You are on page 1of 15

BJD

M E D I C A L D E R MA T O L O GY British Journal of Dermatology

Nonclinical and human pharmacology of the potent and


selective topical retinoic acid receptor-c agonist trifarotene*
J. Aubert,1 D. Piwnica,1 B. Bertino,1 S. Blanchet-Rethore,1 I. Carlavan,1 S. De
ret,1 B. Dreno iD ,2,3 B. Gamboa,1
A. Jomard, A.P. Luzy, P. Mauvais, C. Mounier, J. Pascau, I. Pelisson, T. Portal,1 M. Rivier,1 P. Rossio,1
1 1 4 1 1 1

E. Thoreau,1 E. Vial1 and J.J. Voegel iD 1


1
Research Department, Galderma R&D, Les Templiers, 2400 Route des Colles, 06410 Biot, France
2
Department of Dermatology, Nantes University Hospital, Nantes, France
3
CIC, Inserm U892-CNRS 6299, Nantes, France
4
Pharma & Life Sciences Xpert, Antibes, France

Linked Editorial: Balak. Br J Dermatol 2018; 179:231–232.

Summary

Correspondence Background First- and third-generation retinoids are the main treatment for acne.
Johannes J. Voegel. Even though efficacious, they lack full selectivity for retinoic acid receptor (RAR)
E-mail: johannes.voegel@galderma.com c, expressed in the epidermis and infundibulum.
Objectives To characterize the in vitro metabolism and the pharmacology of the
Accepted for publication
7 February 2018
novel retinoid trifarotene.
Materials and methods In vitro assays determined efficacy, potency and selectivity on
Funding sources RARs, as well as the activity on the expression of retinoid target genes in human
The studies have been financed by Galderma R&D, keratinocytes and ex vivo cultured skin. In vivo studies investigated topical comedo-
Sophia Antipolis, France. lytic, anti-inflammatory and depigmenting properties. The trifarotene-induced
gene expression profile was investigated in nonlesional skin of patients with acne
Conflicts of interest
With the exception of B.D. and P.M. the authors
and compared with ex vivo and in vivo models. Finally, the metabolic stability in
are employees of Galderma R&D. human keratinocytes and hepatic microsomes was established.
Results Trifarotene is a selective RARc agonist with > 20-fold selectivity over RARa
J.A. and D.P. contributed equally to this study. and RARb. Trifarotene is active and stable in keratinocytes but rapidly metabolized
by human hepatic microsomes, predicting improved safety. In vivo, trifarotene
*Plain language summary available online
001% applied topically is highly comedolytic and has anti-inflammatory and
DOI 10.1111/bjd.16719 antipigmenting properties. Gene expression studies indicated potent activation of
known retinoid-modulated processes (epidermal differentiation, proliferation,
stress response, retinoic acid metabolism) and novel pathways (proteolysis, trans-
port/skin hydration, cell adhesion) in ex vivo and in vivo models, as well as in human
skin after 4 weeks of topical application of trifarotene 0005% cream.
Conclusions Based on its RARc selectivity, rapid degradation in human hepatic
microsomes and pharmacological properties including potent modulation of epi-
dermal processes, topical treatment with trifarotene could result in good efficacy
and may present a favourable safety profile in acne and ichthyotic disorders.

What’s already known about this topic?


• All-trans and 13-cis retinoic acid, which binds all three retinoic acid receptors
(RARs), and adapalene and tazarotene, which interact preferentially with RARb
and RARc, are first-line treatments for acne and are also used in other inflam-
matory skin diseases.
• Topical formulations of these retinoids avoid significant systemic retinoid-related
side-effects.

442 British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 443

What does this study add?


• Trifarotene is the first fourth-generation retinoid with potent and selective RARc
agonist activity, potentially associated with an improved efficacy/safety ratio com-
pared with less selective RAR agonists.
• The pharmacological potency of trifarotene translates from in vitro models to topi-
cally treated rodent and human skin in vivo. The modulated pathways collectively
are expected to translate into strong clinical efficacy in acne.
• Based on its rapid degradation in human hepatic microsomes, trifarotene is
expected to be rapidly eliminated in the blood stream, thereby potentially provid-
ing good safety, which should be particularly useful for the treatment of patients
with ichthyosis with application on large surface areas.
• Based on the favourable metabolic and pharmacological characteristics of tri-
farotene, it is worth investigating the clinical efficacy of this fourth-generation reti-
noid in acne and lamellar ichthyosis.

Retinoids are an important class of nuclear receptor agonists generation retinoids, is expected to demonstrate a favourable
used in dermatology. They are classified into three genera- systemic safety profile, allowing in addition to acne the topical
tions. Retinol, tretinoin (all-trans retinoic acid, ATRA) and iso- treatment of patients with diseases like lamellar ichthyosis
tretinoin (13-cis retinoic acid) belong to the first generation; where application on large surface areas is required.19–23
etretinate and its metabolite acitretin to the second generation; The present report describes for the first time the in vitro
and tazarotene, through its metabolite tazarotenic acid, and metabolism and the nonclinical and clinical pharmacology of
adapalene, to the third generation.1,2 First- and third-genera- the fourth generation, i.e. the fully RARc-selective, retinoid
tion retinoids are mainstay topical treatments for acne, and trifarotene [see Figure S1 (Supporting Information) for a
target comedones and microcomedones.3,4 In addition, third- depiction of the chemical structure of trifarotene].24
generation retinoids exert anti-inflammatory activity on the
innate immune system.5–7 Materials and methods
Adapalene is a selective agonist of nuclear receptors retinoic
acid receptor (RAR) b and RARc, with lower affinity on Retinoic acid receptor profiling
RARa. Its activity on proliferation and differentiation can be
blocked by a RAR b-c antagonist.8 Like adapalene, tazarotenic RAR activity and selectivity of trifarotene and reference reti-
acid is a selective agonist of RARb and RARc.9 Both adapalene noids were evaluated using transactivation assays with RAR
and tazarotenic acid modulate abnormal keratinocyte differen- and retinoid X receptor (RXR) reporter gene technology
tiation, hyperproliferation, and the expression of inflammatory (details of all materials and methods are given in File S1; see
markers, and both result in low systemic exposure after topical Supporting Information).25
administration.8–12 Even though RARc activity has been shown
to be associated with skin irritation in rodent models,13 we Stability testing in keratinocytes and hepatic microsomes
and others14 have hypothesized that a fully RARc-selective Metabolic stability of retinoids in human keratinocyte mono-
compound should retain strong efficacy for the modulation of cultures and hepatic microsomes was evaluated by measuring
epidermal biology while potentially minimizing side-effects. the loss of the parent product by HPLC/MS/MS (high-perfor-
Our hypothesis is based on the observation that (i) RARb is mance liquid chromatography tandem mass spectrometry) as a
strongly and rapidly induced in human dermal fibroblasts trea- function of time (for details see File S1).
ted with retinoic acid,15–17 (ii) retinoic acid-induced glandular
metaplasia in mouse skin is linked to the dermal expression of
Gene expression in keratinocytes, reconstructed
RARb,18 and (iii) RARb is induced in vivo in retinoid-treated
epidermis and ex vivo cultured skin
rat skin, likely due to dermal induction (our unpublished
observations). We therefore hypothesized that dermal RARb, The activity of retinoids on the expression of target genes in
induced and activated by topical application of nonselective DK-7 human immortalized keratinocytes was evaluated using
retinoids, might significantly participate in retinoid-induced Multiplexed Molecular Profiling (High Throughput Genomics,
skin irritation. A selective RARc agonist would avoid these Tucson, AZ, U.S.A.) assays according to the manufacturer’s
RARb-mediated effects. Moreover, a molecule with further instructions. The expression of target genes in reconstructed
increased hepatic instability, compared with first- and third- human epidermis (RHE) and ex vivo cultured human skin was

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
444 Pharmacology of trifarotene, Aubert et al.

evaluated using reverse transcriptase-polymerase chain reac- weekends) were performed. Biopsies of nonlesional areas of tri-
tion. Detailed methods are provided in File S1. farotene- and vehicle-treated skin were made under local anaes-
thesia at day 27. Biopsies were stored in RNAlater TissueProtect
In vivo topical comedolytic activity Tubes (Qiagen, Les Ulis, France).
Skin biopsies were also collected from ex vivo organ-cultured
The comedolytic properties of trifarotene were assessed in the human skin and fuzzy rats topically treated with trifarotene.
rhino mouse model26,27 after 11 daily topical applications. The Detailed methods are provided in File S1.
dose–response of cream formulations of trifarotene was com-
pared with ATRA 005% (Retacnylâ, Galderma International,
France) and tazarotene 01% (Zoracâ, Pierre Fabre Dermatology, Large-scale gene expression profiling
Boulogne, France). Detailed methods are provided in File S1. Samples from all three studies were homogenized with a
Potter-Elvehjem tissue grinder in lysis buffer for RNA extrac-
In vivo topical anti-inflammatory activity tion (Qiagen). Total RNA was extracted using miRNeasy
extraction kits (Qiagen) according to the manufacturer’s pro-
The anti-inflammatory activity of trifarotene 01%, tazarotene
tocol. RNA quantity was measured using a Thermo ScientificTM
01% and tazarotenic acid 01% was assessed after a single
Nano Drop 8000 spectrophotometer (Thermo Fisher, Illkirch,
topical administration in the TPA (12-O-tetradecanoylphorbol-
France). RNA quality was monitored using a 2100 Bioanalyzer
13-acetate)-induced ear oedema mouse model.28,29 Detailed
(Agilent Technologies, Waldbronn, Germany). Probes were
methods are provided in File S1.
synthesized and then hybridized on Affymetrix U133 Plus 2.0
chips (GeneChipTM, Santa Clara, CA, U.S.A.). All chips were
In vivo topical depigmenting and antipigmenting activity normalized using the robust multi-array average (RMA)
The activity of trifarotene was investigated in studies lasting method.32 Only Affymetrix identifiers with expression ≥ 26 in
6 weeks in the SKH2 mouse. The depigmentation activity of at least one condition were selected. Data analysis was per-
trifarotene 001% was compared with that of adapalene 01% formed on Array Studio software (Omicsoft Corporation, Cary,
and ATRA 001% on the mouse tail. The antipigmenting activ- NC, U.S.A.). Mean expression levels were obtained by calculat-
ity on ultraviolet radiation (UVR)-induced pigmentation of ing the geometric means of the RMA-normalized data for
the tail was determined according to the model described by involved and noninvolved sample group. A two-sided paired
Warren.30 Detailed methods are provided in File S1. t-test was performed using Array Studio (Omicsoft Corpora-
tion), to determine which genes were significantly differen-
tially expressed between trifarotene- and vehicle-treated skin.
Sample collection for large-scale gene expression
The Benjamini–Hochberg false discovery rate (FDR) multiple
profiling
testing correction was applied.33 The raw data are available at
Clinical investigations were conducted in accordance with the NCBI GEO, accession number GSE 107232.
Declaration of Helsinki principles, and the ICH Guideline for
Good Clinical Practice. The clinical study received approval from
Ontology analysis
the ethics committee of Brest, France (reference CPP Ouest 6 -
755). Subjects provided written informed consent prior to biop- Gene Ontology (GO) category enrichment analysis was per-
sies. Subjects with moderate inflammatory acne on the back at formed using DAVID 67 (http://david.abcc.ncifcrf.gov/).
the screening visit, defined by scores of between 2 and 4 for the
whole back, with at least one area scored at 2 and with a maxi-
Immunohistochemistry
mum of three nodules using the ECLA (Acne Lesion Score Scale)
scale,31 were included in the study. Nineteen applications of a Five-lm sections were prepared from paraffin-embedded
dose of 2 mg cm 2 of trifarotene 0005% (50 lg g 1) and biopsies, then deparaffinized and rehydrated. Antigen retrieval
vehicle cream on the back for 4 weeks (every day except was performed using heat at pH = 8. The list of antibodies

Table 1 Retinoic acid receptor profiling

RARa RARb RARc


1 1
EC50 (nmol L ) Efficacy (%) EC50 (nmol L ) Efficacy (%) EC50 (nmol L 1) Efficacy (%)
ATRA 09 105 09 82 07 89
Adapalene67 22 100 2 100 9 100
Tazarotenic acid 11 104 25 74 11 86
Trifarotene 498 63 125 58 77 85

ATRA, all-trans retinoic acid.

British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 445

used for detection is detailed in File S1. Labelling was revealed Table 2 Half-life of retinoids in human keratinocytes and hepatic
using a detection kit (alkaline phosphatase/fast red substrate) microsomes
and nuclei were counterstained with haematoxylin. Slides
were observed and scanned using a NanoZoomer (Hamamatsu T1/2: human T1/2: human
Retinoid keratinocytes hepatic microsomes
Photonics, Hamamatsu City, Japan).
Adapalene > 24 h > 60 min
Tazarotene NA (not stable in 7 min
Results
medium)
Tazarotenic acid > 24 h 57 min
Trifarotene is a pure and potent retinoic acid receptor-c Trifarotene > 24 h 5 min
agonist
*
NA, not applicable.
RAR profiling assays showed that trifarotene is RARc-selective
with an efficacy of 81% and a half maximal effective concentration
(EC50) of 77 nmol L 1, which is 16-fold and 65-fold lower than tazarotene was metabolized in keratinocytes and more rapidly
the EC50 on RARb and RARa, respectively (Table 1). The mea- in hepatocytes. The active compound, tazarotenic acid, was
sured EC50 of ATRA on RARc was 07 nmol L 1 (89% efficacy), 10-fold more stable than trifarotene in human hepatic micro-
that of adapalene was 9 nmol L 1 (100% efficacy) and that of somes, with a t1/2 of 57 min. Adapalene had a t1/2 in hepatic
tazarotenic acid 11 nmol L 1 (86% efficacy). Efficacy and microsomes of > 60 min.
potency on RARc are therefore similar for adapalene, tazarotene
and trifarotene. Moreover, trifarotene was inactive on RXR (the Trifarotene is a potent modulator of target gene
chemical structure of trifarotene and details of pharmacological expression in keratinocytes, reconstructed epidermis and
profiling are given in Figures S1–S3; see Supporting Information). ex vivo cultured human skin
Gene expression of a set of target genes was assessed in DK-7
Trifarotene is stable in keratinocytes and unstable in
immortalized keratinocytes. The mean EC50 on the combined
hepatic microsomes
target genes was determined at 009 nmol L 1 for trifarotene,
Trifarotene was stable in human keratinocytes for more than 12 nmol L 1 for tazarotenic acid and 12 nmol L 1 for ATRA
24 h and very rapidly metabolized in human liver micro- (Fig. 1a). Table S1 (see Supporting Information) provides
somes, with a half-life (t1/2) of 5 min (Table 2). The prodrug individual EC50 values for each retinoid response gene.

(a)
Trifarotene Tazaarotenic acid ATTRA
Mean EC50 = 0.09 nmol L–1
Mean EC50 = 1·2 nmol L–1 Mean EC50 = 12 nmol L–1

(b) mol L–1


EC50 = 3 µm EC50 = 1.9 µmol L–1 EC50 = 30 µmol L–1

Fig 1. In vitro pharmacology of trifarotene. (a) Dose–response curves of trifarotene, tazarotenic acid and all-trans retinoic acid (ATRA) in DK-7
human immortalized keratinocytes. Individual EC50 values for each induced retinoid response gene are reported in Table S1 (see Supporting
Information). (b) Topical activity of trifarotene on gene markers in human reconstructed epidermis. Genes for which the expression was
significantly modulated by trifarotene: KRT2, KRT4, KRT10, KRT19, TGM3 and DSC1 (differentiation process); IL1B, LCN2 and PTGES (inflammation
process). The geometric mean EC50 of fold modulations (FM) is reported. Mean EC50 values include induced and repressed genes. For repressed
genes (FM < 1), the transformed 1/FM was used.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
446 Pharmacology of trifarotene, Aubert et al.

In reconstructed human epidermis (RHE), expression of details see File S1). As expected, following 11 daily applica-
KRT2, KRT4, KRT10, KRT19, TGM3 and DSC1, involved in epider- tions, the trifarotene placebo cream had no comedolytic activ-
mal differentiation, and of IL1B, LCN2 and PTGES, involved in ity (61 comedones mm 1) and was well tolerated. ATRA
inflammation, was modulated by the topical application of 005% cream had a marked comedolytic activity, with a 61%
retinoids. The mean EC50 on the combined target genes of tri- reduction of comedones (24 comedones mm 1). At day 12 it
farotene and tazarotenic acid was 3 lmol L 1 and also produced an increase of 240% in epidermal thickness
19 lmol L 1, respectively, and that of ATRA was (60 lm) and increased transepidermal water loss (TEWL) by
30 lmol L 1 (Fig. 1b). 285% (26 g h 1 m 2) (Fig. 2c, d). Both parameters are typi-
Finally, in an ex vivo human skin explant model with a skin cally associated with retinoid activity in vivo.
barrier similar to human skin, trifarotene modulated genes The dose of 01% of tazarotene, required to reach full effi-
involved in keratinization, desquamation, cornification and cell cacy (Fig. S5; see Supporting Information), resulted in a 99%
adhesion (Table 3). The mean EC50 on the combined target reduction of comedones (007 comedones mm 1); it
genes was 00048% for trifarotene and 0013% for tazarotenic increased epidermal thickness and TEWL by 254% (62 lm)
acid (Fig. S4; see Supporting Information). and 451% (37 g h 1 m 2), respectively.
Trifarotene showed dose-dependent comedolytic activity,
being fully efficacious at 001% (98% reduction, 015 come-
Trifarotene is an efficient comedolytic agent in the rhino
dones mm 1), a concentration 10-fold lower than that
mouse
required for tazarotene to achieve similar results. At the same
The comedolytic activity of retinoids was evaluated in the dose trifarotene increased the epidermis thickness by 275%
well-established rhino mouse model (Fig. 2a, b)8,34 (for (66 lm) and the TEWL by 285% (26 g h 1 m 2).

Table 3 Topical activity on gene markers in human ex vivo cultured skin from three donors. Mean fold modulations, normalized on the nontreated
condition, are indicated for each dose applied, of either trifarotene or tazarotenic acid

British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 447

(a) Number of comedones


8

Number of comedones mm–1


–1% Nontreated
ns
–14% Placebo
6 *
Trifarotene 0·001%
–45%
Trifarotene 0·0025%
*
4 Trifarotene 0·005%
–61%
*
–77% Trifarotene 0·01%
2 *
Tretinoin 0·05%
–98% –99%
*
Tazarotene 0·1%
*
0
(b)

Fig 2. Efficacy of trifarotene in the rhino


mouse model. (a) Following 11 daily topical
applications with placebo, trifarotene 0001%,
00025%, 0005% and 001%, Retacnyl cream
(tretinoin) 005% or Zorac cream 01%
(tazarotene), the number of comedones was
counted on skin histology sections stained
with haematoxylin–phloxin–saffron. Results
are expressed as mean number of comedones
mm 1 of epidermis  SEM (3 sections per/
animal, n = 6). Percentage reductions
compared with the nontreated condition are
indicated. (b) Images of representative skin
histology sections: placebo (upper left panel),
trifarotene 001% (upper right panel), (c) Epidermal thickness
80 275%
Retacnyl cream 005% (lower left panel) and 254%
Nontreated
Epidermal thickness (µm)

*
223% 240% *
Zorac cream 01% (lower right panel). 215%
* * * Placebo
Inverted triangles, epidermis; arrows, 60 159%
Trifarotene 0·001%
*
sebaceous glands; triangles, utriculi. (c) On Trifarotene 0·0025%
the same sections, epidermal thickness was 40
36% Trifarotene 0·005%
also evaluated and expressed in mean ns
Trifarotene 0·01%
lm  SEM. Percentage increases compared 20 Tretinoin 0·05%
with the nontreated condition are indicated. Tazarotene 0·1%
(d) Transepidermal water loss (TEWL) was 0
monitored on restrained animals at the end of
the treatment using a Tewameter device and (d) 50 TEWL
451% Nontreated
expressed in mean g h 1 m 2  SEM *
40 Placebo
(n = 6). Percentage increases compared with
TEWL (g h–1 m–2)

271%
285% 285% Trifarotene 0·001%
the nontreated condition are indicated. All 30 238% 230%
* *
*
*
Trifarotene 0·0025%
statistical analyses were performed using *
Trifarotene 0·005%
multiple comparisons by Wilcoxon test and 20
22% Trifarotene 0·01%
Bonferroni P-value correction. As Retacnyl ns
Tretinoin 0·05%
10
005% and Zorac 01% placebos were not Tazarotene 0·1%
available for testing, all results are presented 0
vs. the nontreated group. *P-value < 005.

Trifarotene demonstrates anti-inflammatory properties Trifarotene shows depigmenting and antipigmenting


in vivo activity in vivo
The anti-inflammatory effects of retinoids, known to be Depigmenting activity of retinoids has been observed in SKH2
mediated in part by anti-AP-1 activity,35 were evaluated in mice.37 Unlike other retinoids, and potentially due to a
the TPA-induced ear oedema mouse model.8,36 All tested weaker penetration in hyperkeratotic tail skin, adapalene 01%
compounds, including the RARc-selective agonist trifarotene, showed no significant depigmenting activity after 6 weeks of
showed potent anti-inflammatory characteristics in vivo, result- topical application on the SKH2 mouse tail (Fig. 3b). In con-
ing in ear oedema reduction of 73%, 99% and 95% for tri- trast, trifarotene and ATRA showed significant depigmenting
farotene, tazarotene and tazarotenic acid, respectively activity at 001% ( 11 and 1, respectively, on the pigmen-
(Fig. 3a). tation score at day 43). After UVR induction, in the same

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
448 Pharmacology of trifarotene, Aubert et al.

(a) TPA inflammation


Fig 3. Anti-inflammatory and antipigmenting
20
TPA + vehicle activities of trifarotene in vivo. The anti-
Ear oedema (1/100 mm)

TPA + trifarotene 0·1% inflammatory activity of vehicle, trifarotene


15 01%, tazarotene 01% and tazarotenic acid
TPA + tazarotene 0·1%
01% was evaluated after a single topical co-
TPA + tazarotenic acid 0·1%
application with TPA 001% on Balb/c mouse
10
ears. The ear oedema (increase in ear
–75%
***
thickness) was measured using a micrometer
5 –95% (Oditest) 6 h after application and expressed
–99%
***
*** in mean 1/100 mm increase  SEM (n = 5).
Percentage decreases compared with the
0
vehicle-treated condition are indicated.
(b) Pigmentation Statistical analyses were performed using
Student’s t-test: *001 < P-value < 005;
0·0
Trifarotene 0·01% **0005 < P-value < 001; ***P-value
ns
Tretinoin 0·01% < 0005. (b) The depigmenting activity of
Pigmentation score

–0·5 vehicle, trifarotene 001%, tretinoin 001%


Adapalene 0·1%
and adapalene 01% was evaluated after 6-
* Vehicle
week daily topical applications on the tail of
–1·0
SKH2 mice. Results are expressed as mean
***
pigmentation score [from 0 (naturally
–1·5 pigmented) to 4 (totally depigmented)] 
SEM (n = 5). Statistical analyses at day 43
were performed using Student’s t-test:
–2·0
D1 D8 D15 D22 D29 D36 D43
*001 < P-value < 005; **0005 < P-value
< 001; ***P-value < 0005. (c) The
(c) Pigmentation + UV antipigmenting activity of trifarotene 0003%
6
UV + trifarotene 0·003% was evaluated after 6-week daily topical
UV + vehicle applications on the tail of SKH2 mice in
4
Pigmentation score

***

Trifarotene 0·003%
conjunction with ultraviolet (UV) B
irradiations (90 mJ cm 2 three times a week
2 Vehicle
for 6 weeks). Results are expressed as mean
pigmentation score [from 4 (totally
0
pigmented) to 4 (totally depigmented)] 
SEM (n = 5). Statistical analyses at day 42
–2
*** were performed using Student’s t-test:
*** *001 < P-value < 005; **0005 < P-value
–4
D1 D8 D15 D22 D29 D36 D42
< 001; ***P-value < 0005. TPA, 12-O-
tetradecanoylphorbol-13-acetate.

mouse model, the antipigmenting activity of trifarotene was metabolism (CYP26A1, STRA6, DHRS9, CRABP2), epidermal differ-
again highly significant (Fig. 3c). entiation (KRT4, ELF3, PPARD, ID1), proliferation (FOSL1, P2RY2,
RIT1, CCNG2, ZBTB20, FGFR2, BTC) and response to stress
(S100A9, GPX2, IL1RN, CXCR2, TYMS, F3). Interestingly, three reti-
Translational transcriptomics with trifarotene, including
noid-modulated pathways were identified that have not yet been
human skin in vivo, demonstrates potent activation of
described: proteolysis (PRSS27, KLK6, KLK8, KLK10, CTSD, SER-
known and novel retinoid-modulated pathways
PINA12, MME) (Fig. 4b), transport/skin hydration (PADI1, AQP3,
The pharmacodynamic effects of trifarotene were investigated in RHCG, ATP11B) (Fig. 4c) and cell adhesion (PRRG4, EFHD2,
noninflammatory skin of patients with acne after 4 weeks of CHL1, SPON1, DST) (Fig. 4d). For the proteolysis pathway, the
topical treatment with trifarotene 0005% cream. Retinoid- induction of KLK6, KLK8 and KLK10 expression was confirmed by
modulated genes observed in the human study were compared immunohistochemistry (Fig. 4e).
with transcriptomics data obtained after topical treatment in
human ex vivo skin culture and in the fuzzy rat in vivo. Forty-one
Discussion
genes were modulated in the same direction in all three models,
and formed the basis for biological interpretation (Fig. 4a and We described the pharmacology of trifarotene, the first potent
Table 4). Trifarotene regulated the expression of genes involved RARc-selective agonist entering clinical trials. The molecule
in known retinoid-induced pathways, including retinoid was designed to present good metabolic stability in cultured

British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 449

(a) Noninflammatory Fuzzy rat


skin of in vivo
acne patients 0 72 (312 genes)
(214 genes)
1

47
95 192
Ex vivo cultured
human skin 0
(334 genes)

(b)

Fig 4. Comparative analysis of trifarotene


(Trif)-induced gene modulation in three
pharmacological studies: ex vivo cultured
human skin, fuzzy rat in vivo and
noninflammatory skin of patients with acne.
(a) Venn diagram including data of three
experimental studies, depicting genes
modulated with a fold modulation > 16 and
a Benjamini–Hochberg false discovery
rate < 005. Forty-seven genes were found in
the union of all three studies. Of these, 41
were modulated in the same direction in all
three studies, and used for further analysis.
(b) Genes involved in proteolysis and
modulated by topical trifarotene application in
all three studies. Expression levels are
provided in arbitrary units (AU). Continued
over (c) Genes involved in skin hydration and
transport, and modulated by topical
trifarotene application in all three studies.
Expression levels are provided in AU. (d)
Genes involved in cell adhesion and
modulated by topical trifarotene application in
all three studies. Expression levels are
provided in AU. (e) Immunohistochemical
staining of three kallikrein members on skin
sections obtained from vehicle- and
trifarotene-treated noninflammatory skin of
patients with acne. Ctrl, control.

keratinocytes, while being rapidly metabolized in human liver compounds.38,39 Importantly, topically applied trifarotene was
microsomes. The in vitro metabolic profile of trifarotene is active in ex vivo cultured human skin, with a stratum corneum
expected to result in low systemic levels, while retaining barrier close to human skin in vivo. Interestingly, in this assay
strong cutaneous activity. These properties are particularly trifarotene was threefold more potent than tazarotene, which
important when treating large body surface areas topically in is in agreement with the demonstrated overall higher in vitro
diseases such as lamellar ichthyosis, as potential systemic safety potency of trifarotene compared with tazarotenic acid, the
issues may thus be reduced. active metabolite of the prodrug tazarotene.
The potent pharmacological efficacy of trifarotene was con- In vivo, trifarotene eliminated almost all comedones from the
firmed in the pluristratified RHE model. Although presenting classical retinoid-responsive rhino mouse model, with a dose
only limited barrier properties, the RHE model is routinely 10 times lower than that required for tazarotene and ATRA to
used for the topical efficacy evaluation of skin-targeted achieve the same effects.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
450 Pharmacology of trifarotene, Aubert et al.

(c)

(d)

Fig 4. Continued

In line with the described effects of retinoids,40–42 tri- note, toATRA is already recommended to reduce PIHP sec-
farotene demonstrated strong anti-inflammatory properties, ondary to acne.43,44
which in addition to the comedolytic activity is expected to To evaluate if the potent in vitro and in vivo pharmacological
contribute to its efficacy in inflammatory skin diseases such as activities of trifarotene translate to the clinical setting, topical
acne. trifarotene 0005% cream was applied to noninflammatory
Inflammatory acne is often accompanied by post-inflamma- skin of patients with acne, followed by large-scale gene
tory hyperpigmentation (PIHP), especially in Black and Asian expression profiling in skin biopsies. Transcriptomics was per-
subjects. The rapid in vivo antipigmenting activity of trifarotene formed in parallel in ex vivo cultured human skin, and in the
is therefore another useful characteristic of this molecule. Of fuzzy rat in vivo model. Forty-one genes modulated in all three

British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 451

(e) Vehicle Trifarotene

KLK6 KLK6

KLK8 KLK8

KLK10 KLK10

Fig 4. Continued

topical models provided a robust set of trifarotene-biomarkers, associated with late stages of epidermal differentiation, where it
including genes involved in retinoid-modulated processes such catalyses deimination of filaggrin and keratin K1.55
as retinoid metabolism,45 and epidermal differentiation and The upregulation of Rh family C glycoprotein (RHCG), an
proliferation.46,47 ammonium transporter, suggests that trifarotene may impact
Within biological factors influencing cellular proliferation, the NH4+/NH3 balance, and thereby cutaneous pH homeosta-
FGFR2 was downregulated by trifarotene. This observation, not sis. Furthermore, a change in the ammonium ion content may
previously described for retinoids, is of particular interest, as a indirectly affect the natural barrier structure, for example by
potential pathogenic role of FGFR2 in acne has been sug- regulating glutamine synthetase and the production of
gested.48,49 Naturally occurring gain-of-function germline glutamine.56,57
mutations of the FGFR2 gene have been demonstrated to cause SLC2A1 or GLUT-1 in humans is the most common trans-
Apert syndrome, characterized by skin manifestations includ- porter of glucose, the major energy source for cells.
ing severe acne and oily skin.50,51 Of note, a case report with 3 Proteolysis: To date, the downregulation of membrane
oral isotretinoin describes the resolution of acneiform lesions metalloendopeptidase (MME) observed with trifarotene has
in Apert syndrome.52 not been described for other retinoids. Of note, upregulated
Importantly, three novel pathways were identified in our expression of MME has been associated with the degradation
comparative large-scale gene expression analysis: (1) cell of elastin fibres, resulting in wrinkling or sagging of the
adhesion, (2) transport/skin hydration and (3) proteolysis: skin.58,59 MME downregulation may therefore constitute
1 Cell adhesion: Genes associated with the cell adhesion another previously not yet recognized pathway involved in the
process may take part in the renewal of epidermal cells.53 As positive effects of retinoids on skin ageing.60,61
such, the downregulation of dystonin by trifarotene may pro- Interestingly, trifarotene induced the expression of several
mote the weakening of hemidesmosomes, inducing the migra- serine proteases, including kallikreins (KLK6, KLK8, KLK10)
tion of keratinocytes, thereby supporting the anticomedolytic and serine protease PRSS27. The involvement of these pro-
activity of the drug. teases in the desquamation process62,63 and their upregulated
2 Transport/skin hydration: By activating the expression of expression in psoriasis64 has been described previously.
aquaporin 3 (AQP3) and peptidyl arginine deiminase 1 (PADI1), Whereas discordant results, i.e. induced and repressed expres-
trifarotene may improve skin hydration and impact cutaneous sion, respectively, have been described for other kallikrein
barrier function. Water channel AQP3 transports both water and members (KLK5 and KLK7) in cultured keratinocytes treated
small neutral solutes, such as the humectant glycerol,54 important with retinoids,65,66 we demonstrated consistent upregulation
for maintaining hydration of the stratum corneum. PADI1 is of KLK6, KLK8 and KLK10 in three independent experimental

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
Table 4 Gene expression profile induced by trifarotene across three experimental models: ex vivo cultured human skin, fuzzy rat in vivo and noninflammatory skin of patients with acne. Genes commonly
modulated in all three models are grouped according to their main biological function provided by DAVID tools. References for genes known to be regulated by retinoids are indicated in brackets
452 Pharmacology of trifarotene, Aubert et al.

British Journal of Dermatology (2018) 179, pp442–456


© 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 453

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
454 Pharmacology of trifarotene, Aubert et al.

models on the mRNA level and in human skin in vivo on the 12 Attar M, Yu D, Ni J et al. Disposition and biotransformation of the
protein level. This observation may provide a partial molecular acetylenic retinoid tazarotene in humans. J Pharm Sci 2005;
explanation for the effect of trifarotene on the renewal of ter- 94:2246–55.
13 Chen S, Ostrowski J, Whiting G et al. Retinoic acid receptor
minally differentiated corneocytes in human skin.
gamma mediates topical retinoid efficacy and irritation in animal
This report is limited to pharmacology and does not describe models. J Invest Dermatol 1995; 104:779–83.
in vivo pharmacokinetics and toxicology data, or data on human 14 Beard RL, Chandraratna RAS. RAR-selective ligands: receptor sub-
safety and cutaneous tolerance. At this point predictions of type and function selectivity. In: Retinoids: The Biochemical and Molecular
improved safety of topically applied trifarotene are therefore Basis of Vitamin A and Retinoid Action (Nau H, Blaner WS, eds). Berlin,
hypothetical and based only on rapid metabolism in hepatic Heidelberg: Springer, 1999; 185–213.
microsomes in vitro and strong selectivity of trifarotene for 15 Elder JT, Fisher GJ, Zhang QY et al. Retinoic acid receptor gene
expression in human skin. J Invest Dermatol 1991; 96:425–33.
RARc. Future investigations with a suitably formulated drug
16 Redfern CP, Todd C. Retinoic acid receptor expression in human
product will have to investigate safety and tolerance as well as skin keratinocytes and dermal fibroblasts in vitro. J Cell Sci 1992;
clinical efficacy in dermatological diseases and in skin ageing. 102 (Pt 1):113–21.
Today, based on the favourable metabolic and pharmaco- 17 Tsou HC, Lee X, Si SP, Peacocke M. Regulation of retinoic acid
logical characteristics of trifarotene described in this report, receptor expression in dermal fibroblasts. Exp Cell Res 1994;
this fourth-generation retinoid is under investigation for clini- 211:74–81.
cal efficacy in acne and lamellar ichthyosis. 18 Viallet JP, Ruberte E, du Manoir S et al. Retinoic acid-induced
glandular metaplasia in mouse skin is linked to the dermal expres-
sion of retinoic acid receptor beta mRNA. Dev Biol 1991; 144:424–8.
Acknowledgments 19 Beckenbach L, Baron JM, Merk HF et al. Retinoid treatment of skin
diseases. Eur J Dermatol 2015; 25:384–91.
The excellent scientific and experimental contributions of G. 20 Fleckman P, Newell BD, van Steensel MA, Yan AC. Topical treat-
Duvert, G. Feraille, C. Feret, P. Fogel, M. Freylinger, D. ment of ichthyoses. Dermatol Ther 2013; 26:16–25.
Froude, C. Menigot, T. Minier, K. Nicolas, P. Reiniche and D. 21 Hernandez-Martin A, Aranegui B, Martin-Santiago A, Garcia-Doval
Spiesse are gratefully acknowledged. The authors acknowledge I. A systematic review of clinical trials of treatments for the con-
genital ichthyoses, excluding ichthyosis vulgaris. J Am Acad Dermatol
the writing assistance of Karl Patrick G€
oritz, SMWS, Scientific
2013; 69:544–9.e8.
and Medical Writing Services France. 22 Hofmann B, Stege H, Ruzicka T, Lehmann P. Effect of topical
tazarotene in the treatment of congenital ichthyoses. Br J Dermatol
1999; 141:642–6.
References 23 Marulli GC, Campione E, Chimenti MS et al. Type I lamellar
1 Chambon P. A decade of molecular biology of retinoic acid recep- ichthyosis improved by tazarotene 0.1% gel. Clin Exp Dermatol 2003;
tors. FASEB J 1996; 10:940–54. 28:391–3.
2 Germain P, Chambon P, Eichele G et al. International Union of 24 Thoreau E, Arlabosse JM, Bouix-Peter C et al. Structure-based
Pharmacology. LX. Retinoic acid receptors. Pharmacol Rev 2006; design of trifarotene (CD5789), a potent and selective RARc ago-
58:712–25. nist for the treatment of acne. Bioorg Med Chem Lett 2018; 28:1736–
3 Dreno B, Gollnick HP, Kang S et al. Understanding innate immu- 41.
nity and inflammation in acne: implications for management. J Eur 25 Cheng Z, Garvin D, Paguio A et al. Luciferase reporter assay system
Acad Dermatol Venereol 2015; 29 (Suppl. 4):3–11. for deciphering GPCR pathways. Curr Chem Genom 2010; 4:84–91.
4 Webster GF. Evidence-based review: fixed-combination therapy 26 Kligman LH, Kligman AM. The effect on rhino mouse skin of
and topical retinoids in the treatment of acne. J Drugs Dermatol agents which influence keratinization and exfoliation. J Invest Derma-
2011; 10:636–44. tol 1979; 73:354–8.
5 Leyden J. Adapalene in clinical practice. Cutis 2001; 68 (Suppl. 27 Van Scott EJ, Yu RJ. Control of keratinization with alpha-hydroxy
4):7–9. acids and related compounds. I. Topical treatment of ichthyotic
6 Bershad S, Poulin YP, Berson DS et al. Topical retinoids in the disorders. Arch Dermatol 1974; 110:586–90.
treatment of acne vulgaris. Cutis 1999; 64 (Suppl. 2):8–20; quiz 28 Bird J, Kim HP, Lee HJ. Topical anti-inflammatory activity of esters
1–3. of steroid 21-oic acids. Steroids 1986; 47:35–40.
7 Chandraratna RA. Tazarotene – first of a new generation of recep- 29 Carlson RP, O’Neill-Davis L, Chang J, Lewis AJ. Modulation of
tor-selective retinoids. Br J Dermatol 1996; 135 (Suppl. 49):18–25. mouse ear edema by cyclooxygenase and lipoxygenase inhibitors
8 Michel S, Jomard A, Demarchez M. Pharmacology of adapalene. Br and other pharmacologic agents. Agents Actions 1985; 17:197–204.
J Dermatol 1998; 139 (Suppl. 52):3–7. 30 Warren R. Sensitivity of mouse Skh-HR-2 to ultraviolet light:
9 Nagpal S, Thacher SM, Patel S et al. Negative regulation of two mouse pigmentation model. Photochem Photobiol 1986; 43:41–7.
hyperproliferative keratinocyte differentiation markers by a 31 Dreno B, Bodokh I, Chivot M et al. [ECLA grading: a system of
retinoic acid receptor-specific retinoid: insight into the mecha- acne classification for every day dermatological practice]. Ann Der-
nism of retinoid action in psoriasis. Cell Growth Differ 1996; matol Venereol 1999; 126:136–41 (in French).
7:1783–91. 32 Irizarry RA, Hobbs B, Collin F et al. Exploration, normalization,
10 Tenaud I, Khammari A, Dreno B. In vitro modulation of TLR-2, and summaries of high density oligonucleotide array probe level
CD1d and IL-10 by adapalene on normal human skin and acne data. Biostatistics 2003; 4:249–64.
inflammatory lesions. Exp Dermatol 2007; 16:500–6. 33 Benjamini Y, Hochberg Y. Controlling the false discovery rate: a
11 Wolf JE Jr. Potential anti-inflammatory effects of topical retinoids practical and powerful approach to multiple testing. J Royal Stat Soc
and retinoid analogues. Adv Ther 2002; 19:109–18. B Method 1995; 57:289–300.

British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists
Pharmacology of trifarotene, Aubert et al. 455

34 Shroot B, Michel S. Pharmacology and chemistry of adapalene. J 55 Dong S, Ying S, Kojima T et al. Crucial roles of MZF1 and Sp1 in
Am Acad Dermatol 1997; 36 (6 Pt 2):S96–103. the transcriptional regulation of the peptidylarginine deiminase
35 Fisher GJ, Talwar HS, Lin J et al. Retinoic acid inhibits induction of type I gene (PADI1) in human keratinocytes. J Invest Dermatol 2008;
c-Jun protein by ultraviolet radiation that occurs subsequent to 128:549–57.
activation of mitogen-activated protein kinase pathways in human 56 Danielyan L, Zellmer S, Sickinger S et al. Keratinocytes as deposi-
skin in vivo. J Clin Invest 1998; 101:1432–40. tory of ammonium-inducible glutamine synthetase: age- and anat-
36 Dawson MI, Park JH, Chen G et al. Retinoic acid (RA) receptor omy-dependent distribution in human and rat skin. PLOS One
transcriptional activation correlates with inhibition of 12-O-tetra- 2009; 4:e4416.
decanoylphorbol-13-acetate-induced ornithine decarboxylase 57 Baday S, Orabi EA, Wang S et al. Mechanism of NH4+ recruitment
(ODC) activity by retinoids: a potential role for trans-RA-induced and NH3 transport in Rh proteins. Structure 2015; 23:1550–7.
ZBP-89 in ODC inhibition. Int J Cancer 2001; 91:8–21. 58 Imokawa G, Nakajima H, Ishida K. Biological mechanisms under-
37 Nair X, Tramposch KM. UVB-induced pigmentation in hairless lying the ultraviolet radiation-induced formation of skin wrinkling
mice as an in vivo assay for topical skin-depigmenting activity. Skin and sagging II: over-expression of neprilysin plays an essential
Pharmacol 1989; 2:187–97. role. Int J Mol Sci 2015; 16:7776–95.
38 Bernard FX, Pedretti N, Rosdy M, Deguercy A. Comparison of 59 Imokawa G. Epithelial–mesenchymal interaction mechanisms lead-
gene expression profiles in human keratinocyte mono-layer cul- ing to the over-expression of neprilysin are involved in the UVB-
tures, reconstituted epidermis and normal human skin; transcrip- induced formation of wrinkles in the skin. Exp Dermatol 2016; 25
tional effects of retinoid treatments in reconstituted human (Suppl. 3):2–13.
epidermis. Exp Dermatol 2002; 11:59–74. 60 Mukherjee S, Date A, Patravale V et al. Retinoids in the treatment
39 Eichner R. Epidermal effects of retinoids: in vitro studies. J Am Acad of skin aging: an overview of clinical efficacy and safety. Clin Interv
Dermatol 1986; 15 (4 Pt 2):789–97. Aging 2006; 1:327–48.
40 Akdeniz N, Calka O, Ozbek H, Metin A. Anti-inflammatory effects 61 Varani J, Fisher GJ, Kang S, Voorhees JJ. Molecular mechanisms of
of tretinoin (all-trans-retinoic acid) 0.1% and adapalene 0.1% in intrinsic skin aging and retinoid-induced repair and reversal. J
rats. Clin Exp Dermatol 2005; 30:570–2. Investig Dermatol Symp Proc 1998; 3:57–60.
41 Brogden RN, Goa KE. Adapalene. A review of its pharmacological 62 Miyai M, Matsumoto Y, Yamanishi H et al. Keratinocyte-specific
properties and clinical potential in the management of mild to mesotrypsin contributes to the desquamation process via kallikrein
moderate acne. Drugs 1997; 53:511–19. activation and LEKTI degradation. J Invest Dermatol 2014; 134:1665–
42 Becherel PA, Mossalayi MD, LeGoff L et al. Mechanism of anti- 74.
inflammatory action of retinoids on keratinocytes. Lancet 1994; 63 Borgono CA, Michael IP, Komatsu N et al. A potential role for mul-
344:1570–1. tiple tissue kallikrein serine proteases in epidermal desquamation. J
43 Yin NC, McMichael AJ. Acne in patients with skin of color: practi- Biol Chem 2007; 282:3640–52.
cal management. Am J Clin Dermatol 2014; 15:7–16. 64 Komatsu N, Saijoh K, Kuk C et al. Aberrant human tissue kallikrein
44 Woolery-Lloyd HC, Keri J, Doig S. Retinoids and azelaic acid to levels in the stratum corneum and serum of patients with psoria-
treat acne and hyperpigmentation in skin of color. J Drugs Dermatol sis: dependence on phenotype, severity and therapy. Br J Dermatol
2013; 12:434–7. 2007; 156:875–83.
45 Heise R, Mey J, Neis MM et al. Skin retinoid concentrations are 65 Morizane S, Yamasaki K, Kabigting FD, Gallo RL. Kallikrein expres-
modulated by CYP26AI expression restricted to basal keratinocytes sion and cathelicidin processing are independently controlled in
in normal human skin and differentiated 3D skin models. J Invest keratinocytes by calcium, vitamin D, and retinoic acid. J Invest Der-
Dermatol 2006; 126:2473–80. matol 2010; 130:1297–306.
46 Varani J, Zeigler M, Dame MK et al. Heparin-binding epidermal- 66 Kim MY, Lee SE, Chang JY, Kim SC. Retinoid induces the degrada-
growth-factor-like growth factor activation of keratinocyte ErbB tion of corneodesmosomes and downregulation of corneodesmo-
receptors mediates epidermal hyperplasia, a prominent side-effect somal cadherins: implications on the mechanism of retinoid-
of retinoid therapy. J Invest Dermatol 2001; 117:1335–41. induced desquamation. Ann Dermatol 2011; 23:439–47.
47 Fisher GJ, Voorhees JJ. Molecular mechanisms of retinoid actions 67 Grosshans E, Marks R, Mascaro JM et al. Evaluation of clinical effi-
in skin. FASEB J 1996; 10:1002–13. cacy and safety of adapalene 0.1% gel versus tretinoin 0.025% gel
48 Rees J. FGFR2 mutations and acne. Lancet 1998; 352:668–9. in the treatment of acne vulgaris, with particular reference to the
49 Melnik B, Schmitz G. FGFR2 signaling and the pathogenesis of onset of action and impact on quality of life. Br J Dermatol 1998;
acne. J Dtsch Dermatol Ges 2008; 6:721–8. 139 (Suppl. 52):26–33.
50 Freiman A, Tessler O, Barankin B. Apert syndrome. Int J Dermatol 68 Szeto W, Jiang W, Tice DA et al. Overexpression of the retinoic acid-
2006; 45:1341–3. responsive gene Stra6 in human cancers and its synergistic induction
51 Zhang Y, Gorry MC, Post JC, Ehrlich GD. Genomic organization of by Wnt-1 and retinoic acid. Cancer Res 2001; 61:4197–205.
the human fibroblast growth factor receptor 2 (FGFR2) gene and 69 Soref CM, Di YP, Hayden L et al. Characterization of a novel airway
comparative analysis of the human FGFR gene family. Gene 1999; epithelial cell-specific short chain alcohol dehydrogenase/reductase
230:69–79. gene whose expression is up-regulated by retinoids and is
52 Gilaberte M, Puig L, Alomar A. Isotretinoin treatment of acne in a involved in the metabolism of retinol. J Biol Chem 2001;
patient with Apert syndrome. Pediatr Dermatol 2003; 20:443–6. 276:24194–202.
53 Tang HY, Chaffotte AF, Thacher SM. Structural analysis of the 70 Giguere V, Lyn S, Yip P et al. Molecular cloning of cDNA encoding
predicted coiled-coil rod domain of the cytoplasmic bullous a second cellular retinoic acid-binding protein. Proc Natl Acad Sci
pemphigoid antigen (BPAG1). Empirical localization of the N- U S A 1990; 87:6233–7.
terminal globular domain-rod boundary. J Biol Chem 1996; 71 Virtanen M, Sirsjo A, Vahlquist A, Torma H. Keratins 2 and 4/13 in
271:9716–22. reconstituted human skin are reciprocally regulated by retinoids
54 Hara-Chikuma M, Verkman AS. Roles of aquaporin-3 in the epi- binding to nuclear receptor RARalpha. Exp Dermatol 2010; 19:674–
dermis. J Invest Dermatol 2008; 128:2145–51. 81.

© 2018 British Association of Dermatologists British Journal of Dermatology (2018) 179, pp442–456
456 Pharmacology of trifarotene, Aubert et al.

72 Kim HJ, Lotan R. Identification of retinoid-modulated proteins in 82 Sheikh MS, Augereau P, Chalbos D et al. Retinoid regulation of
squamous carcinoma cells using high-throughput immunoblotting. human cathepsin D gene expression. J Steroid Biochem Mol Biol 1996;
Cancer Res 2004; 64:2439–48. 57:283–91.
73 Berry DC, Noy N. All-trans-retinoic acid represses obesity and insu- 83 Bellemere G, Von Stetten O, Oddos T. Retinoic acid increases
lin resistance by activating both peroxisome proliferation-activated aquaporin 3 expression in normal human skin. J Invest Dermatol
receptor beta/delta and retinoic acid receptor. Mol Cell Biol 2009; 2008; 128:542–8.
29:3286–96.
74 Villano CM, White LA. Expression of the helix-loop-helix protein
inhibitor of DNA binding-1 (ID-1) is activated by all-trans retinoic Supporting Information
acid in normal human keratinocytes. Toxicol Applied Pharmacol 2006;
Additional Supporting Information may be found in the online
214:219–29.
75 Ma Y, Koza-Taylor PH, DiMattia DA et al. Microarray analysis version of this article at the publisher’s website:
uncovers retinoid targets in human bronchial epithelial cells. Onco- File S1. Materials and Methods. In vitro retinoic acid recep-
gene 2003; 22:4924–32. tor (RAR) and retinoid X receptor (RXR) profiling; In vitro
76 Fujishita K, Koizumi S, Inoue K. Upregulation of P2Y2 receptors assessment of the activity of trifarotene on the expression of
by retinoids in normal human epidermal keratinocytes. Purinergic retinoid target genes; In vivo assessment of the comedolytic,
Signal 2006; 2:491–8.
anti-inflammatory and depigmenting activity of trifarotene;
77 Rittie L, Varani J, Kang S et al. Retinoid-induced epidermal hyper-
Samples for large-scale gene expression analysis; Antibodies
plasia is mediated by epidermal growth factor receptor activation
via specific induction of its ligands heparin-binding EGF and and dilutions for immunohistochemistry on human skin
amphiregulin in human skin in vivo. J Invest Dermatol 2006; sections.
126:732–9. Table S1 Activity in DK-7 human immortalized ker-
78 Liu TX, Zhang JW, Tao J et al. Gene expression networks underly- atinocytes in culture.
ing retinoic acid-induced differentiation of acute promyelocytic Fig. S1. Chemical structures.
leukemia cells. Blood 2000; 96:1496–504.
Fig. S2. Pharmacological activity on retinoic acid receptors
79 Morbitzer M, Herget T. Expression of gastrointestinal glutathione
(RARa, RARb, RARc).
peroxidase is inversely correlated to the presence of hepatitis C
virus subgenomic RNA in human liver cells. J Biol Chem 2005; Fig. S3. Pharmacological activity on retinoid X receptors
280:8831–41. (RXRa, RXRb).
80 Oeth P, Yao J, Fan ST, Mackman N. Retinoic acid selectively inhi- Fig. S4. Topical activity on gene markers in human skin
bits lipopolysaccharide induction of tissue factor gene expression ex vivo.
in human monocytes. Blood 1998; 91:2857–65. Fig. S5. Dose–response of tazarotene and tazarotenic acid in
81 Zeng M, Zhang Y, Bhat I et al. The human kallikrein 10 promoter
the rhino mouse model.
contains a functional retinoid response element. Biol Chem 2006;
Powerpoint S1 Journal Club Slide Set.
387:741–7.

British Journal of Dermatology (2018) 179, pp442–456 © 2018 British Association of Dermatologists

You might also like