Abstract
Itaconate, an endogenous immunomodulator from the tricarboxylic acid (TCA) cycle, shows therapeutic effects in various disease models, but is highly polar with poor cellular permeability. We previously reported a novel, topical itaconate derivative, SCD-153, for the treatment of alopecia areata. Here, we present the discovery of orally available itaconate derivatives for systemic and skin disorders. Four sets of prodrugs were synthesized using pivaloyloxymethyl (POM), isopropyloxycarbonyloxymethyl (POC), (5-methyl-2-oxo-1,3-dioxol-4-yl) methyl (ODOL), and 3-(hexadecyloxy)propyl (HDP) pro-moieties pairing with itaconic acid (IA), 1-methyl itaconate (1-MI), and 4-methyl itaconate (4-MI). Among these, POC-based prodrugs (P2, P9, P13) showed favorable stability, permeability, and pharmacokinetics. Notably, P2 and P13 significantly inhibited Poly(I:C)/IFNγ-induced inflammatory cytokines in human epidermal keratinocytes. Oral studies demonstrated favorable pharmacokinetics releasing micromolar concentrations of IA or 4-MI from P2 and P13, respectively. These findings highlight the potential of prodrug strategies to enhance itaconate’s cellular permeability and oral bioavailability, paving the way for clinical translation.
Graphical Abstract

INTRODUCTION
Itaconic acid (IA, itaconate) is an endogenous immunomodulatory metabolite produced by diverting aconitate from the tricarboxylic acid (TCA) cycle during the activation of inflammatory macrophages. IA is an unsaturated dicarboxylic acid synthesized by immune-responsive gene 1 (IRG1).1 IA acts on multiple inflammatory pathways and is shown to be an essential functional component of activated macrophages.2,3 Despite being a widely used commercial biomaterial derived from Aspergillus for decades, its identification as a mammalian immunometabolite was not established until 2011.2,4 It acts as an immunomodulator through several mechanisms: inhibition of succinate dehydrogenase (SDH),5 activation of Nrf2 via alkylation of KEAP1,6 regulation of ATF3/IκBζ inflammatory axis,7 inhibition of glycolysis,5 regulation of type I IFNs8 and inhibition of NLRP3 inflammasome.9
Itaconate contains an α,β-unsaturated alkene and shares structural similarity with other metabolites like phosphoenolpyruvate, succinate, malonate, and fumarate. There is a growing body of evidence suggesting that itaconate, as well as its derivatives, have promise in various disease models.6,10 Despite promising efficacy in preclinical models, clinical potential of IA is limited due to its highly polar structure, making it impermeable to biological membranes. Several cell-permeable itaconate analogs, including dimethyl itaconate (DMI), 4-octyl itaconate (4-OI), and 4-ethyl itaconate (4-EI) have been developed to mimic the actions of endogenous itaconate. DMI and 4-OI, interestingly have been shown to conserve the anti-inflammatory effects of IA11,12 but fall short in their ability to convert to intracellular IA once delivered.8,13 Additionally, it has been reported that DMI is intracellularly converted into a mixture of 1-methyl itaconate (1-MI) and 4-methyl itaconate (4-MI) and presumably exerts its effects through these active metabolites, rather than itaconate itself.13
The immunomodulatory properties of itaconate and its derivatives make them promising pharmacologic candidates for treating inflammatory conditions such as psoriasis, rheumatoid arthritis, systemic lupus erythematosus, and multiple sclerosis.1,14,15 We have recently designed and reported a topical, cell-permeable prodrug of 4-MI, termed SCD-153 as a novel treatment for alopecia areata,16 which is a chronic autoimmune disorder characterized by the targeted destruction of hair follicles by CD8+ T cells, leading to hair loss. Interestingly, topical administration of SCD-153 to C57BL/6 mice resulted in a significant increase in hair growth, exhibiting statistically superior effects compared to the vehicle (dimethyl sulfoxide), less cell-permeable itaconate analogues (4-MI and DMI), as well as the clinically used JAK inhibitor tofacitinib. SCD-153 demonstrated considerable skin concentrations, indicating that the prodrug strategy has promise in topically delivering IA to previously impermeable tissues.16
Building on the initial strategy, this work aimed to develop orally available, cell-permeable prodrugs of IA or its monoesters, 1-MI and 4-MI, to target tissues such as skin systemically. We employed FDA-approved promoieties such as pivaloyloxymethyl (POM), isopropyloxycarbonyloxymethyl (POC), (5-methyl-2-oxo-1,3-dioxol-4-yl) methyl (ODOL) and 3-(hexadecyloxy)-propyl (HDP) to mask either the 1-carboxylate or 4-carboxylate on itaconate, thereby enhancing its permeability and pharmacokinetic properties.
CHEMISTRY
The synthesis of IA prodrugs focused on three structural types: (a) itaconate diesters with identical ester groups, (b) itaconate monoesters with ester groups at the 4-position of the carboxylate, and (c) itaconate diesters derived from 1-methyl or 4-methyl itaconate, featuring various promoieties at the second carboxylic function. The prodrug groups utilized were FDA approved promoieties, including pivaloyloxymethyl (POM), isopropyloxycarbonyloxymethyl (POC), (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl (ODOL), and 3-(hexadecyloxy)-propyl (HDP) to minimize challenges related to translation and toxicity.17–19 For example, these groups were applied in the prodrug development of acyclic nucleoside phosphonates, including Adefovir, Tenofovir, and Cidofovir which led to substantial improvement in pharmacokinetics and oral absorption.20,21 We have also reported their application in successfully synthesizing prodrugs of the multiply charged GCPII inhibitor 2-(phosphonomethyl)pentanedioic acid (2-PMPA) and demonstrated enhancement in oral bioavailability (20–69 fold) in mice and dogs.22,23
Synthesis of itaconate diesters bis-POM (P1), bis-POC (P2), and bis-ODOL (P3) were performed by alkylation of IA with carbonyloxymethyl chlorides or (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl chloride, respectively, under basic conditions in the presence of sodium iodide (Scheme 1).
Scheme 1. Synthesis of Bis-POM, Bis-POC and Bis-ODOL Dialkyl Esters P1–3a.
aReagents and conditions: (i) K2CO3, NaI, MeCN, 40–55 °C, 16 h, 14–57%.
Selective monoesterification of free IA to POM, POC, or ODOL was challenging; various reaction conditions were attempted, but only a nonseparable mixture of 1- and 4-monoesters were obtained. Thus, to synthesize pure 4-monoesters with a free 1-carboxylate, 4-tert-butyl itaconate (5) was prepared according to a published protocol24 and alkylation was performed under the same conditions as described above. The protected diesters 6–8 were subsequently treated with trifluoroacetic acid to give the desired monoesters P4, P5, and P6 (Scheme 2). Preparation of 3-(hexadecyloxy)propyl monoester (P7) was achieved by reaction of itaconic anhydride (9) with hexadecyloxypropanol (10) at 70 °C in chloroform proceeding selectively to the position 4 (Scheme 2).
Scheme 2. Synthesis of POM, POC, ODOL, and HDP Itaconic Monoesters P4–6 and P7a.
aReagents and conditions: (i) K2CO3, NaI, MeCN, 40–55 °C, 16 h, 71–85%; (ii) trifluoroacetic acid in DCM, rt, 2 h, 84–97%; (iii) CHCl3, 70 °C, 16 h, 83%.
Mixed diesters P8–15 were prepared by alkylation of 1-MI (11) or 4-MI (12) with a chloromethyl derivative (analogously as described for compounds P1–3) or by Steglich esterification with 3-(hexadecyloxy)propanol (compounds P11 and P15, Scheme 3).
Scheme 3. Synthesis of Itaconate Diesters Designed to Release 1-MI or 4-MIa.
aReagents and conditions: (i) K2CO3, NaI, MeCN, 40–45 °C, 16 h, 84–94%; (ii) DCC, DMAP, 0 °C to rt, 16 h, 28–68%.
RESULTS AND DISCUSSION
In Vitro Chemical Stability, Permeability, and Metabolic Stability Assessments.
To develop orally available prodrugs for IA, 1-MI, and 4-MI, it was crucial to achieve high permeability, good chemical stability, and the ability to release active compounds upon oral absorption. Consequently, the prodrugs were assessed for their chemical stability under gastrointestinal pH conditions (1.2, 4.5, and 7.4) to ensure stability in the gut for oral delivery. Those demonstrating good pH stability were further evaluated for their ability to permeate via the parallel artificial membrane permeation assay (PAMPA, GIT) assay.25 Finally, stable and highly permeable prodrugs were assessed for stability in mouse and human plasma, as well as in skin homogenates to assess release of actives at the desired site, as detailed in Table 1 below.
Table 1. Structure, Stability, and Permeability of Itaconate Prodrugs.
| Compound ID | Molecular Structure (Blue=active moiety; Red=promoiety) | ClogP | Chemical Stability (% remaining at 1h) | Permeability (Pe, × 10−6 cm/sec) pH 7.4 | Metabolic Stability (% remaining at 1h) | |||||
|---|---|---|---|---|---|---|---|---|---|---|
| pH 1.2 | pH 4.5 | pH 7.4 | Mouse Plasma | Mouse Skin | Human Plasma | Human Skin | ||||
| P1 |
|
2.74 | 106 ± 9 | 92 ± 3 | 104 ± 17 | 35.8 | 0 ± 0 | 0 ± 0 | 0 ± 0 | 0 ± 0 |
| P4 |
|
1.27 | 100 ± 2 | 74 ± 3 | 22 ± 4 | 0.08 | 3 ± 1 | 14 ± 0.5 | 83 ± 2 | 40 ± 0.7 |
| P8 |
|
1.79 | 101 ± 2 | 104 ± 5 | 86 ± 8 | 35.2 | 0 ± 0 | 0 ± 0 | 0 ± 0 | 9.4 ± 0.9 |
| P12 |
|
1.61 | 99 ± 2 | 109 ± 4 | 94 ± 1 | 33.2 | 0 ± 0 | 5.3 ± 0.4 | 0 ± 0 | 15 ± 2.9 |
| P2 |
|
1.83 | 104 ± 2 | 109 ± 6 | 47 ± 4 | 35.9 | 0 ± 0 | 2.5 ± 2.4 | 0 ± 0 | 1.1 ± 0.4 |
| P5 |
|
0.82 | 104 ± 0 | 79 ± 11 | 44 ± 1 | 0 | 0 ± 0 | 4.9 ± 1.3 | 53 ± 1 | 29 ± 0.9 |
| P9 |
|
1.34 | 98 ± 2 | 94 ± 2 | 63 ± 3 | 29.3 | 0 ± 0 | 0 ± 0 | 0 ± 0 | 0 ± 0 |
| P13 |
|
1.16 | 108 ± 1 | 103 ± 4 | 102 ± 2 | 35.3 | 0 ± 0 | 0 ± 0 | 0 ± 0 | 1.6 ± 0.4 |
| P3 |
|
−0.94 | 102 ± 1 | 91 ± 2 | 91 ± 14 | 17.0 | 0 ± 0 | 0 ± 0 | 0 ± 0 | 30 ± 0.4 |
| P6 |
|
−0.66 | 109 ± 4 | 88 ± 1 | 0 ± 0 | 0 | 20 ± 8 | 74 ± 30 | 0 ± 0 | 93 ± 4 |
| P10 |
|
−0.13 | 100 ± 6 | 97 ± 4 | 69 ± 5 | 16.3 | 0 ± 0 | 0 ± 0 | 0 ± 0 | 56 ± 0.9 |
| P14 |
|
−0.14 | 98 ± 3 | 87 ± 6 | 99 ± 4 | 15.4 | 0 ± 0 | 16 ± 0.4 | 0 ± 0 | 76 ± 4 |
| P7 |
|
8.38 | 91 ± 2 | 90 ± 10 | 65 ± 4 | 3.2 | 92 ± 8 | 100 ± 9.5 | 68 ± 9 | 87 ± 7 |
| P11 |
|
8.79 | 109 ± 14 | 98 ± 4 | 97 ± 4 | 0 | 89 ± 2 | 61 ± 10 | 90 ± 4 | 101 ± 8 |
| P15 |
|
8.91 | 103 ± 12 | 108 ± 16 | 75 ± 8 | 0 | 44 ± 10 | 67 ± 11 | 32 ± 2 | 94 ± 5 |
| IA |
|
−0.33 | 0 | - | - | |||||
| 4-MI |
|
0.14 | 0 | - | - | |||||
| 1-MI |
|
0.20 | 0 | - | - | |||||
All prodrugs were either highly stable (>80% remaining at 1 h) or moderately stable (50–80% remaining at 1 h) across the three pH conditions, with the exception of POM, POC, and ODOL monoesters of IA (P4, P5, and P6 respectively), which all exhibited complete instability at pH 7.4.
The calculated c log P values of the prodrugs expectedly showed significant improvement in lipophilicity, approximately 10-fold for POC and POM, and 50-fold for HDP prodrugs, compared to their respective active moieties (c log P: −0.33, 0.14, and 0.20 for IA, 4-MI, and 1-MI, respectively) (SwissADME, Lausanne, Switzerland).26 Given the polar nature of the ODOL promoieties, its addition did not enhance itaconate calculated lipophilicity. In terms of permeability via PAMPA-GIT assay to mimic permeability via the gastrointestinal tract, bis-POM and bis-POC-protected IA prodrugs (P1, P2), as well as POM/POC prodrugs of 1-MI (P8, P9) and 4-MI (P12, P13), demonstrated excellent in vitro permeability with a permeation index (Pe) greater than 10 × 10−6 cm/s. These results are consistent with the increase in their c log P, aligning with previous studies showing that permeability increases with lipophilicity when cLogP values are 3.5 or lower.27 Conversely, POM or POC monoesters of IA (P4, P5), as expected, displayed poor permeability, with a Pe values less than 1 × 10−6 cm/s, due to the retain charged site on one terminus. Regarding the ODOL prodrugs, the trend was similar, with the bis-ODOL ester of IA (P3) and ODOL-esters of 1-MI (P10) and 4-MI (P14) demonstrating high permeability, while the monoester of IA (P6) demonstrating limited permeability. Overall, all ODOL prodrugs (P3, P6, P10, and P14) exhibited a significant enhancement in the permeability (Pe ∼ 15 × 10−6 cm/s) of both IA and 4-MI. These results align with previously reported ODOL-based prodrugs, such as azilsartan medoxomil, faropenem medoxomil and 2-PMPA-tetraODOL, which demonstrated significantly enhanced oral bioavailability.22,28,29 However, in the case of itaconate and analogs ODOL promoiety did not outperform POM and POC based prodrugs in enhancing permeability attributed to the highly polar surface area of the ODOL promoiety. Consequently, ODOL prodrugs were not further advanced into pharmacokinetic studies.
Interestingly, all HDP prodrugs displayed poor permeability (Pe < 1 × 10−6 cm/s) except for the IA monoester HDP prodrug (P7) which had a slightly better, but comparably lower permeability, (Pe = 3.21 × 10−6 cm/s) to POM and POC analogs. This poor permeability was attributed to their exceptionally high lipophilicity, with c log P values exceeding 5 for P7, P11, and P15 (c log P: 8.38, 8.79, and 8.91, respectively). Such high c log P values are also associated with poor solubility, reflecting an unfavorable hydrophilic–lipophilic balance and resulting in suboptimal properties.30 These findings suggest that, overall, HDP prodrugs were not effective candidates for improving the oral bioavailability of IA analogs.
To evaluate the potential for oral delivery of active compounds to systemic circulation in inflammatory conditions and to ensure skin targeting in the in vivo PK study, we focused on mouse and human plasma and skin homogenates. These models were selected because they closely mimic the enzymatic and metabolic environments of the in vivo matrices under investigation. However, as most of these compounds are ester-based prodrugs, we anticipate that other highly metabolic organs, such as the liver, may also play a significant role in their metabolic pathways, although this was not tested in the current study.
All POC, POM and ODOL prodrugs except the monoesters of IA (P4 and P5) were found to hydrolyze in mouse and human plasma readily. However, the monoesters of IA were chemically unstable, and showed poor permeability (Table 1) and therefore were not selected for further consideration. HDP prodrugs, interestingly, showed stability in both mouse and human plasma. However, when incubated in mouse skin homogenate, the HDP-based prodrugs were unstable (<50% remaining at 1 h). In human skin homogenate, POM and POC-based prodrugs demonstrated instability similar to mouse skin homogenate. Interestingly, ODOL-based prodrugs, except the bis-ODOL prodrug (P3), had moderate stability (>50% remaining after 1h) while HDP-based prodrugs were completely stable in human skin homogenates (>90% remaining at 1 h).
Collectively, POM (P1, P8 and P12) and POC (P2, P9 and P13) based prodrugs showed the most promise, with enhanced PAMPA permeability and effective release of active moieties in plasma and skin. While the POM-based prodrugs showed comparable stability and permeability results, the well-known additional toxicity attributed to POM promoieties due to carnitine depletion31 led us to advance the three POC-based prodrugs P2 (IA), P9 (1-MI) and P13 (4-MI) for in vitro gene expression efficacy studies, and in vivo mouse pharmacokinetic evaluation as described below.
Immunomodulatory Effects of P2, P9 and P13 in Stimulated Human Epidermal Keratinocytes.
We theorized that itaconate-based cell permeable prodrugs may offer benefits for alopecia areata (as we have previously shown) by counteracting immune stimulants such as Poly(I:C) and IFNγ as these stimulants have shown to contribute to the disease by activating the NLRP3 inflammasome activity32,33 and JAK-STAT signaling.34–37 IA and its derivatives have been shown to suppress proinflammatory cytokines such as IL-6, a JAK-STAT activator, and IL-1β, which disrupts hair cycling and is elevated in alopecia areata scalp lesions.38–41 Thus, to validate the therapeutic potential of P2, P9 and P13, we evaluated their dose-dependent cytotoxicity and ability to block Poly(I:C) and IFNγ-induced cytokines and chemokines release in vitro in normal human epidermal keratinocytes (NHEKs) as previously described (Figure 1).16 First, the in vitro cytotoxicity of the prodrugs was assessed to ensure they did not adversely affect cell viability. After 8 h of treatment, NHEKs treated with P2, P9, and P13 all demonstrated good cell viability, with over 80% of cells remaining viable at the highest concentration of 100 μM (data not shown). The immunomodulatory effects were next evaluated by adding P2, P9 and P13 to NHEKs stimulated with Poly(I:C) or IFNγ to induce IL-6, IL-1β, IFNβ, and IFN-inducible chemokines (CXCL9, CXCL10, CXCL11), which are recognized as characteristic markers of alopecia areata.42 For IFNγ-inducible chemokines (CXCL9, CXCL10, and CXCL11), P2 and P13 showed significant effects, with over 50% inhibition at 30–100 μM. However, P9 exhibited limited inhibitory effects, with less than 50% inhibition at 100 μM. This suggests that although the IA- and 4-MI-releasing prodrugs were effective, the 1-MI-releasing prodrug was less so, indicating that the free carboxylate at the 1-position on IA is essential for activity, and masking it resulted in a decrease in effectiveness. Similarly, IL-1β although showed low induction (2–3 fold) when stimulated with Poly(I:C) + IFNγ, P2 and P13 demonstrated over 50% inhibition of this signal at concentrations ≥10 μM, and P9 showed effects at 100 μM. P2 and P13 also showed significant, dose-dependent inhibition of IL-6 and P2 in addition showed dose dependent inhibition of IFNβ, while P9 did not, further suggesting that IA or 4-MI are more active versus 1-MI in this assay.
Figure 1.
Effect of P2, P9 and P13 on gene expression in Poly(I:C) + IFNγ stimulated NHEK cells. Induction of the chemokine and cytokine genes in primary human keratinocyte cells was measured at 8 h following incubation of P2, P9 and P13 (1–100 μM) as follows: CXCL 9 (A), CXCL 10 (B), CXCL 11 (C), IFNβ (D), IL-1β (E), and IL-6 (F). Poly(I:C) (50 μg/mL) + IFNγ (5 ng/mL) was used as the stimulant. Data expressed as mean ± SEM, n = 3. Statistical analysis was performed using one-way ANOVA by Dunnett’s multiple comparisons test with comparing with Poly(I:C) + IFNγ alone; ***p < 0.0005, **p < 0.005; *p < 0.05.
Overall, among POC-based prodrugs, P2 and P13 showed significantly enhanced immunomodulatory effects in NHEKs, supporting our hypothesis that cell permeable prodrugs may offer benefits for alopecia areata. This was further evidenced in our screening data in NHEKs, where neither 4-MI nor 1A affected the mRNA expression of TLR3 (Toll-like receptor 3) and IL-6 involved in inflammatory response while the prodrugs P2 and P13 caused >90% inhibition (Figure S1).
Pharmacokinetic Studies in Mice.
Given their promising immunomodulatory effects in NHEKs, we next conducted pharmacokinetic evaluation of P2 and P13 at a dose of 100 mg/kg equivalent in mice. The dose was selected based on previously reported dose of 25–150 mg/kg43–45 and to achieve effective concentrations (30–100 μM) in in vitro NHEK model.
The concentration–time profiles of released IA from P2 and released 4-MI from P13 in plasma and skin are presented in Figure 2. Following oral administration, intact P2 was not detectable in the plasma, suggesting rapid hydrolysis of P2 into IA. The Cmax of released IA in plasma following P2 administration (100 mg/kg IA equivalent) was 83.8 ± 18.8 μM, observed at 15 min postdose (Tmax), as shown in Figure 2C. The Cmax of IA in skin was 65.9 ± 10.2 nmol/g. Notably, P2 showed significant distribution to skin tissue, indicated by higher skin versus plasma AUC0−t values (173 ± 30.0 nmol·h/g and 108 ± 2.68 μM·h, respectively), resulting in a skin/plasma ratio of 1.61 ± 0.29. The half-life of itaconate released from P2 was calculated to be 1.42 ± 0.24 h in plasma and 3.32 ± 1.23 h in skin, demonstrating longer retention and slower clearance from the skin compared to plasma.
Figure 2.
Delivery of IA or 4-MI in plasma and skin of mice dosed with P2 and P13, respectively. (A), release of IA in mice dosed with P2 (B), release of 4-MI in mice dosed with P13, and (C), PK parameters of IA released from P2, and 4-MI released from P13 with data expressed as means ± SEM (n = 3 per time point).
Similarly, following oral administration of P13 (100 mg/kg eq of 4-MI), intact P13 was undetectable in plasma, indicating that P13 was rapidly hydrolyzed into 4-MI. The released 4-MI showed high exposure in plasma, with a Cmax of 349 ± 39.3 μM and an AUC0−t of 415 ± 16.8 μM·h. 4-MI released from P13 also exhibited high exposure in skin tissue, with an AUC0−t value of 234 ± 1.30 nmol·h/g. The skin/plasma ratio of 4-MI after P13 administration was calculated to be 0.59 ± 0.01. The half-life of 4-MI released from P13 was calculated to be 0.87 ± 0.09 h in plasma and 0.97 ± 0.20 h in skin. Overall, its exposure in the skin exceeded 100 μM, a concentration that resulted in over 50% inhibition of cytokines in NHEKs (Figure 1).
Collectively, P2 and P13 demonstrated efficient delivery of active moieties to systemic circulation and skin. While comparative PK data for IA and 4-MI versus their prodrugs are lacking in this study, our results clearly support the prodrug strategy. IA and 4-MI showed poor permeability in the PAMPA assay (Table 1), failed to inhibit cytokine production in NHEKs (Figure S1), and 4-MI was ineffective in vivo in the alopecia areata model, unlike P13 when compared at equimolar doses.16 These findings highlight the limitations of the parent compounds and underscore the potential of prodrugs like P2 and P13 to overcome these challenges, aligning with prior literature on the importance of enhancing cellular permeability for itaconate derivatives.13,14,46,47
Another limitation of this study is the absence of direct comparisons between our prodrugs and traditional treatments for alopecia areata, such as JAK inhibitors. Although future studies could address this gap, in our previous work, topical application of 5% P13 using an alternate-day, two-dose regimen significantly promoted hair growth compared to 5% tofacitinib under the same conditions16 Moreover, while JAK inhibitors like tofacitinib are effective, they carry risks of serious adverse effects, including venous thromboembolism and other on-target toxicities.48,49 In contrast, itaconate prodrugs act through a distinct mechanism, reducing the likelihood of such on-target side effects. In support, we assessed P13’s preliminary toxicity profile (given its robust efficacy in alopecia areata) at two doses via systemic administration. No hematological changes or weight loss were observed (Figure S2 and Table S1), supporting the safety of P13. These findings suggest that itaconate prodrugs offer a promising, potentially safer alternative for treating alopecia areata.
CONCLUSIONS
In summary, we synthesized four sets of IA, 4-MI, and 1-MI prodrugs using the FDA-approved promoieties POM, POC, ODOL, and HDP by incorporating itaconate diester, itaconate monoester, and itaconate diesters derived from monomethyl itaconate. These prodrugs were evaluated in a panel of in vitro and pharmacokinetic assays. The POC-based prodrugs of IA (P2) and 4-MI (P13) demonstrated the best properties including chemical stability in gastric conditions, high permeability, release of the active moiety in skin homogenates, and positive immunomodulatory effects in NHEK assays. P2 and P13 also exhibited good pharmacokinetic properties following oral administration. The ability of these prodrugs to effectively deliver the active IA and 4-MI to skin tissue following oral administration, suggests that they hold potential as oral treatments for alopecia areata and other inflammatory skin diseases.
EXPERIMENTAL SECTION
Chemicals and Materials.
A commercially available reagent or HPLC-grade solvents and materials were used for the synthesis of the compounds described. All chemicals were reagent grade purchased from Sigma-Aldrich, Combi-Blocks, or Tokyo Chemical Industry (TCI) Co. Ltd. TLC was performed on Silica gel 60 F254-coated aluminum sheets (Merck) and spots were visualized with UV light and by the solution of Ce(SO4)2·4H2O (1%) and H3P(Mo3O10)4 (2%) in sulfuric acid (10%). Column chromatography was performed on silica gel 60 (0.063–0.200 mm, Fluorochem). NMR spectra were measured on Bruker AVANCE 400 instrument. 1H NMR was recorded at 401 MHz, and signals of TMS (δ 0.0, CDCl3) and CDCl3 (δ 7.26) were used for standardization. 13C NMR spectra were recorded at 101 MHz, and the signal of CDCl3 (δ 77.16) was used for standardization. The chemical shifts are given in δ scale; the coupling constants J are given in Hz. The low-resolution ESI mass spectra were recorded using a ZQ micromass mass spectrometer (Waters). High-resolution ESI mass spectra were recorded using an LTQ Orbitrap XL spectrometer (Thermo Fisher Scientific). The purity of all compounds subjected to biological testing was over 95%.
General Synthetic Procedure for Prodrugs P1–3.
IA (200 mg, 1.53 mmol, 1 equiv), appropriate chloride 2–4 (2.5–3 equiv), sodium iodide (46.1 mg, 0.307 mmol, 0.2 equiv), and potassium carbonate (3–4 equiv) were dissolved in anhydrous MeCN (5–10 mL) and the mixture was stirred for 16 h at 45–55 °C under inert atmosphere. EtOAc (60 mL) was added, and the mixture was washed with brine (20 mL) and sat. Sodium thiosulfate (10 mL). The organic phase was dried over anhydrous Na2SO4, volatiles were evaporated, and the residue was purified by flash column chromatography on silica (various mobile phases) to afford compounds P1–3.
Bis((pivaloyloxy)methyl) 2-Methylenesuccinate (P1).
Chloromethyl pivalate (2) (579 mg, 554 μL, 3.84 mmol, 2.5 equiv); potassium carbonate (637 mg, 4.61 mmol, 3 equiv); MeCN (5 mL); 45 °C; mobile phase: cyclohexane/EtOAc, 5:1. Compound P1 was isolated as a colorless oil (199 mg) in 36% yield. 1H NMR (401 MHz, CDCl3): δH 1.20 (s, 18H), 3.38 (d, J = 1.0 Hz, 2H), 5.74 (s, 2H), 5.82 (s, 3H), 6.41 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 26.83, 26.84, 37.12, 38.75, 38.78, 79.70, 79.82, 130.52, 132.33, 164.45, 169.12, 177.07. ESI MS: 381.2 ([M + Na]+). HRMS (ESI): calcd. For C17H26O8Na 381.15199, found: 381.15158.
Bis(((isopropoxycarbonyl)oxy)methyl) 2-Methylenesuccinate (P2).
Chloromethyl isopropyl carbonate (3) (586 mg, 514 μL, 3.84 mmol, 2.5 equiv); potassium carbonate (637 mg, 4.61 mmol, 3 equiv); MeCN (5 mL); 45 °C; mobile phase: cyclohexane/EtOAc, 5:1. Compound P2 was isolated as a colorless oil (79.2 mg) in 14% yield. 1H NMR (401 MHz, CDCl3): δH 1.30 (s, 6H), 1.31 (s, 6H), 3.41 (s, 2H), 4.82–4.98 (m, 2H), 5.75 (s, 2H), 5.82 (s, 2H), 5.85 (s, 1H), 6.46 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 21.63, 36.95, 73.13, 82.00, 82.22, 131.05, 131.99, 153.26, 153.29, 164.28, 168.95. ESI MS: 385.1 ([M + Na]+). HRMS (ESI): calcd. For C15H22O10Na 385.11052, found: 385.11069.
Bis((5-methyl-2-oxo-1,3-dioxol-4-yl)methyl) 2-Methylenesuccinate (P3).
(4-Chloromethyl)-5-methyl-1,3-dioxol-2-one (4) (685 mg, 504 μL, 4.61 mmol, 3 equiv); potassium carbonate (850 mg, 6.15 mmol, 4 equiv); MeCN (10 mL); 55 °C; mobile phase: cyclohexane/EtOAc, 1:1. Compound P3 was isolated as a colorless oil (311 mg) in 57% yield. 1H NMR (401 MHz, CDCl3): δH 2.17 (s, 3H), 2.19 (s, 3H), 3.38 (d, J = 1.1 Hz, 2H), 4.86 (s, 2H), 4.92 (s, 2H), 5.81 (s, 1H), 6.40 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 9.48, 9.52, 37.57, 54.39, 54.50, 130.62, 132.59, 133.32, 133.34, 140.45, 140.49, 152.22, 165.41, 170.05. ESI MS: 279.0 ([M + Na]+). HRMS (ESI): calcd. For C11H12O7Na 279.04752, found: 279.04800.
General Synthetic Procedure for Compounds 6–8.
3-(tert-Butoxycarbonyl)but-3-enoic acid (5) (400 mg, 2.15 mmol, 1 equiv), appropriate chloride 2–4 (1.2–1.3 equiv), sodium iodide (64.4 mg, 0.430 mmol, 0.2 equiv) and potassium carbonate (445 mg, 3.22 mmol, 1.5 equiv) were dissolved in anhydrous MeCN (5 mL) and the mixture was stirred for 16 h at 45–55 °C. EtOAc (60 mL) was added, and the mixture was washed with brine (20 mL) and sat. Sodium thiosulfate (10 mL). The organic phase was dried over anhydrous Na2SO4, volatiles were evaporated, and the residue was purified by flash column chromatography on silica (various mobile phases) to afford desired compounds 6–8 as colorless oils.
1-(tert-Butyl) 4-((Pivaloyloxy)methyl) 2-Methylenesuccinate (6).
Chloromethyl pivalate (2) (421 mg, 402 μL, 2.79 mmol, 1.3 equiv); 45 °C; mobile phase: cyclohexane/EtOAc, 6:1. Compound 6 was isolated as a colorless oil (458 mg) in 71% yield. 1H NMR (401 MHz, CDCl3): δH 1.21 (s, 9H), 1.48 (s, 9H), 3.33 (d, J = 1.2 Hz, 2H), 5.63 (d, J = 1.2 Hz, 1H), 5.76 (s, 2H), 6.25 (d, J = 1.0 Hz, 1H). 13C NMR (101 MHz, CDCl3): δC 27.00, 28.10, 37.72, 38.90, 79.88, 81.44, 127.92, 134.86, 165.13, 169.79, 177.23. ESI MS: 323.2 ([M + Na]+). HRMS (ESI): calcd. For C15H24O6Na 323.14651, found: 323.14622.
4-(tert-Butyl) 1-(((Isopropoxycarbonyl)oxy)methyl) 2-Methylene-succinate (7).
Chloromethyl isopropyl carbonate (3) (393 mg, 345 μL, 2.58 mmol, 1.2 equiv); 50 °C; mobile phase: cyclohexane/EtOAc, 5:1. Compound 7 was isolated as a colorless oil (552 mg) in 85% yield. 1H NMR (401 MHz, CDCl3): δH 6.28–6.23 (m, 1H), 5.74 (s, 2H), 5.62 (td, J = 1.2 Hz, 1H), 4.95–4.84 (m, 1H), 3.36–3.31 (m, 2H), 1.46 (s, 9H), 1.29 (d, J = 6.2 Hz, 6H). 13C NMR (101 MHz, CDCl3): δC 169.6, 165.0, 153.4, 134.6, 128.2, 82.0, 81.5, 73.1, 37.7, 28.0, 21.7. ESI MS: 325.1 ([M + Na]+). HRMS (ESI): calcd. for C14H22O7Na 325.12577; found: 325.12580.
4-(tert-Butyl) 1-(((Isopropoxycarbonyl)oxy)methyl) 2-Methylene-succinate (8).
(4-Chloromethyl)-5-methyl-1,3-dioxol-2-one (4) (383 mg, 282 μL, 2.58 mmol, 1.2 equiv); 55 °C; mobile phase: cyclohexane/EtOAc, 4:1. Compound 8 was isolated as a colorless oil (493 mg) in 77% yield. 1H NMR (401 MHz, CDCl3): δH 1.46 (s, 9H), 2.16 (s, 3H), 3.31 (d, J = 0.8 Hz, 2H), 4.84 (s, 2H), 5.63 (td, J = 1.1 Hz, 1H), 6.23–6.25 (m, 1H). 13C NMR (101 MHz, CDCl3): δC 9.49, 28.05, 37.83, 54.15, 81.48, 128.05, 133.53, 134.91, 140.30, 152.15, 165.15, 170.54. ESI MS: 377.0 ([M + Na]+). HRMS (ESI): calcd. for C15H14O10Na 377.04792; found: 377.04773.
General Synthetic Procedure for Prodrugs P4–6.
Compounds P6–8 (1 equiv) were dissolved in anhydrous DCM (0.5–1 mL), trifluoroacetic acid (4–6 mL) was added, and the mixture was stirred for 2 h at room temperature. Volatiles were evaporated, and the residue was dissolved in DCM (3 × 15 mL) and evaporated 3 times. The residue was purified by flash column chromatography on silica (mobile phase: cyclohexane/EtOAc, 1:1) to afford desired prodrugs P4–6.
2-Methylene-4-oxo-4-((pivaloyloxy)methoxy)butanoic Acid (P4).
Compound 6 (140 mg, 0.466 mmol); DCM (0.5 mL), trifluoroacetic acid (4 mL). Prodrug P4 was isolated as a colorless oil (110 mg) in 97% yield. 1H NMR (401 MHz, CDCl3): δH 1.21 (s, 9H), 3.37 (s, 2H), 5.77 (s, 2H), 5.86 (s, 1H), 6.49 (s, 1H), 11.12 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 26.81, 36.97, 38.76, 79.66, 131.36, 132.54, 169.22, 171.11, 177.13. ESI MS: 267.1 ([M + Na]+). HRMS (ESI): calcd. For C11H16O6Na 267.08391; found: 267.08375.
4-(((Isopropoxycarbonyl)oxy)methoxy)-2-methylene-4-oxobutanoic Acid (P5).
Compound 7 (500 mg, 1.65 mmol); DCM (1 mL), trifluoroacetic acid (6 mL). Prodrug P5 was isolated as a colorless solid (375 mg) in 92% yield. 1H NMR (401 MHz, CDCl3): δH 1.30 (s, 3H), 1.32 (s, 3H), 3.40 (s, 2H), 4.84–4.99 (m, 1H), 5.76 (s, 2H), 5.88 (d, J = 1.1 Hz, 1H), 6.50 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 21.75, 36.94, 73.31, 82.14, 131.72, 132.49, 153.44, 169.22, 171.37. ESI MS: 245.1 ([M – H]+). HRMS (ESI): calcd. for C10H13O7 245.06668; found: 245.06667.
4-((5-Methyl-2-oxo-1,3-dioxol-4-yl)methoxy)-2-methylene-4-oxobutanoic Acid (P6).
Compound 8 (470 mg, 1.58 mmol); DCM (1 mL), trifluoroacetic acid (6 mL). Prodrug P6 was isolated as a colorless solid (321 mg) in 84% yield. 1H NMR (401 MHz, CDCl3): δH 2.19 (s, 3H), 3.40 (d, J = 1.1 Hz, 2H), 4.89 (s, 2H), 5.89 (d, J = 1.0 Hz, 1H), 6.52 (s, 1H), 11.46 (bs, 1H). 13C NMR (101 MHz, CDCl3): δC 9.47, 37.17, 54.34, 131.73, 132.72, 133.43, 140.43, 152.21, 170.20, 171.42. ESI MS: 265.0 ([M + Na]+). HRMS (ESI): calcd. for C10H10O7Na 265.03187; found: 265.03191.
Synthetic Procedure for Prodrug P7.
4-(3-(Hexadecyloxy)-propoxy)-2-methylene-4-oxobutanoic Acid (P7).
Itaconic anhydride (336 mg, 3.00 mmol, 1 equiv) and 3-(hexadecyloxy)propanol (992 mg, 3.30 mmol, 1.1 equiv) were dissolved in CHCl3 (3 mL) and the mixture was stirred for 16 h at 70 °C. Volatiles were evaporated, and the residue was purified by flash column chromatography on silica (mobile phase: DCM/MeOH, 35:1) to afford 1.03 g (83%) of prodrug P7 as a colorless solid. 1H NMR (401 MHz, CDCl3): δH 0.88 (t, 3H, J = 6.8, 3H), 1.20–1.35 (m, 26 H), 1.60–1.51 (m, 2H), 1.89 (p, 2H), 3.34 (d, J = 1.0, 2H), 3.39 (t, J = 6.7, 2H), 3.46 (t, J = 6.2, 2H), 4.20 (t, J = 6.4, 2H), 5.83 (d, J = 1.1, 1H), 6.46 (d, J = 0.9, 1H). 13C NMR (101 MHz, CDCl3): δC 14.28, 22.85, 26.31, 29.12, 29.52, 29.67, 29.77, 29.80, 29.82, 29.84, 29.86, 32.08, 37.47, 62.54, 67.16, 71.34, 130.79, 133.38, 170.62, 170.93. ESI MS: 411.3 ([M – H]+). HRMS (ESI): calcd. for C24H43O5 411.31160; found: 411.31088.
General Synthetic Procedure for Prodrugs P8–10 and P11–14.
1-MI (11) or 4-MI (12) (200 mg, 1.39 mmol, 1 equiv), appropriate chloride 2–4 (1.2–1.3 equiv), sodium iodide (41.6 mg, 0.278 mmol, 0.2 equiv) and potassium carbonate (288 mg, 2.08 mmol, 1.5 equiv) were dissolved in anhydrous MeCN (5 mL) and the mixture was stirred for 16 h at 40–45 °C. EtOAc (60 mL) was added, and the mixture was washed with brine (20 mL) and sat. sodium thiosulfate (10 mL). The organic phase was dried over anhydrous Na2SO4, volatiles were evaporated, and the residue was purified by flash column chromatography on silica (cyclohexane/EtOAc, 5:1) to afford desired prodrugs P8–10 and P11–14.
1-Methyl 4-((Pivaloyloxy)methyl) 2-Methylenesuccinate (P8).
1-MI (11); chloromethyl pivalate (272 mg, 260 μL, 1.80 mmol, 1.3 equiv); 40 °C. Compound P8 was isolated as a colorless oil (301 mg) in 84% yield. 1H NMR (401 MHz, CDCl3): δH 1.22 (s, 9H), 3.38 (s, 2H), 3.77 (s, 3H), 5.74 (dd, J = 2.1, 1.1 Hz, 1H), 5.76 (s, 2H), 6.35 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 26.95, 37.52, 38.86, 52.29, 79.75, 129.07, 133.15, 166.43, 169.51, 177.17. ESI MS: 281.1 ([M + Na]+). HRMS (ESI): calcd. for C12H18O6Na 281.09956; found: 281.09993.
4-(((Isopropoxycarbonyl)oxy)methyl) 1-Methyl 2-Methylenesuccinate (P9).
1-MI (11); chloromethyl isopropyl carbonate (254 mg, 223 μL, 1.67 mmol, 1.2 equiv); 40 °C. Compound P9 was isolated as a colorless oil (336 mg) in 93% yield. 1H NMR (401 MHz, CDCl3): δH 1.31 (s, 3H), 1.32 (s, 3H), 3.40 (s, 2H), 3.76 (s, 3H), 4.86−4.97 (m, 1H), 5.78−5.71 (m, 3H), 6.36 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 21.65, 37.33, 52.20, 73.12, 81.96, 129.18, 132.88, 153.31, 166.33, 169.27. ESI MS: 283.1 ([M + Na]+). HRMS (ESI): calcd. for C11H16O7Na 283.07882; found: 283.07925.
1-Methyl 4-((5-Methyl-2-oxo-1,3-dioxol-4-yl)methyl) 2-Methylenesuccinate (P10).
1-MI (11); (4-chloromethyl)-5-methyl-1,3-dioxol-2-one (268 mg, 197 μL, 1.80 mmol, 1.3 equiv); 40 °C. Compound P10 was isolated as a colorless oil (298 mg) in 84% yield. 1H NMR (401 MHz, CDCl3): δH 2.16 (s, 3H), 3.36 (s, 2H), 3.76 (s, 3H), 4.85 (s, 2H), 5.74 (d, J = 1.1 Hz, 1H), 6.35 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 9.49, 37.60, 52.35, 54.25, 129.25, 133.20, 133.43, 140.34, 152.19, 166.50, 170.33. ESI MS: 279.0 ([M + Na]+). HRMS (ESI): calcd. for C11H12O7Na 279.04752; found: 279.04800.
4-Methyl 1-((Pivaloyloxy)methyl) 2-Methylenesuccinate (P12).
4-MI (12); chloromethyl pivalate (272 mg, 260 μL, 1.80 mmol, 1.3 equiv); 40 °C. Compound P12 was isolated as a colorless oil (330 mg) in 92% yield. 1H NMR (401 MHz, CDCl3): δH 1.20 (s, 9H), 3.34 (s, 2H), 3.68 (s, 3H), 5.79 (d, J = 1.1 Hz, 1H), 5.82 (s, 2H), 6.39 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 26.95, 37.38, 38.90, 52.21, 79.91, 130.30, 133.06, 164.82, 170.90, 177.19. ESI MS: 281.1 ([M + Na]+). HRMS (ESI): calcd. for C12H18O6Na 281.09956; found: 281.09921.
1-(((Isopropoxycarbonyl)oxy)methyl) 4-Methyl 2-Methylenesuccinate (P13).
4-MI (12); chloromethyl isopropyl carbonate (254 mg, 223 μL, 1.67 mmol, 1.2 equiv); 45 °C. Compound P13 was isolated as a colorless oil (340 mg) in a 94% yield. 1H NMR (401 MHz, CDCl3): δH 1.33 (s, 3H), 1.34 (s, 3H), 3.38 (s, 2H), 3.72 (s, 3H), 4.86−5.02 (m, 1H), 5.82−5.88 (m, 3H), 6.46 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 21.64, 37.20, 52.12, 73.10, 82.20, 130.50, 132.76, 153.29, 164.52, 170.77. ESI MS: 283.1 ([M + Na]+). HRMS (ESI): calcd. for C11H16O7Na 283.07882; found: 283.07855.
4-Methyl 1-((5-Methyl-2-oxo-1,3-dioxol-4-yl)methyl) 2-Methylenesuccinate (P14).
4-MI (12); (4-chloromethyl)-5-methyl-1,3-dioxol2-one (268 mg, 197 μL, 1.80 mmol, 1.3 equiv); 40 °C. Compound P14 was isolated as a light-yellow oil (320 mg) in 90% yield. 1H NMR (401 MHz, CDCl3): δH 2.17 (s, 3H), 3.33 (s, 2H), 3.67 (s, 3H), 4.91 (s, 2H), 5.78 (d, J = 1.1 Hz, 1H), 6.35 (s, 1H). 13C NMR (101 MHz, CDCl3): δC 9.36, 37.36, 52.13, 54.28, 129.98, 132.99, 133.36, 140.30, 152.11, 165.51, 170.91. ESI MS: 279.1 ([M + Na]+). HRMS (ESI): calcd. For C11H12O7Na 279.04752; found: 279.04757.
General Synthetic Procedure for Prodrugs P11 and P15.
1-MI (11) or 4-MI (12) (400 mg, 2.78 mmol, 1 equiv) were dissolved in anhydrous DCM (8 mL), 3-(hexadecyloxy)propanol (959 mg, 3.19 mmol, 1.15 equiv) and DMAP (509 mg, 4.16 mmol, 1.5 equiv) was added and the solution was cooled to 0 °C and stirred under inert atmosphere. A solution of DCC (859 mg, 4.16 mmol, 1.5 equiv) in anhydrous DCM (5 mL) was added dropwise for 10 min, and the resulting mixture was stirred for 1 h at 0 °C and then overnight (15 h) at room temperature. The mixture was filtered, DCM (30 mL) was added, and the organic phase was washed with 10% KHSO4 (3 × 10 mL), sat. NaHCO3 (10 mL) and brine (10 mL). The organic phase was dried over anhydrous Na2SO4, volatiles were evaporated, and the residue was purified by flash column chromatography on silica (mobile phase: cyclohexane/EtOAc, 80:15) to afford prodrugs P11 and P15.
4-(3-(Hexadecyloxy)propyl) 1-Methyl 2-Methylenesuccinate (P11).
1-MI (11). Compound P11 was isolated as a colorless amorphous compound (332 mg) in 28% yield. 1H NMR (401 MHz, CDCl3): δH 0.87 (t, J = 7.0 Hz, 3H), 1.23−1.33 (m, 26 H), 1.50−1.58 (m, 2H), 1.84−1.92 (p, 2H), 3.33 (d, J = 1.2 Hz, 2H), 3.38 (t, J = 6.7 Hz, 2H), 3.45 (t, J = 6.3 Hz, 2H), 3.76 (s, 3H), 4.19 (t, J = 6.5 Hz, 2H), 5.69−5.71 (m, 1H), 6.32 (d, J = 1.1 Hz, 1H). 13C NMR (101 MHz, CDCl3): δC 14.27, 22.84, 26.30, 29.14, 29.51, 29.66, 29.76, 29.78, 29.80, 29.82, 29.84, 29.86, 32.07, 37.89, 52.26, 62.43, 67.16, 71.32, 128.59, 128.73, 133.88, 166.79, 170.79. ESI MS: 449.3 ([M + Na]+). HRMS (ESI): calcd. for C25H46O5Na 449.32375; found: 449.32382.
1-(3-(Hexadecyloxy)propyl) 4-Methyl 2-Methylenesuccinate (P15).
4-MI (12). Compound P15 was isolated as a colorless amorphous compound (805 mg) in 68% yield. 1H NMR (401 MHz, CDCl3): δH 0.88 (t, J = 7.0 Hz, 3H), 1.21−1.34 (m, 26 H), 1.50−1.60 (m, 2H), 1.89−1.97 (p, 2H), 3.34 (d, J = 1.2 Hz, 2H), 3.39 (t, J = 6.7 Hz, 2H), 3.48 (t, J = 6.3 Hz, 2H), 3.70 (s, 3H), 4.26 (t, J = 6.5 Hz, 2H), 5.69−5.71 (m, 1H), 6.31−6.35 (m, 1H). 13C NMR (101 MHz, CDCl3): δC 14.26, 22.84, 26.32, 29.22, 29.51, 29.66, 29.76, 29.78, 29.81, 29.82, 29.85, 29.87, 32.08, 37.69, 52.17, 62.58, 67.23, 71.36, 128.53, 134.04, 166.20, 171.27. ESI MS: 427.3 ([M + H]+). HRMS (ESI): calcd. for C25H47O5 427.34180; found: 427.34158.
CHEMICAL AND METABOLIC STABILITY
For measuring chemical stability, prodrugs were spiked (10 μM) in pH buffers (pH 1.2, 4.5, and 7.4) in triplicate. These mixtures were incubated at 37 °C for 1 h. Prodrug disappearance was monitored using the developed LC–MS/MS methods described below.
For measuring metabolic stability, prodrugs (10 μM) were spiked in mouse and human plasma and incubated in an orbital shaker at 37 °C. Aliquots (100 μL) were sampled at predetermined times (0 and 60 min) and quenched with cold acetonitrile (300 μL) containing internal standard (losartan 0.5 μM). The samples were vortexed for 30 s and centrifuged at 14,000g for 10 min. The supernatant (50 μL) was diluted with water (50 μL) and transferred to a 250 μL polypropylene vial sealed with a Teflon cap. These samples were run using liquid chromatography and high-resolution mass spectrometry (LCMS), and the disappearance of prodrugs was noted over the stipulated time. Briefly, prodrugs were analyzed on a Thermo Scientific Dionex Ultimate 3000 UPLC system coupled to Dionex Ultimate 3000 pump and autosampler using EclipsePlus C18 UPLC column from Agilent [Santa Clara, CA] (100 mm × 2.1 mm id, 1.8 μm). The autosampler was maintained at 4 °C and the column compartment at 35 °C for the duration of the LC-MS runs. Chromatographic separation was achieved using acetonitrile/water containing 0.1% formic acid as a mobile phase while pumping a flow of 0.3 mL/min for 9 min using gradient elution. The eluent was analyzed using a Thermo Scientific Q Exactive Focus mass spectrometer, equipped with a heated electrospray ionization (HESI) probe set in the positive ionization mode. Samples were introduced into the ionization source through a heated nebulized probe (350 °C). Disappearance of prodrugs was measured from the ratio of peak areas of analyte to IS.
PAMPA-GIT.
To assess intestinal permeability, we utilized STIRWELL PAMPA sandwiches (pION INC). A 5 μL of GIT-0 lipid solution was applied to coat each well of the top (acceptor) compartment of STIRWELL PAMPA sandwich. Before assembling the sandwich, the bottom (donor) plate was loaded with 200 μL of test compounds and reference compounds (10 μM) dissolved in the pION buffer at pH 7.4, containing 0.2% DMSO. The acceptor plate was filled with 200 μL of pH 7.4 sink buffer. The sandwich was incubated at room temperature for 4 h. Following the incubation, 50 μL of samples were collected from both donor and acceptor compartments and analyzed by LC-MS/MS. value is calculated using the following equation.
where is the effective permeability coefficient (cm/s), A is the filter area (0.3 cm2) multiplied by a nominal porosity of 70% according to the manufacturer, and are the volumes in the donor and acceptor phase, t is the incubation time, is the steady state time (s), is the concentration (mol cm−3) of the compound in the donor phase at time , is the concentration (mol cm−3) of the compound in the donor phase at time 0, and is the membrane retention factor
NHEK Cytotoxicity.
Normal human epidermal keratinocytes (Lonza, Basel, Switzerland) were revived, allowed to expand, and seeded in 96-well plates. P2, P9 and P13 were diluted to concentrations of 100, 30, 10, and 1 μM and added to the assay plates containing NHEKs. The plates were checked for compound effects after 8 h of incubation to determine % viability of NHEKs. Cell viability was measured with CellTiter-Glo assay (Promega, Madison, WI).
Poly(I:C)/IFNγ-Induced Gene Expression Profiles in NHEKs.
NHEKs were seeded in plates and incubated overnight at 37 °C and 5% CO, followed by stimulation of Poly(I:C) (50 μg/mL) and IFNγ (5 ng/mL) with or without the presence of prodrugs at the concentrations of 1, 10, 30, and 100 μM. After incubation, RNA was isolated and analyzed with TaqMan RT-PCR (Thermo Fisher Scientific, Waltham, MA) to quantify fold changes in the expression of genes of IL-6, IFNβ, IL-1β, CXCL-9, CXCL-10, and CXCL-11.
Pharmacokinetic Studies.
All animal studies were conducted in accordance with protocols reviewed and approved by the Institutional Animal Care and Use Committee of Johns Hopkins University (JHU). C57BL6 mice weighing between 25–30 g and 8.5 weeks of age were maintained on a 12 h light-dark cycle, with access to food and water, ad libitum.
For PK studies, P2 or P13 were formulated in 10% DMSO, 80% PEG, 10% HBS v/v/v and administered PO at a dose of 100 mg/kg equivalent to either IA or MMI. All of the formulations were freshly prepared prior to the dosing. The mice were sacrificed at specified time points (0.25, 0.5, 1, 2, 4, 6, and 24 h) post drug administration. Blood samples were collected in heparinized microtubes by cardiac puncture and spun at 2000× g for 15 min to collect plasma and then 100 μL plasma was stabilized by the addition of 50 μL 2% formic acid in water and immediately frozen and stored at −80 °C.
Skin was dissected, flash frozen in liquid nitrogen, then weighed. Four microliters of 0.1% FA in 30:70 water:ACN/mg tissue was added as stabilizer. Samples were stored at −80 °C.
For quantifying intact P2 or P13, IA, and 4-MI in plasma, naive mouse plasma was combined 2:1 with 2% formic acid stabilizer, aliquoted, and standard and QC stock solutions of each analyte were spiked to obtain standards (0.01–1000 nmol/mL), and QCs (0.05–500 nmol/mL). These then were protein precipitated using methanol (5 × plasma volume) containing internal standards (0.5 μM Losartan: Millipore Sigma, Burlington MA; 10 μM itaconic acid-C[13]5, Cambridge isotope libraries, Tewksbury, MA; 10 μM 5-methoxy-2-methylene-5-oxopentanoic acid (MMOPA), Enamine; Monmouth Jct., NJ.) Samples were prepared in a similar fashion (without spiking standard stock solutions) to match the ratio of matrix to solvent achieved in standards and QCs. The solutions were centrifuged at 16,000 g for 5 min at 4 °C, and the supernatant was analyzed using LC-MS/MS to quantify P2 or P13, IA and 4-MI using the bioanalytical methods described below.
For quantifying intact P2 or P13, IA, and 4-MI in the skin, naive skin was homogenized, and protein precipitated simultaneously using methanol (5 × tissue weight) containing internal standards in a Geno grinder at 1500 rpm for 3 min. This matrix was aliquoted, and standard and QC stock solutions of each analyte were spiked to obtain standards (0.01–1000 nmol/g), and QCs (0.05–500 nmol/g). Samples were prepared in a similar fashion (without spiking standard stock solutions) to match the ratio of matrix to solvent achieved in standards and QCs. These were then centrifuged at 16,000g for 5 min at 4 °C, and the supernatant was analyzed using LC-MS/MS to quantify the analytes using the bioanalytical methods described below.
Bioanalysis.
Chromatographic analysis was performed using an Ultimate 3000 ultrahigh-performance system consisting of coupled with a QExactive Focus orbitrap mass spectrometer (Thermo Fisher Scientific Inc., Waltham, MA). Analyte separation was achieved at 35 °C using an Agilent EclipsePlus column (100 mm × 2.1 mm i.d.) packed with 1.8 μm C18 stationary phase. The mobile phase consisted of 0.1% formic acid in methanol and 0.1% formic acid in water with gradient elution. The [M + H]+ ion transition for P2 was m/z 363.1286 → 105.0592 and 113.0291 with losartan as internal standard. The [M + H]+ transition for losartan were m/z 423.1695 → 207.0915 and 377.1518. The [M + H]+ ion transition for P13 was m/z 261.0969 → 69.0339, 99.0474, and 127.0386 with losartan as internal standard as above. The [M − H]− ion transition for IA was m/z 129.0193 → 85.0294 with IA-[13C]5 as internal standard. The [M − H]− ion transition for IA-[13C]5 was m/z 134.0361 → 89.0429. The [M + H]+ ion transition for 4-MI was m/z 145.0495 → 69.0339 and 99.0474 with MMOPA as internal. standard The [M + H]+ transition for MMOPA were m/z 159.0652 → 113.0656 and 127.0388.
Supplementary Material
ACKNOWLEDGMENTS
The synthetic part of this work was supported by a grant from Ministry of Education, Youths and Sports of the Czech Republic (LM2023053, program EATRIS-CZ).
ABBREVIATIONS USED
- ACN
Acetonitrile
- DCM
dichloromethane
- DMI
dimethyl itaconate
- EtOAc
ethyl acetate
- 4-EI
4-ethyl itaconate
- HDP
3-(hexadecyloxy)propyl
- HPLC
high-performance liquid chromatography
- HRMS
high-resolution mass spectrometry
- IRG1
immune-responsive gene 1
- POC
isopropoxycarbonyloxymethyl
- IA
itaconic acid
- 1-MI
1-methyl itaconate
- 4-MI
4-methyl itaconate
- NMR
(5-methyl-2-oxo-1,3-dioxol-4-yl)methyl (ODOL);nuclear magnetic resonance
- NHEK
normal human epidermal keratinocyte
- 4-OI
4-octyl itaconate
- PAMPA
parallel artificial membrane permeability assay
- POM
pivaloyloxymethyl
- NaI
sodium iodide
- SDH
succinate dehydrogenase
- TCA
tricarboxylic acid
Footnotes
ASSOCIATED CONTENT
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.4c02646.
1H and 13C (APT) NMR spectra and LC-MS chromatogram and spectra for prodrugs P1–P15; molecular formula strings and the associated biochemical and biological data (PDF)
SMILES, permeability data, mouse plasma and skin stability, human plasma and skin stability for prodrugs P1–P15, IA, 1-MI, 4-MI, DMI, 4-OI (CSV)
Complete contact information is available at: https://pubs.acs.org/10.1021/acs.jmedchem.4c02646
The authors declare the following competing financial interest(s): L.T., I.S., M.K., L.G., P.M., B.S.S., and R.R. are inventors on Johns Hopkins University (JHU) patents covering novel compositions of itaconate prodrugs and their utility. These patents have been licensed to Sun Pharma Advanced Research Company Ltd. (SPARC). This arrangement has been reviewed and approved by the JHU in accordance with its conflict-of-interest policies. Other authors declare that no conflicts of interest exist.
Contributor Information
Chae Bin Lee, Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.
Ivan Šnajdr, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague 160 00, Czech Republic.
Lukáš Tenora, Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States; Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague 160 00, Czech Republic.
Jesse Alt, Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.
Sadakatali Gori, Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.
Marcela Krečmerová, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague 160 00, Czech Republic.
R. Michael Maragakis, Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.
James Paule, Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.
Sandhya Tiwari, In Vitro Biology, Sun Pharma Advanced Research Company Ltd., Vadodara 391775 Gujarat, India.
Jitesh Iyer, In Vitro Biology, Sun Pharma Advanced Research Company Ltd., Vadodara 391775 Gujarat, India.
Rashmi Talwar, In Vitro Biology, Sun Pharma Advanced Research Company Ltd., Vadodara 391775 Gujarat, India.
Luis Garza, Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.
Pavel Majer, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague 160 00, Czech Republic.
Barbara S. Slusher, Johns Hopkins Drug Discovery, Departments of Neurology, Psychiatry and Behavioral Sciences, Neuroscience, Medicine, Oncology, and Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
Rana Rais, Johns Hopkins Drug Discovery, Departments of Neurology, and Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.
REFERENCES
- (1).O’Neill LAJ; Artyomov MN Itaconate: the poster child of metabolic reprogramming in macrophage function. Nat. Rev. Immunol 2019, 19 (5), 273–281. [DOI] [PubMed] [Google Scholar]
- (2).Ferreira AV; Netea MG; Dominguez-Andres J. Itaconate as an immune modulator. Aging 2019, 11 (12), 3898–3899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (3).Lampropoulou V; Sergushichev A; Bambouskova M; Nair S; Vincent EE; Loginicheva E; Cervantes-Barragan L; Ma X; Huang SC; Griss T; et al. Itaconate Links Inhibition of Succinate Dehydrogenase with Macrophage Metabolic Remodeling and Regulation of Inflammation. Cell Metab. 2016, 24 (1), 158–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (4).Lawrence GW; Ovsepian SV; Wang J; Aoki KR; Dolly JO Extravesicular intraneuronal migration of internalized botulinum neurotoxins without detectable inhibition of distal neurotransmission. Biochem. J 2012, 441 (1), 443−452. [DOI] [PubMed] [Google Scholar]
- (5).Qin W; Qin K; Zhang Y; Jia W; Chen Y; Cheng B; Peng L; Chen N; Liu Y; Zhou W; et al. S-glycosylation-based cysteine profiling reveals regulation of glycolysis by itaconate. Nat. Chem. Biol 2019, 15 (10), 983–991. [DOI] [PubMed] [Google Scholar]
- (6).Song H; Xu T; Feng X; Lai Y; Yang Y; Zheng H; He X; Wei G; Liao W; Liao Y; et al. Itaconate prevents abdominal aortic aneurysm formation through inhibiting inflammation via activation of Nrf2. EBioMedicine 2020, 57, No. 102832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (7).Bambouskova M; Gorvel L; Lampropoulou V; Sergushichev A; Loginicheva E; Johnson K; Korenfeld D; Mathyer ME; Kim H; Huang LH; et al. Electrophilic properties of itaconate and derivatives regulate the IkappaBzeta-ATF3 inflammatory axis. Nature 2018, 556 (7702), 501–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (8).Swain A; Bambouskova M; Kim H; Andhey PS; Duncan D; Auclair K; Chubukov V; Simons DM; Roddy TP; Stewart KM; Artyomov MN Comparative evaluation of itaconate and its derivatives reveals divergent inflammasome and type I interferon regulation in macrophages. Nat. Metab 2020, 2 (7), 594–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (9).Hooftman A; Angiari S; Hester S; Corcoran SE; Runtsch MC; Ling C; Ruzek MC; Slivka PF; McGettrick AF; Banahan K; et al. The Immunomodulatory Metabolite Itaconate Modifies NLRP3 and Inhibits Inflammasome Activation. Cell Metab. 2020, 32 (3), 468–478.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (10).Runtsch MC; Angiari S; Hooftman A; Wadhwa R; Zhang Y; Zheng Y; Spina JS; Ruzek MC; Argiriadi MA; McGettrick AF; et al. Itaconate and itaconate derivatives target JAK1 to suppress alternative activation of macrophages. Cell Metab. 2022, 34 (3), 487–501.e8. [DOI] [PubMed] [Google Scholar]
- (11).Yang S; Zhang X; Zhang H; Lin X; Chen X; Zhang Y; Lin X; Huang L; Zhuge Q. Dimethyl itaconate inhibits LPS-induced microglia inflammation and inflammasome-mediated pyroptosis via inducing autophagy and regulating the Nrf-2/HO-1 signaling pathway. Mol. Med. Rep 2021, 24 (3), No. 672, DOI: 10.3892/mmr.2021.12311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Li W; Li Y; Kang J; Jiang H; Gong W; Chen L; Wu C; Liu M; Wu X; Zhao Y; Ren J. 4-octyl itaconate as a metabolite derivative inhibits inflammation via alkylation of STING. Cell Rep. 2023, 42 (3), No. 112145. [DOI] [PubMed] [Google Scholar]
- (13).ElAzzouny M; Tom CT; Evans CR; Olson LL; Tanga MJ; Gallagher KA; Martin BR; Burant CF Dimethyl Itaconate Is Not Metabolized into Itaconate Intracellularly. J. Biol. Chem 2017, 292 (12), 4766–4769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (14).Hooftman A; O’Neill LAJ The Immunomodulatory Potential of the Metabolite Itaconate. Trends Immunol. 2019, 40 (8), 687–698. [DOI] [PubMed] [Google Scholar]
- (15).Lin J; Ren J; Gao DS; Dai Y; Yu L. The Emerging Application of Itaconate: Promising Molecular Targets and Therapeutic Opportunities. Front. Chem 2021, 9, No. 669308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (16).Tsai J; Gori S; Alt J; Tiwari S; Iyer J; Talwar R; Hinsu D; Ahirwar K; Mohanty S; Khunt C; et al. Topical SCD-153, a 4-methyl itaconate prodrug, for the treatment of alopecia areata. PNAS Nexus 2023, 2 (1), No. pgac297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Hecker SJ; Erion MD Prodrugs of phosphates and phosphonates. J. Med. Chem 2008, 51 (8), 2328–2345. [DOI] [PubMed] [Google Scholar]
- (18).Babu KS; Reddy MS; Tagore AR; Reddy GS; Sebastian S; Varma MS; Venkateswarlu G; Bhattacharya A; Reddy PP; Anand RV Efficient synthesis of olmesartan medoxomil, an antihypertensive drug. Synth. Commun 2008, 39 (2), 291–298. [Google Scholar]
- (19).Garaga S; Misra NC; Reddy AVR; Prabahar KJ; Takshinamoorthy C; Sanasi PD; Babu KR Commercial synthesis of Azilsartan Kamedoxomil: An angiotensin II receptor blocker. Org. Process Res. Dev 2015, 19 (4), 514–519. [Google Scholar]
- (20).Hostetler KY Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: current state of the art. Antiviral Res. 2009, 82 (2), A84–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Pradere U; Garnier-Amblard EC; Coats SJ; Amblard F; Schinazi RF Synthesis of nucleoside phosphate and phosphonate prodrugs. Chem. Rev 2014, 114 (18), 9154–9218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Dash RP; Tichy T; Veeravalli V; Lam J; Alt J; Wu Y; Tenora L; Majer P; Slusher BS; Rais R. Enhanced Oral Bioavailability of 2-(Phosphonomethyl)-pentanedioic Acid (2-PMPA) from its (5-Methyl-2-oxo-1,3-dioxol-4-yl)methyl (ODOL)-Based Prodrugs. Mol. Pharmaceutics 2019, 16 (10), 4292–4301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Majer P; Jancarik A; Krecmerova M; Tichy T; Tenora L; Wozniak K; Wu Y; Pommier E; Ferraris D; Rais R; Slusher BS Discovery of Orally Available Prodrugs of the Glutamate Carboxypeptidase II (GCPII) Inhibitor 2-Phosphonomethylpentanedioic Acid (2-PMPA). J. Med. Chem 2016, 59 (6), 2810–2819. [DOI] [PubMed] [Google Scholar]
- (24).Chollet AM; Le Diguarher T; Kucharczyk N; Loynel A; Bertrand M; Tucker G; Guilbaud N; Burbridge M; Pastoureau P; Fradin A; et al. Solid-phase synthesis of alpha-substituted 3-bisarylthio N-hydroxy propionamides as specific MMP inhibitors. Bioorg. Med. Chem 2002, 10 (3), 531–544. [DOI] [PubMed] [Google Scholar]
- (25).Faller B. Artificial membrane assays to assess permeability. Curr. Drug Metab 2008, 9 (9), 886–892. [DOI] [PubMed] [Google Scholar]
- (26).Daina A; Michielin O; Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep 2017, 7, No. 42717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Wils P; Warnery A; Phung-Ba V; Legrain S; Scherman D. High lipophilicity decreases drug transport across intestinal epithelial cells. J. Pharmacol. Exp. Ther 1994, 269 (2), 654–658. [PubMed] [Google Scholar]
- (28).Kawaguchi N; Ebihara T; Takeuchi T; Morohashi A; Yamasaki H; Tagawa Y; Takahashi J; Kondo T; Asahi S. Absorption of TAK-491, a new angiotensin II receptor antagonist, in animals. Xenobiotica 2013, 43 (2), 182–192. [DOI] [PubMed] [Google Scholar]
- (29).Schurek KN; Wiebe R; Karlowsky JA; Rubinstein E; Hoban DJ; Zhanel GG Faropenem: review of a new oral penem. Expert Rev. Anti-Infect. Ther 2007, 5 (2), 185–198. [DOI] [PubMed] [Google Scholar]
- (30).Tsaioun K; Blaauboer BJ; Hartung T. Evidence-based absorption, distribution, metabolism, excretion (ADME) and its interplay with alternative toxicity methods. ALTEX 2016, 33 (4), 343–358. [DOI] [PubMed] [Google Scholar]
- (31).Heidel KM; Dowd CS Phosphonate prodrugs: an overview and recent advances. Future Med. Chem 2019, 11 (13), 1625–1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (32).Shin JM; Choi DK; Sohn KC; Kim SY; Min Ha J; Ho Lee Y; Im M; Seo YJ; Deok Kim C; Lee JH; et al. Double-stranded RNA induces inflammation via the NF-kappaB pathway and inflammasome activation in the outer root sheath cells of hair follicles. Sci. Rep 2017, 7, No. 44127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (33).Shin JM; Choi DK; Sohn KC; Koh JW; Lee YH; Seo YJ; Kim CD; Lee JH; Lee Y. Induction of alopecia areata in C3H/HeJ mice using polyinosinic-polycytidylic acid (poly[I:C]) and interferon-gamma. Sci. Rep 2018, 8 (1), No. 12518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (34).Xing L; Dai Z; Jabbari A; Cerise JE; Higgins CA; Gong W; de Jong A; Harel S; DeStefano GM; Rothman L; et al. Alopecia areata is driven by cytotoxic T lymphocytes and is reversed by JAK inhibition. Nat. Med 2014, 20 (9), 1043–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (35).Pratt CH; King LE Jr.; Messenger AG; Christiano AM; Sundberg JP Alopecia areata. Nat. Rev. Dis Primers 2017, 3, No. 17011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (36).Glickman JW; Dubin C; Renert-Yuval Y; Dahabreh D; Kimmel GW; Auyeung K; Estrada YD; Singer G; Krueger JG; Pavel AB; Guttman-Yassky E. Cross-sectional study of blood biomarkers of patients with moderate to severe alopecia areata reveals systemic immune and cardiovascular biomarker dysregulation. J. Am. Acad. Dermatol 2021, 84 (2), 370–380. [DOI] [PubMed] [Google Scholar]
- (37).McPhee CG; Duncan FJ; Silva KA; King LE Jr.; Hogenesch H; Roopenian DC; Everts HB; Sundberg JP Increased expression of Cxcr3 and its ligands, Cxcl9 and Cxcl10, during the development of alopecia areata in the mouse. J. Invest. Dermatol 2012, 132 (6), 1736–1738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (38).Hoffmann R. The potential role of cytokines and T cells in alopecia areata. J. Invest. Dermatol. Symp. Proc 1999, 4 (3), 235–238. [DOI] [PubMed] [Google Scholar]
- (39).Hoffmann R; Eicheler W; Wenzel E; Happle R. Interleukin-1beta-induced inhibition of hair growth in vitro is mediated by cyclic AMP. J. Invest. Dermatol 1997, 108 (1), 40–42. [DOI] [PubMed] [Google Scholar]
- (40).Hoffmann R; Eicheler W; Huth A; Wenzel E; Happle R. Cytokines and growth factors influence hair growth in vitro. Possible implications for the pathogenesis and treatment of alopecia areata. Arch. Dermatol. Res 1996, 288 (3), 153–156. [DOI] [PubMed] [Google Scholar]
- (41).Hoffmann R; Wenzel E; Huth A; van der Steen P; Schaufele M; Henninger HP; Happle R. Cytokine mRNA levels in Alopecia areata before and after treatment with the contact allergen diphenylcyclopropenone. J. Invest. Dermatol 1994, 103 (4), 530–533. [DOI] [PubMed] [Google Scholar]
- (42).Fetter T; de Graaf DM; Claus I; Wenzel J. Aberrant inflammasome activation as a driving force of human autoimmune skin disease. Front. Immunol 2023, 14, No. 1190388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (43).Chen LL; Morcelle C; Cheng ZL; Chen X; Xu Y; Gao Y; Song J; Li Z; Smith MD; Shi M; et al. Itaconate inhibits TET DNA dioxygenases to dampen inflammatory responses. Nat. Cell Biol 2022, 24 (3), 353–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (44).Xie QM; Chen N; Song SM; Zhao CC; Ruan Y; Sha JF; Liu Q; Jiang XQ; Fei GH; Wu HM Itaconate Suppresses the Activation of Mitochondrial NLRP3 Inflammasome and Oxidative Stress in Allergic Airway Inflammation. Antioxidants 2023, 12 (2), No. 489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (45).Aso K; Kono M; Kanda M; Kudo Y; Sakiyama K; Hisada R; Karino K; Ueda Y; Nakazawa D; Fujieda Y; et al. Itaconate ameliorates autoimmunity by modulating T cell imbalance via metabolic and epigenetic reprogramming. Nat. Commun 2023, 14 (1), No. 984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (46).Mills EL; Ryan DG; Prag HA; Dikovskaya D; Menon D; Zaslona Z; Jedrychowski MP; Costa ASH; Higgins M; Hams E; et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 2018, 556 (7699), 113–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (47).McGettrick AF; Bourner LA; Dorsey FC; O’Neill LAJ Metabolic Messengers: itaconate. Nat. Metab 2024, 6 (9), 1661–1667. [DOI] [PubMed] [Google Scholar]
- (48).Hoisnard L; Lebrun-Vignes B; Maury S; Mahevas M; El Karoui K; Roy L; Zarour A; Michel M; Cohen JL; Amiot A; et al. Adverse events associated with JAK inhibitors in 126,815 reports from the WHO pharmacovigilance database. Sci. Rep 2022, 12 (1), No. 7140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (49).Mori S; Ogata F; Tsunoda R. Risk of venous thromboembolism associated with Janus kinase inhibitors for rheumatoid arthritis: case presentation and literature review. Clin. Rheumatol 2021, 40 (11), 4457–4471. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





