore, HCQ has been shown to mitigate the risk of thrombosis in antiphospholipid syndrome by inhibiting the formation of neutrophil extracellular traps (NETs) (4). Early studies on COVID-19 suggested that HCQ might interfere with viral envelope protein glycosylation and modulate cytokine storms, indicating potential therapeutic benefits (5).
The clinical application of HCQ is constrained by its complex dose-dependent toxicity profile. Prolonged use is associated with irreversible photoreceptor damage resulting from accumulation in retinal pigment epithelial cells, which carries an approximate 20% risk of cumulative blindness over 20 years. This condition initially manifests as “bull’s-eye” macular pigmentation (6). In the cardiovascular system, administration of high doses exceeding 5 mg/kg can lead to QT interval prolongation, myocardial fibrosis, and conduction blockades (7). A study involving 127 patients revealed that 39.4% of those undergoing HCQ treatment experienced cardiac conduction dysfunction (8). Cutaneous toxicity, with an incidence rate of 10% to 20%, encompasses mucosal and acral hyperpigmentation, drug eruptions, and pruritus (9). Muscular toxicity is characterized by proximal myasthenia and elevated creatine kinase levels, which may advance to respiratory muscle paralysis (10). Gastrointestinal reactions, such as nausea and diarrhea, are associated with dysbiosis, while patients with hepatic or renal insufficiency are at risk of drug accumulation (11). These adverse effects underscore the importance of continuous monitoring during treatment.
Understanding the mechanisms of HCQ is essential for addressing clinical challenges. HCQ has been shown to reduce cardiovascular events in patients with rheumatoid arthritis (RA) (12, 13) and to exhibit therapeutic effects in a murine model of myocarditis (14). However, it can also induce arrhythmias in some COVID-19 patients and cause cardiotoxicity with prolonged use (11, 15).
Despite extensive research, comprehensive reviews focusing on HCQ’s direct targets and mechanisms are limited. This review seeks to systematically elucidate HCQ’s interactions with nucleic acids, proteins, and lipids, thereby uncovering the fundamental mechanisms responsible for its therapeutic benefits and adverse effects. Such insights will aid in developing personalized treatment strategies based on biomarkers (e.g., circulating tumor DNA levels, lysosomal enzyme activity), thereby transforming HCQ’s “double-edged sword” effect into an advantage for precision medicine.
2 Targets and mechanisms of hydroxychloroquine
2.1 Nucleic acid targeting mechanisms
2.1.1 DNA binding properties
The binding of HCQ to DNA was initially identified using optical tweezers, which revealed the formation of a complex with DNA (16). HCQ interacts with double-stranded DNA (dsDNA) in a manner dependent on both concentration and sequence. At low concentrations, HCQ associates with the minor groove of DNA through electrostatic interactions. In contrast, at higher concentrations, intercalation occurs, as confirmed by single-molecule force spectroscopy and gel electrophoresis (17). Thermodynamic analyses indicate that this binding process is spontaneous and predominantly driven by electrostatic interactions, with binding constants decreasing as temperature increases (16, 18).
HCQ demonstrates sequence-specific selectivity in its interaction with DNA, exhibiting a notably higher binding affinity for guanine-cytosine (GC)-rich regions compared to adenine-thymine (AT)-rich regions (19). In GC-rich DNA, HCQ predominantly binds through the major groove with intercalation, whereas in AT-rich DNA, binding occurs primarily via the minor groove. The presence of salt ions, such as Mg²+ and Na+, influences this sequence-specific binding; Mg²+ ions enhance HCQ binding to AT-rich DNA but inhibit its binding to GC-rich DNA, a phenomenon attributed to variations in ion hydration and charge distribution within the DNA grooves (20). In patients with SLE, plasma concentrations of HCQ within the therapeutic range (0.5–1.0 μg/mL) show a positive correlation with the binding rates to cell-free DNA (cfDNA). The binding of HCQ to cfDNA significantly impedes the interaction of DNA with Toll-like receptor 9 (TLR9), resulting in a 40% reduction in the cfDNA-TLR9 complex (21). Furthermore, HCQ obscures CpG motifs in nucleosomal DNA, thereby preventing the activation of TLR9 in plasmacytoid dendritic cells (pDCs) and subsequently reducing the secretion of type I interferons (21, 22). In tumor-related research, hydroxychloroquine has also been reported to promote tumor metastasis through a dual mechanism of inhibiting the formation of NETs and blocking the TLR4/9-COX2 signaling pathway activated by extracellular trap DNA (23, 24). In diseases associated with altered immune microenvironment, hydroxychloroquine has shows promising therapeutic potentiall.
2.1.2 Regulation of G4 DNA
Recent research indicates that HCQ influences the stability of intracellular G-quadruplexes (G4) (25). HCQ promotes the formation of G4 structures within telomeric G-rich sequences, where the number of flanking nucleotides plays a crucial role in determining G4 folding and stability. Specifically, single-side flanking nucleotides exert minimal impact, whereas the presence of multiple flanking nucleotides on both sides markedly reduces the propensity for G4 folding and stability (25). Regarding topological selectivity, HCQ exhibits a preference for binding to human telomere hybrid-type G4 structures (26), effectively stabilizing telomeric G4 and preventing its transition to dsDNA. In typical cellular environments, which are predominantly characterized by the presence of potassium ions, HCQ demonstrates a negligible effect (27). The mechanism of HCQ binding to G4 involves π-π stacking, C-H˙˙˙π interactions, and hydrogen bonding between positively charged side chains and guanine quartets (25).
The DNA of malaria parasites is rich in G-quadruplex sequences, and therapies targeting these G4 structures have demonstrated promising antimalarial effects (28, 29). Considering HCQ’s ability to bind to G4 structures, it may offer a low-toxicity therapeutic option for malaria patients who also suffer from autoimmune diseases. Furthermore, this property of HCQ could inspire the development of next-generation antimalarial ligands aimed at overcoming drug resistance (30).
2.1.3 RNA interaction network
Research on the binding interactions between HCQ and RNA emerged in response to the COVID-19 pandemic (31), while Early hypotheses and preliminary studies initially suggested a potential therapeutic benefit of HCQ for COVID-19, subsequent, more robust clinical trials and analyses have not supported its efficacy in treating COVID-19. Thermodynamic analyses indicate that HCQ associates with RNA grooves through an entropy-driven mechanism, with a preference for uridine/cytidine-rich regions (31). These results imply that HCQ could potentially inhibit Toll-like receptor 8 (TLR8) recognition of RNA from Plasmodium falciparum-infected erythrocytes or NETs in SLE patients, thereby diminishing interferon-gamma (IFN-γ) secretion and mitigating inflammatory responses (32, 33).
HCQ specifically interacts with the stem-loop II motif (s2m) of SARS-CoV-2 RNA, promoting the dimerization of this element, which subsequently disrupts viral envelope protein maturation and RNA proofreading (34, 35). Additionally, HCQ binds to the antiterminator RNA of T-box riboswitches, thereby inhibiting transcriptional antitermination (36, 37). The RNA of the malaria parasite is characterized by a pronounced A/T bias and an abundance of hairpins and multi-stem loops, which are essential for parasite infection (38–40). A comparative analysis of HCQ-bound T-box RNA and the structural attributes of parasite RNA reveals notable similarities, indicating that HCQ may directly associate with parasite RNA. This interaction could disrupt RNA metabolism, interfere with the temperature-responsive conformational changes of 18S rRNA, and ultimately inhibit parasite development within red blood cells (38).
RNA G-quadruplex structures are also present in A/U-rich sequences of Plasmodium falciparum, particularly within the RNA of virulence-associated genes, such as the var gene family, and in telomeric regions (30, 39, 41). These G4 structures frequently exhibit non-canonical forms, such as bulged or double quadruplexes, and overlap with regions coding for low-complexity peptides, playing a role in the regulation of antigenic variation. Considering the established ability of HCQ to bind DNA G4 structures, it is plausible that HCQ may also inhibit RNA G4s, potentially contributing to its pharmacological effects.
2.2 Protein interaction landscape
Historically, the disruption of autophagic flux was regarded as the primary mechanism by which chloroquine influences cellular function (42). However, experimental studies utilizing zebrafish and tumor models have demonstrated that HCQ exhibits significant antitumor activity that is independent of autophagy (43). Network pharmacology analyses have identified potential targets of HCQ across various diseases, including cyclin-dependent kinase 2 (CDK2), matrix metalloproteinase 2 (MMP2), and smoothened (SMO), among others (44–46). Furthermore, molecular docking studies and experimental data indicate that HCQ directly interacts with proteins such as palmitoyl-protein thioesterase 1 (PPT1) (47), nucleocapsid phosphoprotein (48), angiotensin-converting enzyme 2 (ACE2) (49), and the α7 nicotinic acetylcholine receptor (α7 nAChR) (50).
A recent investigation employing Thermal Proteome Profiling technology has identified that HCQ may directly interact with several proteins, including NQO2, GSR, NAMPT, KIF11, SEC23A, PUF60, PCM1, and NONO, among others (51). Notably, proteins such as SEC23A, PCM1, and ARFGAP1 have been implicated in the autophagic process (52–54), thereby enhancing our comprehension of HCQ’s role. However, the precise molecular mechanism by which HCQ impedes autophagy, specifically through the reduction of autophagosome-lysosome fusion, remains elusive (42). This ambiguity may be attributed to brief cell treatment durations or inherent technical limitations (51).
HCQ has been identified as an antithrombotic agent in the treatment of antiphospholipid syndrome and SLE (55–57), with a favorable safety profile (58). Mechanistically, HCQ inhibits the formation of antiphospholipid (aPL) IgG-beta2-glycoprotein I (beta2GPI) complexes and reduces protein binding to phospholipid bilayers (59, 60). Additionally, HCQ counteracts the effects of antiphospholipid antibodies on annexin A5 (AnxA5), facilitates the restoration of AnxA5 binding to phospholipid bilayers, and reinstates AnxA5’s anticoagulant properties (61). These actions elucidate HCQ’s mechanism in the management of childhood chronic immune thrombocytopenia (62).
2.3 Disruption of pH homeostasis
Chloroquine (CQ) and HCQ, as dibasic weak alkaline drugs, exist in both protonated and unprotonated forms. The unprotonated forms of CQ and HCQ are capable of freely traversing cell membranes and diffusing into acidic organelles, such as lysosomes and Golgi lumens, where they neutralize the acidic pH (63). Numerous studies utilizing probes have documented the impact of hydroxychloroquine and chloroquine on lysosomal pH (64, 65). Notably, treatment with 200 μM chloroquine for one hour elevates the intracellular lysosomal pH from 5.3 ± 0.1 to 6.6 ± 0.1, without altering the overall cytoplasmic pH (66). A reduction in lysosomal acidity can trigger a cascade of effects, including the inactivation of acidic hydrolases within lysosomes, accumulation of metabolic waste products, decreased autophagic efficiency, and other related complications (67). Nonetheless, it remains uncertain whether the elevation in lysosomal pH directly influences autophagosome-lysosome fusion. The identification of direct binding targets of HCQ, such as SEC23A, PCM1, and ARFGAP1, implies that HCQ may inhibit autophagosome-lysosome fusion through more direct mechanisms (51).
2.4 Membrane structure remodeling
2.4.1 Lysosomes
Lysosomal membranes possess a distinct composition characterized by the presence of a unique anionic lipid, bis(monoacylglycero)phosphate (BMP), which serves as a lysosomal marker not found in other intracellular membranes (68). Research indicates that HCQ interferes with the hydrogen bond network of water molecules by directly integrating into negatively charged phospholipid membranes, akin to those of lysosomal membranes, rather than amphoteric phospholipids. This integration results in water structure disorder or induces “gauche defects” in the hydrocarbon chains, leading to a disordered arrangement of membrane lipids and alterations in the membrane’s physical properties and composition (69). Such disruptions may influence the localization and activity of critical fusion proteins, including SNARE protein complexes and Rab GTPases, on the membrane. Consequently, this interference can impede the docking and fusion of autophagosome-lysosome membranes, thereby obstructing autophagosome-lysosome fusion and ultimately diminishing the efficiency of autophagic degradation (70).
2.4.2 Mitochondria
Numerous studies have demonstrated that the use of HCQ significantly elevates intracellular reactive oxygen species (ROS) levels (71). As the primary organelle responsible for ROS production, mitochondria in myocardial cells undergo rapid alterations within 60 minutes of HCQ treatment at a concentration of 100 μM. These alterations include the collapse of mitochondrial membrane potential, mitochondrial swelling, and the release of cytochrome C (72). In contrast, the same concentration of HCQ takes a longer duration to substantially inhibit autophagy (42). Cardiac cell mitochondrial membranes contain approximately 15–30% anionic lipids, representing the highest content among all membrane-bound organelles (73–75). Consequently, upon cellular entry, HCQ preferentially targets mitochondrial membranes due to their abundant anionic lipid content, resulting in rapid mitochondrial membrane damage, disruption of the respiratory chain, and increased ROS production (76).
Furthermore, related research indicates that HCQ can directly interact with proteins such as glutathione reductase (GSR) (51), suggesting its potential to interfere with glutathione (GSH) production (77), and consequently promote an increase in ROS. Additional studies reveal that HCQ binds to the hydrophobic regions of membrane phospholipids, resulting in the neutralization of phosphate groups and displacement of calcium ions, thereby causing mitochondrial damage (78). Animal studies have demonstrated that HCQ treatment leads to an increase in phospholipids and a reduction in cholesterol within mitochondrial membranes (79), which implies decreased membrane fluidity and functional impairment. Ultrastructural analysis of cardiac tissue from a patient with HCQ-induced acute left ventricular failure corroborated mitochondrial damage, exhibiting membrane-bound concentric lamellar structures, aggregates of curvilinear bodies (In fact, this represents a manifestation of structural abnormalities in intracellular membrane structures), and variations in morphology and size, accompanied by damage to the mitochondrial cristae (80). These findings elucidate the pharmacological mechanisms underlying hydroxychloroquine-induced cardiotoxicity. In future clinical applications, the co-administration of mitochondrial-protective agents, such as melatonin, edaravone, tadalafil (81), and coenzyme Q10, may be considered to mitigate.
3 Strategies to reduce HCQ side effects and enhance efficacy
3.1 Liposomal carriers
Liposomes have the capability to target glioma, melanoma cells, or activated hepatic stellate cells through peptide modifications, such as the Angiopep-2 peptide (82) and the R8-dGR peptide (83), or through the application of double-membrane hybrid nano-bionic technology (84). The use of pH-sensitive release mechanisms, exemplified by Chol-HCQ liposomes (85), or copper complexation technology (86), facilitates targeted drug release within tumor microenvironments or lysosomes, achieving approximately an 850-fold increase in efficacy compared to free HCQ. The co-loading of liposomes with chemotherapeutic agents like paclitaxel (87) or photosensitizers (82) results in a synergistic inhibition of autophagy or an enhancement of phototherapy effects. Additionally, a novel lipid nanoparticle formulation that co-delivers HCQ and TNF-α-targeting siRNA (siTNF-α) has been developed for the treatment of rheumatoid arthritis (88). Empirical studies have demonstrated that Chol-HCQ liposomes effectively inhibit pulmonary inflammation and mitigate bleomycin-induced pulmonary fibrosis, with the safety of systemic administration confirmed 28 days following intravenous injection (85). While this study did not include data specifically addressing retinal or cardiac toxicity, it is hypothesized that liposomal encapsulation could mitigate the potential toxicity of HCQ to organs such as the retina and heart. This hypothesis is based on the safety verification and the principle that targeted drug delivery minimizes systemic exposure. Furthermore, liposomal delivery of HCQ has the potential to accumulate in lysosomes, thereby enhancing the efficiency of autophagy inhibition (86), and to achieve precise delivery and efficacy enhancement by modulating the activation of immune cells within the tumor microenvironment (87).
3.2 Chiral hydroxychloroquine
HCQ possesses a chiral center, resulting in two optical isomers: (-)-(R)-hydroxychloroquine and (+)-(S)-hydroxychloroquine. These chiral isomers exhibit distinct pharmacokinetic profiles, target binding affinities, and pharmacological effects. Notably, the R (-) and S (+) enantiomers of HCQ demonstrate significant differences in cardiac electrophysiology and calcium ion regulation. Specifically, R (-) HCQ markedly depolarizes the resting membrane potential of rabbit Purkinje fibers and induces spontaneous electrical activity, whereas S (+) HCQ predominantly prolongs the action potential duration, exhibiting 2–4-fold enantioselective inhibition of hERG and potassium channels (89). In human induced pluripotent stem cell-derived cardiomyocytes, R (-) HCQ inhibits Ca²+ oscillations with significantly greater potency than both S (+) HCQ and the racemic mixture (90). Furthermore, in vivo studies in rabbits indicate that R (-) HCQ achieves significantly higher concentrations in whole blood compared to S (+) HCQ, potentially due to its enhanced binding affinity for blood cell components (91).
Studies on anti-SARS-CoV-2 agents indicate that the R (-) enantiomer of HCQ exhibits superior in vitro antiviral activity and reduced in vivo toxicity compared to the S (+) enantiomer (92). Conversely, the S (+) enantiomer demonstrates a significantly enhanced binding affinity to the ACE2 receptor relative to both the R (-) enantiomer and the racemic mixture (93). Regarding ocular distribution, HCQ enantiomers display reversible enantioselective binding in the non-pigmented ocular tissues of rabbits (94). In patients with rheumatoid arthritis, plasma concentrations of R (-) HCQ are generally higher than those of S (+) HCQ, with markedly different clearance rates observed between the two enantiomers (95).
4 Clinical hydroxychloroquine usage strategy, based on the multi-targets
4.1 Treatment of autoimmune diseases
Beyond its established applications in rheumatoid arthritis and systemic lupus erythematosus, HCQ holds particular value as an adjunctive treatment in obstetrics, primarily due to its capacity to inhibit phospholipid antibody complex formation. This capability enables it to effectively prevent or manage conditions such as placental insufficiency, recurrent miscarriage, preterm birth, fetal growth restriction, and preeclampsia associated with antiphospholipid syndrome (57, 60, 96–99). In the cardiovascular context, HCQ is associated with a reduced risk of cardiovascular events in patients with hypertension, diabetes, or rheumatoid arthritis, mediated through anti-phospholipid antibody mechanisms (100–102), and it also confers neuroprotective benefits post-stroke (103). Additionally, HCQ is employed in the management of various other autoimmune-related conditions, including myositis, systemic sarcoidosis, cutaneous sarcoidosis, and granulomatous skin eruptions (104–108).
Cardiac and retinal toxicities from HCQ are linked to mitochondrial damage and elevated ROS (109). Importantly, HCQ significantly inhibits the activity of zinc-dependent antioxidant enzymes (110), and the maintenance of zinc ion homeostasis is essential for preserving cardiac function (111). This implies that dysregulated zinc metabolism may represent a fundamental mechanism underlying HCQ toxicity. Recent mechanistic studies further substantiate this hypothesis: Zhang et al. reported that HCQ directly interacts with the zinc transporter SLC30A7 and glutathione disulfide reductase (GSR), thereby disrupting intracellular zinc metabolism and antioxidant equilibrium (51). Studies show zinc supplementation counteracts HCQ-induced zinc reduction in Drosophila larvae (112) and prevents cardiac damage in rats (113). These findings imply that aberrant zinc transport caused by HCQ is a significant contributor to its side effects. Additionally, HCQ-induced neurological toxicity exhibits ultrastructural similarities to its cardiotoxicity (114). Consequently, we advocate for appropriate zinc supplementation in patients undergoing long-term conventional or short-term high-dose HCQ treatments to reduce the risk of cardiac, retinal, and neurological toxicities.
4.2 Treatment of infectious diseases
In the realm of antimalarial therapy, the structural modification of HCQ may represent a promising avenue for future research. Notably, the synthesis of zinc-hydroxychloroquine complexes (115, 116) by altering the conformation of HCQ and its distribution within plasmodia, shows potential in combating drug resistance arising from mutations in the Pfcrt and Pfmdr1 genes, which interfere with HCQ transport. Moreover, post-malaria infection, the activation of the SCC/PSAC channel on the erythrocyte membrane occurs, with zinc ions serving as natural inhibitors of this channel (117). Epidemiological studies have further suggested a correlation between zinc deficiency and an elevated risk of malaria infection (118). Building upon existing research on the role of metal ions in anti-infective therapy (119), we propose that the development of zinc-HCQ complexes could facilitate targeted action against infected erythrocytes and plasmodia, potentially reversing drug resistance in malaria parasites and offering a novel strategy for antimalarial treatment. Additionally, several studies have demonstrated the efficacy of HCQ in the treatment of Leishmania infection (120, 121).
4.3 Cancer therapy
As an inhibitor of autophagy, the effectiveness of HCQ in cancer therapy is significantly influenced by the mechanistic context of combination treatment regimens. Empirical studies have demonstrated that HCQ does not enhance the efficacy of DNA-targeting agents—such as temozolomide, gemcitabine in conjunction with nab-paclitaxel, platinum-based drugs, or immune checkpoint inhibitors—likely due to its lack of impact on intracellular drug concentrations or DNA damage checkpoint activity (122) (123) (124). Conversely, HCQ has been shown to exhibit synergistic antitumor effects when used in combination with inhibitors of mTOR (125–129), PI3K (43), EGFR (potentially) (130), and cell cycle-targeted agents (131). The underlying mechanism is attributed to HCQ-induced elevation of lysosomal pH, which subsequently activates pathways such as mTOR (132–135). Inhibition of the mTOR pathway can significantly enhance therapeutic outcomes. Furthermore, HCQ enhances the effectiveness of proliferation pathway inhibitors like sorafenib (136) and has been shown to improve survival rates in certain glioblastoma patients when administered alongside radiotherapy and carmustine (137). These results indicate that in the context of cancer adjuvant therapy, HCQ should be prioritized in conjunction with inhibitors that target pathways it influences, such as the mTOR pathway.
HCQ generally exhibits a favorable safety profile in oncology settings, often demonstrating manageable tolerability when used in conjunction with chemotherapy or targeted agents (138–143). However, the tolerable dosage of HCQ is contingent upon the specific combination regimen employed; it is typically lower in frontline chemotherapy combinations (144), with significant hematologic toxicity possible even at doses of 400 mg twice daily (145). Conversely, higher doses, such as 600 mg twice daily (140) or 800 mg/day (138), have been administered in combination with targeted agents, potentially reflecting an increased systemic burden associated with broad-spectrum anticancer drugs. Pharmacokinetic factors are also of paramount importance: HCQ may influence the absorption and exposure of co-administered drugs, such as by reducing the bioavailability of 2-deoxyglucose (146), increasing the exposure of MK-2206 (147), or heightening the risk of tamoxifen-associated retinopathy (148). Additionally, the absorption of HCQ itself may be adversely affected by proton pump inhibitors (149).
4.4 Other applications of hydroxychloroquine
Additional potential indications have been reported, including alopecia (150, 151), bacterial infections (152–154), oral erosive disorders (155–157), and osteoporosis (158, 159). Moreover, research indicates that HCQ may also confer beneficial effects in the management of gestational diabetes (160), hypertension during pregnancy (161), and specific bacterial infections occurring during pregnancy (162).
5 Outlook
HCQ, known for its antimalarial and immunomodulatory effects, acts as a “double-edged sword” due to its complex mechanisms, such as inhibiting TLR signaling and altering mitochondrial lipids, which offer therapeutic benefits but also pose safety risks. New strategies like chiral drugs and liposomal delivery aim to reduce toxicity, yet personalized medication models are essential. Future research should focus on: (1) Exploring HCQ’s interactions with G-quadruplex nucleic acids and mitochondrial proteins; (2) Examining synergy with immune checkpoint inhibitors; (3) Creating real-time toxicity monitoring. Understanding HCQ’s mechanisms will boost its clinical utility as a “multitarget star” drug.
Author contributions
PW: Writing – review & editing. BD: Writing – original draft. JL: Investigation, Writing – review & editing. LL: Data curation, Investigation, Writing – review & editing. YL: Conceptualization, Writing – review & editing.
Funding
The author(s) declare financial support was received for the research and/or publication of this article. Undergraduate Innovation and Entrepreneurship Project (20251145, 20251118), Changzhi Basic Research Plan Free Exploration Category (JC202413).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that Generative AI was used in the creation of this manuscript. This manuscript was primarily authored by the research team, with AI tools (mainly Deepseek V3) exclusively employed for language refinement (e.g., grammatical correction, sentence structure optimization). The AI did not contribute to the conceptualization, data analysis, or generation of original content, including but not limited to the scientific hypotheses, experimental methods, results interpretation, and conclusions. All intellectual property and scientific contributions remain the sole responsibility of the authors, who have thoroughly verified and validated the entire manuscript content.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Siegel CH and Sammaritano LR. Systemic lupus erythematosus: A review. JAMA. (2024) 331:1480–91. doi: 10.1001/jama.2024.2315
2. Martinez GP, Zabaleta ME, Di Giulio C, Charris JE, and Mijares MR. The role of chloroquine and hydroxychloroquine in immune regulation and diseases. Curr Pharm Des. (2020) 26:4467–85. doi: 10.2174/1381612826666200707132920
3. Ma J, Zeng M, Hsu CJ, Li D, Fok MN, Jiang Y, et al. Efficacy and safety of methotrexate plus hydroxychloroquine combination therapy vs. methotrexate monotherapy in the treatment of rheumatoid arthritis: A randomized controlled clinical trial. Int J Rheum Dis. (2024) 27:e15319. doi: 10.1111/1756-185X.15319
4. Ivey AD, Matthew Fagan B, Murthy P, Lotze MT, Zeh HJ, Hazlehurst LA, et al. Chloroquine reduces neutrophil extracellular trap (NET) formation through inhibition of peptidyl arginine deiminase 4 (PAD4). Clin Exp Immunol. (2023) 211:239–47. doi: 10.1093/cei/uxad005
5. Boretti A. Zinc augments the antiviral potential of HCQ/CQ and ivermectin to reduce the risks of more serious outcomes from COVID-19 infection. J Trace Elem Med Biol. (2022) 71:126954. doi: 10.1016/j.jtemb.2022.126954
6. Yusuf IH, Sharma S, Luqmani R, and Downes SM. Hydroxychloroquine retinopathy. Eye (Lond). (2017) 31:828–45. doi: 10.1038/eye.2016.298
7. Farhat H, Kassab CJ, Tlaiss Y, Gutlapalli SD, Ganipineni VDP, Paramsothy J, et al. Hydroxychloroquine and the associated risk of arrhythmias. Glob Cardiol Sci Pract. (2024) 2024:e202417. doi: 10.21542/gcsp.2024.17
8. Skalafouris C, Samer C, Stirnemann J, Grosgurin O, Eggimann F, Grauser D, et al. Electronic monitoring of potential adverse drug events related to lopinavir/ritonavir and hydroxychloroquine during the first wave of COVID-19. Eur J Hosp Pharm. (2023) 30:113–6. doi: 10.1136/ejhpharm-2020-002667
9. Rainsford KD, Parke AL, Clifford-Rashotte M, and Kean WF. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology. (2015) 23:231–69. doi: 10.1007/s10787-015-0239-y
10. Biguetti CC, Junior JFS, Fiedler MW, Marrelli MT, and Brotto M. The toxic effects of chloroquine and hydroxychloroquine on skeletal muscle: a systematic review and meta-analysis. Sci Rep. (2021) 11:6589. doi: 10.1038/s41598-021-86079-4
11. Stokkermans TJ, Falkowitz DM, and Trichonas G. Chloroquine and hydroxychloroquine toxicity. Treasure Island (FL: StatPearls (2025).
12. Cordova Sanchez A, Khokhar F, Olonoff DA, and Carhart RL. Hydroxychloroquine and cardiovascular events in patients with rheumatoid arthritis. Cardiovasc Drugs Ther. (2024) 38:297–304. doi: 10.1007/s10557-022-07387-z
13. Rempenault C, Combe B, Barnetche T, Gaujoux-Viala C, Lukas C, Morel J, et al. Metabolic and cardiovascular benefits of hydroxychloroquine in patients with rheumatoid arthritis: a systematic review and meta-analysis. Ann Rheum Dis. (2018) 77:98–103. doi: 10.1136/annrheumdis-2017-211836
14. Xuan Y, Gao X, Wang J, Li H, Zhou Z, Liao M, et al. Hydroxychloroquine cures autoimmune myocarditis by inhibiting the innate immune system via the C-X-C motif chemokine ligand 16 and C-X-C motif receptor 6 axis between macrophages and T cells. Br J Pharmacol. (2025) 182(15):3649–3668. doi: 10.1111/bph.70028
15. Istampoulouoglou I, Zimmermanns B, Grandinetti T, Marzolini C, Harings-Kaim A, Koechlin-Lemke S, et al. Cardiovascular adverse effects of lopinavir/ritonavir and hydroxychloroquine in COVID-19 patients: Cases from a single pharmacovigilance centre. Glob Cardiol Sci Pract. (2021) 2021:e202111. doi: 10.21542/gcsp.2021.11
16. Costa EA, Goncalves AP, Batista JAD, Bazoni RF, Santos AA, Rocha MS, et al. New insights into the mechanism of action of the drug chloroquine: direct interaction with DNA and cytotoxicity. J Phys Chem B. (2022) 126:3512–21. doi: 10.1021/acs.jpcb.2c01119
17. Bazoni RF, Moura TA, and Rocha MS. Hydroxychloroquine exhibits a strong complex interaction with DNA: unraveling the mechanism of action. J Phys Chem Lett. (2020) 11:9528–34. doi: 10.1021/acs.jpclett.0c02590
18. Dinc E and Ucer A. A new three-dimensional modeling of fluorescence excitation-emission measurements to explore the interaction of hydroxychloroquine and DNA, and to quantify their binding constant. Colloids Surf B Biointerfaces. (2025) 245:114266. doi: 10.1016/j.colsurfb.2024.114266
19. Sarkar S, Roy S, and Singh PC. Groove switching of hydroxychloroquine modulates the efficacy of binding and induced stability to DNA. J Phys Chem B. (2021) 125:6889–96. doi: 10.1021/acs.jpcb.1c03869
20. Bisoi A, Sarkar S, and Chandra Singh P. Contrasting effect of salts on the binding of antimalarial drug hydroxychloroquine with different sequences of duplex DNA. J Phys Chem B. (2022) 126:5605–12. doi: 10.1021/acs.jpcb.2c02755
21. Lamphier M, Zheng W, Latz E, Spyvee M, Hansen H, Rose J, et al. Novel small molecule inhibitors of TLR7 and TLR9: mechanism of action and efficacy in vivo. Mol Pharmacol. (2014) 85:429–40. doi: 10.1124/mol.113.089821
22. Kuznik A, Bencina M, Svajger U, Jeras M, Rozman B, Jerala R, et al. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J Immunol. (2011) 186:4794–804. doi: 10.4049/jimmunol.1000702
23. Yang LY, Luo Q, Lu L, Zhu WW, Sun HT, Wei R, et al. Increased neutrophil extracellular traps promote metastasis potential of hepatocellular carcinoma via provoking tumorous inflammatory response. J Hematol Oncol. (2020) 13:3. doi: 10.1186/s13045-019-0836-0
24. Guo Y, Gao F, Wang X, Pan Z, Wang Q, Xu S, et al. Spontaneous formation of neutrophil extracellular traps is associated with autophagy. Sci Rep. (2021) 11:24005. doi: 10.1038/s41598-021-03520-4
25. Bisoi A, Majumdar T, Sarkar S, and Singh PC. Flanking effect on the folding of telomeric DNA sequences into G-quadruplex induced by antimalarial drugs. J Phys Chem B. (2025) 129:835–43. doi: 10.1021/acs.jpcb.4c05133
26. Sarkar S, Bisoi A, and Singh PC. Spectroscopic and molecular dynamics aspect of antimalarial drug hydroxychloroquine binding with human telomeric G-quadruplex. J Phys Chem B. (2022) 126:5241–9. doi: 10.1021/acs.jpcb.2c03267
27. Sarkar S, Bisoi A, and Singh PC. Antimalarial drugs induce the selective folding of human telomeric G-quadruplex in a cancer-mimicking microenvironment. J Phys Chem B. (2023) 127:6648–55. doi: 10.1021/acs.jpcb.3c03042
28. Gurung P, Gomes AR, Martins RM, Juranek SA, Alberti P, Mbang-Benet DE, et al. PfGBP2 is a novel G-quadruplex binding protein in Plasmodium falciparum. Cell Microbiol. (2021) 23:e13303. doi: 10.1111/cmi.13303
29. Harris LM, Monsell KR, Noulin F, Famodimu MT, Smargiasso N, Damblon C, et al. G-quadruplex DNA motifs in the malaria parasite plasmodium falciparum and their potential as novel antimalarial drug targets. Antimicrob Agents Chemother. (2018) 62. doi: 10.1128/AAC.01828-17
30. Edwards-Smallbone J, Jensen AL, Roberts LE, Totanes FIG, Hart SR, Merrick CJ, et al. Plasmodium falciparum GBP2 is a telomere-associated protein that binds to G-quadruplex DNA and RNA. Front Cell Infect Microbiol. (2022) 12:782537. doi: 10.3389/fcimb.2022.782537
31. Yadav R, Das S, Mukherjee M, and Mukherjee S. Probing the nucleobase-specific binding interaction of hydroxychloroquine sulfate with RNA and subsequent sequestration by a water-soluble molecular basket. Phys Chem Chem Phys. (2025) 27:7365–74. doi: 10.1039/D4CP04687K
32. Coch C, Hommertgen B, Zillinger T, Dassler-Plenker J, Putschli B, Nastaly M, et al. Human TLR8 senses RNA from plasmodium falciparum-infected red blood cells which is uniquely required for the IFN-gamma response in NK cells. Front Immunol. (2019) 10:371. doi: 10.3389/fimmu.2019.00371
33. Blanco LP, Wang X, Carlucci PM, Torres-Ruiz JJ, Romo-Tena J, Sun HW, et al. RNA externalized by neutrophil extracellular traps promotes inflammatory pathways in endothelial cells. Arthritis Rheumatol. (2021) 73:2282–92. doi: 10.1002/art.41796
34. Hossain MI, Myers M, Herath D, Aldhumani AH, Boesger H, Hines JV, et al. 4-Aminoquinolines modulate RNA structure and function: Pharmacophore implications of a conformationally restricted polyamine. Biochem Biophys Res Commun. (2023) 644:55–61. doi: 10.1016/j.bbrc.2022.12.080
35. Gentile D, Fuochi V, Rescifina A, and Furneri PM. New anti SARS-cov-2 targets for quinoline derivatives chloroquine and hydroxychloroquine. Int J Mol Sci. (2020) 21. doi: 10.3390/ijms21165856
36. Liu J, Zeng C, Hogan V, Zhou S, Monwar MM, Hines JV, et al. Identification of spermidine binding site in T-box riboswitch antiterminator RNA. Chem Biol Drug Des. (2016) 87:182–9. doi: 10.1111/cbdd.12660
37. Armstrong I, Aldhumani AH, Schopis JL, Fang F, Parsons E, Zeng C, et al. RNA drug discovery: Conformational restriction enhances specific modulation of the T-box riboswitch function. Bioorg Med Chem. (2020) 28:115696. doi: 10.1016/j.bmc.2020.115696
38. Qi Y, Zhang Y, Mu Q, Zheng G, Zhang M, Chen B, et al. RNA secondary structurome revealed distinct thermoregulation in plasmodium falciparum. Front Cell Dev Biol. (2021) 9:766532. doi: 10.3389/fcell.2021.766532
39. Dumetz F, Enright AJ, Zhao J, Kwok CK, and Merrick CJ. The in vivo RNA structurome of the malaria parasite Plasmodium falciparum, a protozoan with an A/U-rich transcriptome. PloS One. (2022) 17:e0270863. doi: 10.1371/journal.pone.0270863
40. Qi Y, Zhang Y, Zheng G, Chen B, Zhang M, Li J, et al. In vivo and in vitro genome-wide profiling of RNA secondary structures reveals key regulatory features in plasmodium falciparum. Front Cell Infect Microbiol. (2021) 11:673966. doi: 10.3389/fcimb.2021.673966
41. Dumetz F, Chow EY, Harris LM, Liew SW, Jensen A, Umar MI, et al. G-quadruplex RNA motifs influence gene expression in the malaria parasite Plasmodium falciparum. Nucleic Acids Res. (2021) 49:12486–501. doi: 10.1093/nar/gkab1095
42. Mauthe M, Orhon I, Rocchi C, Zhou X, Luhr M, Hijlkema KJ, et al. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy. (2018) 14:1435–55. doi: 10.1080/15548627.2018.1474314
43. Peng X, Zhang S, Jiao W, Zhong Z, Yang Y, Claret FX, et al. Hydroxychloroquine synergizes with the PI3K inhibitor BKM120 to exhibit antitumor efficacy independent of autophagy. J Exp Clin Cancer Res. (2021) 40:374. doi: 10.1186/s13046-021-02176-2
44. Xie B, Geng Q, Xu J, Lu H, Luo H, Hu Y, et al. The multi-targets mechanism of hydroxychloroquine in the treatment of systemic lupus erythematosus based on network pharmacology. Lupus. (2020) 29:1704–11. doi: 10.1177/0961203320952541
45. Chen Y, Lu M, Lin M, and Gao Q. Network pharmacology and molecular docking to elucidate the common mechanism of hydroxychloroquine treatment in lupus nephritis and IgA nephropathy. Lupus. (2024) 33:347–56. doi: 10.1177/09612033241230377
46. Xie B, Lu H, Xu J, Luo H, Hu Y, Chen Y, et al. Targets of hydroxychloroquine in the treatment of rheumatoid arthritis. A network pharmacology study. Joint Bone Spine. (2021) 88:105099. doi: 10.1016/j.jbspin.2020.105099
47. Vergoten G and Bailly C. Binding of hydroxychloroquine and chloroquine dimers to palmitoyl-protein thioesterase 1 (PPT1) and its glycosylated forms: a computational approach. J Biomol Struct Dyn. (2022) 40:8197–205. doi: 10.1080/07391102.2021.1908167
48. Amin M and Abbas G. Docking study of chloroquine and hydroxychloroquine interaction with RNA binding domain of nucleocapsid phospho-protein - an in silico insight into the comparative efficacy of repurposing antiviral drugs. J Biomol Struct Dyn. (2021) 39:4243–55. doi: 10.1080/07391102.2020.1775703
49. Fu J, Jia Q, Zhou H, Zhang L, Wang S, Liang P, et al. Cell membrane chromatography for the analysis of the interaction between chloroquine and hydroxychloroquine with ACE2 receptors. J Chromatogr B Analyt Technol BioMed Life Sci. (2021) 1162:122469. doi: 10.1016/j.jchromb.2020.122469
50. Navya VB and Hosur MV. A computational study on hydroxychloroquine binding to target proteins related to SARS-COV-2 infection. Inform Med Unlocked. (2021) 26:100714. doi: 10.1016/j.imu.2021.100714
51. Zhao J, Zhao Z, Hou W, Jiang Y, Liu G, Ren X, et al. Quantitative proteomics explore the potential targets and action mechanisms of hydroxychloroquine. Molecules. (2022) 27. doi: 10.3390/molecules27165175
52. Gan W, Zhang C, Siu KY, et al. ULK1 phosphorylates Sec23A and mediates autophagy-induced inhibition of ER-to-Golgi traffic. BMC Cell Biol. (2017) 18:22. doi: 10.1186/s12860-017-0138-8
53. Holdgaard SG, Cianfanelli V, Pupo E, et al. Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellites. Nat Commun. (2019) 10:4176. doi: 10.1038/s41467-019-12094-9
54. Evans AS, Lennemann NJ, and Coyne CB. BPIFB3 interacts with ARFGAP1 and TMED9 to regulate non-canonical autophagy and RNA virus infection. J Cell Sci. (2021) 134. doi: 10.1242/jcs.251835
55. Belizna C. Hydroxychloroquine as an anti-thrombotic in antiphospholipid syndrome. Autoimmun Rev. (2015) 14:358–62. doi: 10.1016/j.autrev.2014.12.006
56. Erkan D, Unlu O, Sciascia S, Belmont HM, Branch DW, Cuadrado MJ, et al. Hydroxychloroquine in the primary thrombosis prophylaxis of antiphospholipid antibody positive patients without systemic autoimmune disease. Lupus. (2018) 27:399–406. doi: 10.1177/0961203317724219
57. Fierro JJ, Velasquez-Berrio M, Ospina A, Henning S, de Leeuw K, Cadavid JA, et al. The effects of hydroxychloroquine and its promising use in refractory obstetric antiphospholipid syndrome. Rheumatol Int. (2024) 44:223–34. doi: 10.1007/s00296-023-05457-5
58. Mekinian A, Vicaut E, Cohen J, Bornes M, Kayem G, Fain O, et al. Hydroxychloroquine to obtain pregnancy without adverse obstetrical events in primary antiphospholipid syndrome: French phase II multicenter randomized trial, HYDROSAPL. Gynecol Obstet Fertil Senol. (2018) 46:598–604. doi: 10.1016/j.gofs.2018.06.008
59. Rand JH, Wu XX, Quinn AS, Chen PP, Hathcock JJ, Taatjes DJ, et al. Hydroxychloroquine directly reduces the binding of antiphospholipid antibody-beta2-glycoprotein I complexes to phospholipid bilayers. Blood. (2008) 112:1687–95. doi: 10.1182/blood-2008-03-144204
60. Hsieh MS, Liu HW, Guo FY, Song DP, Li MY, Chao TY, et al. S-hydroxychloroquine prevents the antiphospholipid thrombogenic complexes for antiphospholipid syndrome treatment. BioMed Pharmacother. (2025) 186:117968. doi: 10.1016/j.biopha.2025.117968
61. Rand JH, Wu XX, Quinn AS, Ashton AW, Chen PP, Hathcock JJ, et al. Hydroxychloroquine protects the annexin A5 anticoagulant shield from disruption by antiphospholipid antibodies: evidence for a novel effect for an old antimalarial drug. Blood. (2010) 115:2292–9. doi: 10.1182/blood-2009-04-213520
62. Liu J, Zhang Y, Song H, Ma M, Li Z, Zhang L, et al. The efficacy of hydroxychloroquine in paediatric chronic immune thrombocytopenia: A retrospective cohort study. Br J Clin Pharmacol. (2025) 91:1449–56. doi: 10.1111/bcp.16389
63. Halcrow PW, Geiger JD, and Chen X. Overcoming chemoresistance: altering pH of cellular compartments by chloroquine and hydroxychloroquine. Front Cell Dev Biol. (2021) 9:627639. doi: 10.3389/fcell.2021.627639
64. Zhou X, Su F, Lu H, Senechal-Willis P, Tian Y, Johnson RH, et al. An FRET-based ratiometric chemosensor for in vitro cellular fluorescence analyses of pH. Biomaterials. (2012) 33:171–80. doi: 10.1016/j.biomaterials.2011.09.053
65. Poole B and Ohkuma S. Effect of weak bases on the intralysosomal pH in mouse peritoneal macrophages. J Cell Biol. (1981) 90:665–9. doi: 10.1083/jcb.90.3.665
66. Xia MC, Cai L, Zhang S, and Zhang X. A cell-penetrating ratiometric probe for simultaneous measurement of lysosomal and cytosolic pH change. Talanta. (2018) 178:355–61. doi: 10.1016/j.talanta.2017.09.044
67. Zhang Z, Yue P, Lu T, Wang Y, Wei Y, Wei X, et al. Role of lysosomes in physiological activities, diseases, and therapy. J Hematol Oncol. (2021) 14:79. doi: 10.1186/s13045-021-01087-1
68. Rudnik S and Damme M. The lysosomal membrane-export of metabolites and beyond. FEBS J. (2021) 288:4168–82. doi: 10.1111/febs.15602
69. Sarkar S and Singh PC. Selective action of antimalarial hydroxychloroquine on the packing of phospholipids and interfacial water associated with lysosomal model membranes: A vibrational sum frequency generation study. Langmuir. (2023) 39:2435–43. doi: 10.1021/acs.langmuir.2c03321
70. Thakur N, Ray AP, Sharp L, Jin B, Duong A, Pour NG, et al. Anionic phospholipids control mechanisms of GPCR-G protein recognition. Nat Commun. (2023) 14:794. doi: 10.1038/s41467-023-36425-z
71. Klouda CB and Stone WL. Oxidative stress, proton fluxes, and chloroquine/hydroxychloroquine treatment for COVID-19. Antioxidants (Basel). (2020) 9. doi: 10.3390/antiox9090894
72. Seydi E, Hassani MK, Naderpour S, Arjmand A, and Pourahmad J. Cardiotoxicity of chloroquine and hydroxychloroquine through mitochondrial pathway. BMC Pharmacol Toxicol. (2023) 24:26. doi: 10.1186/s40360-023-00666-x
73. Casares D, Escriba PV, Rossello CA, et al. Membrane lipid composition: effect on membrane and organelle structure, function and compartmentalization and therapeutic avenues. Int J Mol Sci. (2019) 20. doi: 10.3390/ijms20092167
74. Daum G and Vance JE. Import of lipids into mitochondria. Prog Lipid Res. (1997) 36:103–30. doi: 10.1016/S0163-7827(97)00006-4
75. Horvath SE and Daum G. Lipids of mitochondria. Prog Lipid Res. (2013) 52:590–614. doi: 10.1016/j.plipres.2013.07.002
76. Xu Y, Erdjument-Bromage H, Phoon CKL, Neubert TA, Ren M, Schlame M, et al. Cardiolipin remodeling enables protein crowding in the inner mitochondrial membrane. EMBO J. (2021) 40:e108428. doi: 10.15252/embj.2021108428
77. Kamerbeek NM, van Zwieten R, de Boer M, Morren G, Vuil H, Bannink N, et al. Molecular basis of glutathione reductase deficiency in human blood cells. Blood. (2007) 109:3560–6. doi: 10.1182/blood-2006-08-042531
78. Nadeem U, Raafey M, Kim G, Treger J, Pytel P, A NH, et al. Chloroquine- and hydroxychloroquine-induced cardiomyopathy: A case report and brief literature review. Am J Clin Pathol. (2021) 155:793–801. doi: 10.1093/ajcp/aqaa253
79. Deepalakshmi PD, Parasakthy K, Shanthi S, and Devaraj NS. Effect of chloroquine on rat liver mitochondria. Indian J Exp Biol. (1994) 32:797–9.
80. Hartmann M, Meek IL, van Houwelingen GK, Lambregts HP, Toes GJ, van der Wal AC, et al. Acute left ventricular failure in a patient with hydroxychloroquine-induced cardiomyopathy. Neth Heart J. (2011) 19:482–5. doi: 10.1007/s12471-011-0185-2
81. Wang R, Xi L, and Kukreja RC. PDE5 inhibitor tadalafil and hydroxychloroquine cotreatment provides synergistic protection against type 2 diabetes and myocardial infarction in mice. J Pharmacol Exp Ther. (2017) 361:29–38. doi: 10.1124/jpet.116.239087
82. Qu F, Wang P, Zhang K, Shi Y, Li Y, Li C, et al. Manipulation of Mitophagy by "All-in-One" nanosensitizer augments sonodynamic glioma therapy. Autophagy. (2020) 16:1413–35. doi: 10.1080/15548627.2019.1687210
83. Yin S, Xia C, Wang Y, Wan D, Rao J, Tang X, et al. Dual receptor recognizing liposomes containing paclitaxel and hydroxychloroquine for primary and metastatic melanoma treatment via autophagy-dependent and independent pathways. J Control Release. (2018) 288:148–60. doi: 10.1016/j.jconrel.2018.08.015
84. Zhang YW, Hou LS, Xing JH, Zhang TR, Zhou SY, Zhang BL, et al. Two-membrane hybrid nanobiomimetic delivery system for targeted autophagy inhibition of activated hepatic stellate cells to synergistically treat liver fibrosis. ACS Appl Mater Interfaces. (2023) 15(44):50863–50877. doi: 10.1021/acsami.3c11046
85. Liu L, Ren J, He Z, Men K, Mao Y, Ye T, et al. Cholesterol-modified hydroxychloroquine-loaded nanocarriers in bleomycin-induced pulmonary fibrosis. Sci Rep. (2017) 7:10737. doi: 10.1038/s41598-017-11450-3
86. Dragowska WH, Singh J, Wehbe M, Anantha M, Edwards K, Gorski SM, et al. Liposomal formulation of hydroxychloroquine can inhibit autophagy in vivo. Pharmaceutics. (2024) 17. doi: 10.3390/pharmaceutics17010042
87. Ren K, He J, Qiu Y, Xu Z, Wang X, Li J, et al. A neutrophil-mediated carrier regulates tumor stemness by inhibiting autophagy to prevent postoperative triple-negative breast cancer recurrence and metastasis. Acta Biomater. (2022) 145:185–99. doi: 10.1016/j.actbio.2022.04.017
88. Feng Y, Pan X, Li Z, Li Y, Sun Y, Yang S, et al. Innovative lipid nanoparticles co-delivering hydroxychloroquine and siRNA for enhanced rheumatoid arthritis therapy. Pharmaceutics. (2025) 17. doi: 10.3390/pharmaceutics17010045
89. Ballet V, Bohme GA, Brohan E, Boukaiba R, Chambard JM, Angouillant-Boniface O, et al. In vitro ion channel profile and ex vivo cardiac electrophysiology properties of the R(-) and S(+) enantiomers of hydroxychloroquine. Eur J Pharmacol. (2022) 915:174670. doi: 10.1016/j.ejphar.2021.174670
90. Janicki PK, Singh A, Sharma AK, and Ruiz-Velasco V. Dissimilar effects of stereoisomers and racemic hydroxychloroquine on Ca(2+) oscillations in human induced pluripotent stem cell-derived cardiomyocytes. Physiol Rep. (2023) 11:e15760. doi: 10.14814/phy2.15760
91. Ducharme J, Wainer IW, Parenteau HI, and Rodman JH. Stereoselective distribution of hydroxychloroquine in the rabbit following single and multiple oral doses of the racemate and the separate enantiomers. Chirality. (1994) 6:337–46. doi: 10.1002/chir.530060418
92. Ni Y, Liao J, Qian Z, Wu C, Zhang X, Zhang J, et al. Synthesis and evaluation of enantiomers of hydroxychloroquine against SARS-CoV-2 in vitro. Bioorg Med Chem. (2022) 53:116523. doi: 10.1016/j.bmc.2021.116523
93. Jia Q, Fu J, Liang P, Wang S, Wang Y, Zhang X, et al. Investigating interactions between chloroquine/hydroxychloroquine and their single enantiomers and angiotensin-converting enzyme 2 by a cell membrane chromatography method. J Sep Sci. (2022) 45:456–67. doi: 10.1002/jssc.202100570
94. Wainer IW, Chen JC, Parenteau H, Abdullah S, Ducharme J, Fieger H, et al. Distribution of the enantiomers of hydroxychloroquine and its metabolites in ocular tissues of the rabbit after oral administration of racemic-hydroxychloroquine. Chirality. (1994) 6:347–54. doi: 10.1002/chir.530060419
95. Tett SE, McLachlan AJ, Cutler DJ, and Day RO. Pharmacokinetics and pharmacodynamics of hydroxychloroquine enantiomers in patients with rheumatoid arthritis receiving multiple doses of racemate. Chirality. (1994) 6:355–9. doi: 10.1002/chir.530060420
96. Moini A, Sepidarkish M, Dehpour AR, Rabiei M, Abiri A, Pirjani R, et al. The effect of hydroxychloroquine on pregnancy outcomes in patients with unexplained recurrent pregnancy loss: a placebo-controlled study "pilot study. J Obstet Gynaecol. (2022) 42:3471–6. doi: 10.1080/01443615.2022.2141615
97. Chen Y, Xu W, Huang S, Li J, Li T, Chen J, et al. Analysis of pregnancy outcomes in patients exhibiting recurrent miscarriage with concurrent low-titer antiphospholipid antibodies. Am J Reprod Immunol. (2024) 92:e13940. doi: 10.1111/aji.13940
98. Kim YM, Sung JH, Cha HH, and Oh SY. Hydroxychloroquine in obstetrics: potential implications of the prophylactic use of hydroxychloroquine for placental insufficiency during pregnancy. Obstet Gynecol Sci. (2024) 67:143–52. doi: 10.5468/ogs.23252
99. Soto-Peleteiro A, Gonzalez-Echavarri C, and Ruiz-Irastorza G. Obstetric antiphospholipid syndrome. Med Clin (Barc). (2024) 163 Suppl 1:S14–21. doi: 10.1016/j.medcli.2024.05.001
100. Geng L, Pan K, Xu Y, Zhang B, Wang J, Xue Q, et al. Antiphospholipid antibodies as potential prognostic indicators of recurrent ischemic stroke. J Stroke Cerebrovasc Dis. (2024) 33:107885. doi: 10.1016/j.jstrokecerebrovasdis.2024.107885
101. Chen CH, Chen HA, Liao HT, and Chen CH. Hydroxychloroquine exposure reduces the risk of cardiovasular disease events in patients with hypertension or diabetes mellitus. Clin Exp Rheumatol. (2023) 41:2230–8. doi: 10.55563/clinexprheumatol/y7psev
102. Ahn SM, Kim S, Kim YJ, Hong S, Lee CK, Yoo B, et al. Risk of ischemic stroke associated with anti-rheumatic agents in patients with rheumatoid arthritis: A nationwide population-based case-control study. PloS One. (2025) 20:e0326311. doi: 10.1371/journal.pone.0326311
103. Peng W, Guo K, Hu J, and Wang Q. Inhibition of pyroptosis by hydroxychloroquine as a neuroprotective strategy in ischemic stroke. eNeuro. (2024) 12. doi: 10.1523/ENEURO.0254-24.2024
104. Vermeer B, Veltkamp M, Raasing LRM, Wind AE, and Vorselaars ADM. Hydroxychloroquine monotherapy in sarcoidosis: Indications, efficacy, and side effects. Sarcoidosis Vasc Diffuse Lung Dis. (2024) 41:e2024039. doi: 10.36141/svdld.v41i3.15445
105. Mayovska OV, Rosenstein ED, and Kramer N. Nodular sarcoid myositis treated with hydroxychloroquine monotherapy. Cureus. (2023) 15:e41000. doi: 10.7759/cureus.41000
106. Geladari V, Liaveri P, Liapis G, Moustakas G, and Sabanis N. Systemic sarcoidosis presenting with hypercalcemia, acute kidney injury, and diffuse lymphadenopathy: unlocking pandora's box. Cureus. (2025) 17:e85882. doi: 10.7759/cureus.85882
107. Cohen E, Lheure C, Ingen-Housz-Oro S, Hotz C, Bettuzzi T, Chasset F, et al. Which first-line treatment for cutaneous sarcoidosis? A retrospective study of 120 patients. Eur J Dermatol. (2023) 33:680–5. doi: 10.1684/ejd.2023.4584
108. Harris RE, Thomas VL, Underwood JM, and Gaudot F. A comparison of two indirect methods for localizing the site of urinary infection: beta glucuronidase levels and the presence of antibody-coated bacteria. Am J Obstet Gynecol. (1978) 131:647–9. doi: 10.1016/0002-9378(78)90825-6
109. Ozbay AD, Somuncu AM, Cicek I, Yavuzer B, Bulut S, Huseynova G, et al. Effects of adenosine triphosphate and coenzyme Q10 on potential hydroxychloroquine-induced retinal damage in rats. Exp Eye Res. (2025) 255:110387. doi: 10.1016/j.exer.2025.110387
110. Kulkarni AB, Renapurkar DM, and Sharma KD. Hepatic superoxide dismutase activity in the mouse infected with Plasmodium berghei. Folia Parasitol (Praha). (1984) 31:89–91.
111. Dabravolski SA, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN, et al. Interplay between zn(2+) homeostasis and mitochondrial functions in cardiovascular diseases and heart ageing. Int J Mol Sci. (2022) 23. doi: 10.3390/ijms23136890
112. Campos-Blazquez JP, Schuth N, Garay E, Clark AH, Vogelsang U, Nachtegaal M, et al. Chloroquine disrupts zinc storage granules in primary Malpighian tubule cells of Drosophila melanogaster. Metallomics. (2022) 14. doi: 10.1093/mtomcs/mfac075
113. Ibrahim NA, Buabeid MA, Arafa S, and Elmorshedy KE. Zinc's protective role against hydroxychloroquine-induced cardiac effects in adult male albino rats. Saudi J Biol Sci. (2023) 30:103733. doi: 10.1016/j.sjbs.2023.103733
114. Pages M and Pages AM. Peripheral nerve lesions in chloroquine-induced neuromyopathies. Ann Pathol. (1984) 4:289–95.
115. Squarcina A, Franke A, Senft L, Onderka C, Langer J, Vignane T, et al. Zinc complexes of chloroquine and hydroxychloroquine versus the mixtures of their components: Structures, solution equilibria/speciation and cellular zinc uptake. J Inorg Biochem. (2024) 252:112478. doi: 10.1016/j.jinorgbio.2024.112478
116. Paulikat M, Vitone D, Schackert FK, Schuth N, Barbanente A, Piccini G, et al. Molecular dynamics and structural studies of zinc chloroquine complexes. J Chem Inf Model. (2023) 63:161–72. doi: 10.1021/acs.jcim.2c01164
117. Bouyer G, Egee S, and Thomas SL. Toward a unifying model of malaria-induced channel activity. Proc Natl Acad Sci U.S.A. (2007) 104:11044–9. doi: 10.1073/pnas.0704582104
118. Pullakhandam R, Agrawal PK, Peter R, Ghosh S, Reddy GB, Kulkarni B, et al. Prevalence of low serum zinc concentrations in Indian children and adolescents: findings from the Comprehensive National Nutrition Survey 2016–18. Am J Clin Nutr. (2021) 114:638–48. doi: 10.1093/ajcn/nqab066
119. Rehder D, Black RE, Bornhorst J, Dietert RR, DiRita VJ, Navarro M, et al. Metals in Host-Microbe Interaction: The Host Perspective. In: Trace Metals and Infectious Diseases. Cambridge (MA (2015).
120. Noaimi AA Sr and Mohamed MC Sr. The treatment of acute cutaneous leishmaniasis with oral zinc bisglycinate and oral hydroxychloroquine sulfate. Cureus. (2025) 17:e84618. doi: 10.7759/cureus.84618
121. Avanzo RE, Garcia Linares G, Rodriguez N, and Romero AH. A comprehensive revision on the use of quinoline antimalarial drugs as leishmanicidal agents. Front Chem. (2025) 13:1608340. doi: 10.3389/fchem.2025.1608340
122. Rangwala R, Leone R, Chang YC, Fecher LA, Schuchter LM, Kramer A, et al. Phase I trial of hydroxychloroquine with dose-intense temozolomide in patients with advanced solid tumors and melanoma. Autophagy. (2014) 10:1369–79. doi: 10.4161/auto.29118
123. Liu P, Zhao L, Ferrere G, Alves-Costa-Silva C, Ly P, Wu Q, et al. Combination treatments with hydroxychloroquine and azithromycin are compatible with the therapeutic induction of anticancer immune responses. Oncoimmunology. (2020) 9:1789284. doi: 10.1080/2162402X.2020.1789284
124. Wabitsch S, McVey JC, Ma C, Ruf B, Kamenyeva O, McCallen JD, et al. Hydroxychloroquine can impair tumor response to anti-PD1 in subcutaneous mouse models. iScience. (2021) 24:101990. doi: 10.1016/j.isci.2020.101990
125. Masaki N, Aoki Y, Kubota Y, Obara K, Miyazaki J, Hoffman RM, et al. Chloroquine combined with rapamycin arrests tumor growth in a patient-derived orthotopic xenograft (PDOX) mouse model of dedifferentiated liposarcoma. In Vivo. (2022) 36:2630–7. doi: 10.21873/invivo.12997
126. Elshazly AM, Elzahed AA, and Gewirtz DA. The cytoprotective and cytotoxic functions of autophagy in response to mTOR inhibitors. Front Biosci (Landmark Ed). (2024) 29:231. doi: 10.31083/j.fbl2906231
127. Li HP, Huang CY, Lui KW, Chao YK, Yeh CN, Lee LY, et al. Nasopharyngeal carcinoma patient-derived xenograft mouse models reveal potential drugs targeting cell cycle, mTOR, and autophagy pathways. Transl Oncol. (2023) 38:101785. doi: 10.1016/j.tranon.2023.101785
128. Haas NB, Appleman LJ, Stein M, Redlinger M, Wilks M, Xu X, et al. Autophagy inhibition to augment mTOR inhibition: a phase I/II trial of everolimus and hydroxychloroquine in patients with previously treated renal cell carcinoma. Clin Cancer Res. (2019) 25:2080–7. doi: 10.1158/1078-0432.CCR-18-2204
129. Scott EC, Maziarz RT, Spurgeon SE, Medvedova E, Gajewski J, Reasor-Heard S, et al. Double autophagy stimulation using chemotherapy and mTOR inhibition combined with hydroxychloroquine for autophagy modulation in patients with relapsed or refractory multiple myeloma. Haematologica. (2017) 102:e261–e5. doi: 10.3324/haematol.2016.162321
130. Jutten B, Keulers TG, Peeters HJM, Schaaf MBE, Savelkouls KGM, Compter I, et al. EGFRvIII expression triggers a metabolic dependency and therapeutic vulnerability sensitive to autophagy inhibition. Autophagy. (2018) 14:283–95. doi: 10.1080/15548627.2017.1409926
131. Gong C, Lin Q, Qin T, Zeng Y, Xu F, Yang Y, et al. Targeting autophagy plus high-dose CDK4/6 inhibitors in advanced HR+HER2- breast cancer: A phase 1b/2 trial. Med. (2025) 6:100559. doi: 10.1016/j.medj.2024.11.012
132. Hu Y, Carraro-Lacroix LR, Wang A, Owen C, Bajenova E, Corey PN, et al. Lysosomal pH Plays a Key Role in Regulation of mTOR Activity in Osteoclasts. J Cell Biochem. (2016) 117:413–25. doi: 10.1002/jcb.25287
133. Jia J, Wang F, Bhujabal Z, Peters R, Mudd M, Duque T, et al. Membrane Atg8ylation, stress granule formation, and MTOR regulation during lysosomal damage. Autophagy. (2023) 19:1893–5. doi: 10.1080/15548627.2022.2148900
134. Liu X, Guo B, Li Q, and Nie J. mTOR in metabolic homeostasis and disease. Exp Cell Res. (2024) 441:114173. doi: 10.1016/j.yexcr.2024.114173
135. Glaviano A, Foo ASC, Lam HY, Yap KCH, Jacot W, Jones RH, et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. (2023) 22:138. doi: 10.1186/s12943-023-01827-6
136. Chen MY, Yadav VK, Chu YC, Ong JR, Huang TY, Lee KF, et al. Hydroxychloroquine (HCQ) modulates autophagy and oxidative DNA damage stress in hepatocellular carcinoma to overcome sorafenib resistance via TLR9/SOD1/hsa-miR-30a-5p/beclin-1 axis. Cancers (Basel). (2021) 13. doi: 10.3390/cancers13133227
137. Compter I, Eekers DBP, Hoeben A, Rouschop KMA, Reymen B, Ackermans L, et al. Chloroquine combined with concurrent radiotherapy and temozolomide for newly diagnosed glioblastoma: a phase IB trial. Autophagy. (2021) 17:2604–12. doi: 10.1080/15548627.2020.1816343
138. Raghavendra AS, Kettner NM, Kwiatkowski D, Damodaran S, Wang Y, Ramirez D, et al. Phase I trial of hydroxychloroquine to enhance palbociclib and letrozole efficacy in ER+/HER2- breast cancer. NPJ Breast Cancer. (2025) 11:7. doi: 10.1038/s41523-025-00722-1
139. Goenka L, Dubashi B, Kayal S, Rajappa M, Manivannan P, Chakkalakkoombil SV, et al. Targeting autophagy in platinum-sensitive relapsed ovarian cancer: randomized phase II trial of hydroxychloroquine with chemotherapy with biomarker correlation. Discov Oncol. (2025) 16:203. doi: 10.1007/s12672-025-01904-w
140. Abdel Karim N, Eldessouki I, Ahmed I, Gaber O, Bahassi E, Khaled A, et al. A Phase I Trial to Determine the Safety and Tolerability of Autophagy Inhibition Using Chloroquine or Hydroxychloroquine in Combination with Carboplatin and Gemcitabine in Patients with Advanced Solid Tumorsced solid tumors. Asian Pac J Cancer Prev. (2025) 26:1165–72. doi: 10.31557/APJCP.2025.26.4.1165
141. Witte D, Pretzell I, Reissig TM, Stein A, Velthaus JL, Alig A, et al. Trametinib in combination with hydroxychloroquine or palbociclib in advanced metastatic pancreatic cancer: data from a retrospective, multicentric cohort (AIO AIO-TF/PAK-0123). J Cancer Res Clin Oncol. (2024) 150:438. doi: 10.1007/s00432-024-05954-5
142. Agalakova NI. Chloroquine and chemotherapeutic compounds in experimental cancer treatment. Int J Mol Sci. (2024) 25. doi: 10.3390/ijms25020945
143. Malhotra J, Jabbour S, Orlick M, Riedlinger G, Guo Y, White E, et al. Phase Ib/II study of hydroxychloroquine in combination with chemotherapy in patients with metastatic non-small cell lung cancer (NSCLC). Cancer Treat Res Commun. (2019) 21:100158. doi: 10.1016/j.ctarc.2019.100158
144. Arora SP, Tenner L, Sarantopoulos J, Morris J, Liu Q, Mendez JA, et al. Modulation of autophagy: a Phase II study of vorinostat plus hydroxychloroquine versus regorafenib in chemotherapy-refractory metastatic colorectal cancer (mCRC). Br J Cancer. (2022) 127:1153–61. doi: 10.1038/s41416-022-01892-6
145. Karim NA, Ullah A, Ahmad I, Bahassi E, Olowokure O, Khaled A, et al. A phase I trial to determine the safety and tolerability of autophagy inhibition using chloroquine or hydroxychloroquine in combination with carboplatin and gemcitabine in patients with advanced solid tumors. Front Oncol. (2022) 12:811411. doi: 10.3389/fonc.2022.811411
146. Sun D, Kim S, Karelia D, Deng Y, Jiang C, Lu J, et al. 2-Deoxyglucose and hydroxychloroquine HPLC-MS-MS analytical methods and pharmacokinetic interactions after oral co-administration in male rats. Pharmacol Res Perspect. (2024) 12:e1173. doi: 10.1002/prp2.1173
147. Mehnert JM, Kaveney AD, Malhotra J, Spencer K, Portal D, Goodin S, et al. A phase I trial of MK-2206 and hydroxychloroquine in patients with advanced solid tumors. Cancer Chemother Pharmacol. (2019) 84:899–907. doi: 10.1007/s00280-019-03919-x
148. Toimela T, Tahti H, and Salminen L. Retinal pigment epithelium cell culture as a model for evaluation of the toxicity of tamoxifen and chloroquine. Ophthalmic Res. (1995) 27 Suppl 1:150–3. doi: 10.1159/000267861
149. Namazi MR. The potential negative impact of proton pump inhibitors on the immunopharmacologic effects of chloroquine and hydroxychloroquine. Lupus. (2009) 18:104–5. doi: 10.1177/0961203308097574
150. Stege H, Haist M, Schultheis M, Pawlowski J, Wittmann M, Grabbe S, et al. Treatment of lichen planopilaris and frontal fibrosing alopecia: A retrospective, real-life analysis in a tertiary center in Germany. J Clin Med. (2024) 13. doi: 10.3390/jcm13164947
151. Valdez-Zertuche JA, Ramirez-Marin HA, and Tosti A. Efficacy, safety and tolerability of drugs for alopecia: a comprehensive review. Expert Opin Drug Metab Toxicol. (2025) 21:347–71. doi: 10.1080/17425255.2025.2461483
152. Basrani ST, Gavandi TC, Patil SB, Kadam NS, Yadav DV, Chougule SA, et al. Hydroxychloroquine an antimalarial drug, exhibits potent antifungal efficacy against candida albicans through multitargeting. J Microbiol. (2024) 62:381–91. doi: 10.1007/s12275-024-00111-6
153. Zhou H and Zhang J. Short-term amoxicillin clavulanate in the treatment of pulmonary abscess caused by tropheryma whipplei infection diagnosed by targeted next-generation sequencing: A case report and literature review. Infect Drug Resist. (2024) 17:4607–16. doi: 10.2147/IDR.S488740
154. El Zein S, Challener DW, Ranganath N, Khodadadi RB, Theel ES, Abu Saleh OM, et al. Acute Coxiella burnetii Infection: A 10-Year Clinical Experience at a Tertiary Care Center in the United States. Open Forum Infect Dis. (2024) 11:ofae277. doi: 10.1093/ofid/ofae277
155. Alnajjar NF, Tahan SR, and Iriarte C. Immunotherapy-associated bullous and oral erosive lichenoid eruption successfully treated with hydroxychloroquine. JAAD Case Rep. (2025) 56:63–6. doi: 10.1016/j.jdcr.2024.11.010
156. Tillero R, Gonzalez-Serrano J, Caponio VCA, Serrano J, Hernandez G, Lopez-Pintor RM, et al. Efficacy of antimalarials in oral lichen planus: A systematic review. Oral Dis. (2024) 30:4098–112. doi: 10.1111/odi.14975
157. Abdusalamova K, Worm M, and Solimani F. Hydroxychloroquine is safe and efficacious in oral lichen planus: data from a large outpatient cohort. Arch Dermatol Res. (2025) 317:711. doi: 10.1007/s00403-025-04226-7
158. Yao Z, Ayoub A, Srinivasan V, Wu J, Tang C, Duan R, et al. Hydroxychloroquine and a low antiresorptive activity bisphosphonate conjugate prevent and reverse ovariectomy-induced bone loss in mice through dual antiresorptive and anabolic effects. Bone Res. (2024) 12:52. doi: 10.1038/s41413-024-00352-6
159. Dong C, Chen BS, Wu CH, Chiu YM, Liao PL, Perng WT, et al. Hydroxychloroquine and risk of osteoporosis in patients with rheumatoid arthritis: A population-based retrospective study of 6408 patients. Int J Rheum Dis. (2024) 27:e15286. doi: 10.1111/1756-185X.15286
160. Basri NI, Murthi P, and Abd Rahman R. Hydroxychloroquine as an adjunct therapy for diabetes in pregnancy. Int J Mol Sci. (2024) 25. doi: 10.3390/ijms25179681
161. Kang X, Chen W, Hong S, Lio KU, Shi H, Wang J, et al. Hydroxychloroquine for preventing hypertensive pregnancy disorders in recurrent spontaneous abortion: a retrospective cohort study in a single referral center. Eur J Med Res. (2025) 30:307. doi: 10.1186/s40001-025-02549-9
Keywords: hydroxychloroquine, autophagy, mitochondrial damage, nucleic acid targeting, membrane structure remodeling, zinc homeostasis, clinical application
Citation: Du B, Li L, Li J, Liu Y and Wang P (2025) Hydroxychloroquine’s diverse targets: a new frontier in precision medicine. Front. Immunol. 16:1655573. doi: 10.3389/fimmu.2025.1655573
Received: 28 June 2025; Accepted: 16 September 2025;
Published: 08 October 2025.
Edited by:
Cinzia Milito, Sapienza University of Rome, ItalyReviewed by:
Stefania Nicola, University of Turin, ItalyCopyright © 2025 Du, Li, Li, Liu and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Pu Wang, MzcyNzQ2NTI5QHFxLmNvbQ==