Background:

Cetuximab, an EGFR inhibitor used to treat multiple cancer types, including colon cancer, causes severe skin toxicity in 5%–20% of patients, leading to decreased quality of life and treatment delays. Our understanding of which patients have an increased risk of severe toxicities is limited. We conducted a genome-wide association study to identify germline variants predictive of cetuximab-induced severe skin toxicity.

Methods:

Our study included 1,209 patients with stage III colon cancer randomized to receive cetuximab plus 5-fluorouracil and oxaliplatin as part of the NCCTG N0147 (Alliance) clinical trial. Skin toxicity outcomes were collected using the Common Toxicity Criteria for Adverse Events version 3.0. We performed genotyping, evaluating approximately 10 million genetic variants. We used logistic regression to evaluate the association of each genetic variant and severe (grade ≥ 3) skin toxicity, adjusting for age, sex, and genetic ancestry. Genome-wide significance was defined as P < 5 × 10−8.

Results:

Participants were predominantly middle-aged white men; 20% (n = 243) experienced severe skin toxicity. Two genetic variants in the retinoic acid receptor alpha (RARA) gene were significantly associated with severe skin toxicity [OR, 3.93; 95% confidence interval (CI), 2.47–6.25; P < 7.8 × 10−9]. Functional annotations indicate these variants are in the RARA promoter. Additional significantly associated variants were identified in chromosome 2 intergenic regions.

Conclusions:

Identified variants could represent a potential target for risk stratification of patients with colon cancer receiving cetuximab.

Impact:

Retinoids have shown promise in the treatment of cetuximab-induced skin toxicity, so follow-up work could evaluate whether individuals with the RARA variant would benefit from retinoid therapy.

Cetuximab, a mAb inhibiting the EGFR, is commonly used for treatment of metastatic squamous cell carcinoma of the head and neck and RAS-wild-type colorectal cancer. Skin toxicities, generally in the form of a follicular papulopustular (“acneiform”) rash, are a common, dose-dependent side effect of treatment, affecting >90% of patients (1–3). With the high prevalence of rash in cetuximab-treated patients, prophylactic treatment with sunscreen, topical steroids, and oral antibiotics has become an important adjunct therapy (4, 5). This approach has been shown to reduce the severity of skin toxicity without reducing drug efficacy (6–8). Nonetheless, about 5%–20% of patients still develop skin toxicities severe enough to affect quality of life (9–11) and compromise drug delivery by causing treatment delays, dose reductions, or even discontinuation of therapy (1, 6, 12).

Our understanding of why patients develop EGFR-associated skin toxicity and which patients have an increased risk of severe toxicities is limited. It appears to be a class effect related to EGFR inhibition, with both cetuximab and panitumumab (a fully humanized mAb also inhibiting EGFR) capable of generating this rash in patients. Initially, research focused on candidate genetic variants related to EGFR (13–15). However, the clinical utility of such associations are complicated by the finding that rash severity is associated with cetuximab efficacy (1, 16–18). Thus, individuals with the highest risk of rash based on EGFR polymorphisms may also receive the most antitumor benefit from cetuximab therapy. Genome-wide association studies (GWAS) have been successful in identifying novel genetic markers of drug toxicity due to their agnostic and comprehensive approach (19). This approach provides the best opportunity to identify targetable markers of cetuximab-induced skin toxicity that do not impact drug efficacy. While one such GWAS has been conducted for EGFR toxicity, the power and sample size were limited and no variants reached genome-wide significance (20). The goal of our study was to use a large population to agnostically identify germline genetic variants associated with cetuximab-induced severe skin toxicity among patients with stage III colon cancer.

Study population and design

This study included data from the N0147 trial, a NCI-sponsored phase III multicenter randomized clinical trial led by the North Central Cancer Treatment Group (NCCTG). NCCTG is now part of the Alliance for Clinical Trials in Oncology. The details of this efficacy trial have been described previously (21). Briefly, participants with resected stage III colon cancer were recruited from clinical institutions across North America and randomly assigned to a variety of treatment groups. This analysis is limited to those patients who received adjuvant fluorouracil, leucovorin, and oxaliplatin (FOLFOX) every 2 weeks along with weekly cetuximab for a total of 12 cycles, as tolerated. All participants provided written informed consent and the study was approved by the institutional review board of all participating centers.

Classification of adverse events

Detailed information on adverse events was collected by clinicians at each study institution and graded according to the NCI Common Toxicity Criteria for Adverse Events (CTCAE), version 3.0 (22) prior to each chemotherapy dose. Toxicity grade was defined as the maximum grade experienced throughout the treatment period and subsequently dichotomized as grade 0–2 versus grade ≥3 to focus on severe skin toxicity. Skin toxicity was captured as a combined variable including desquamation and acneiform rash.

Genotyping and quality control

DNA was extracted from whole blood collected at study recruitment (n = 1,237 of 1,349 randomized to the cetuximab with FOLFOX treatment arm). As previously described (23), genotyping was performed at the Center for Inherited Disease Research at Johns Hopkins University (Baltimore, MD) using the Illumina HumanOmniExpress + Exome array (HumanOmniExpressExome-8v1–2, BPM annotation version A, genome build GRCh37/hg19), which consisted of a total of 964,193 genetic variants. We included 133 HapMap samples as genotyping controls. Standard quality control (QC) for genetic variants included removing variants with call rates <98%, discordance with technical duplicates, Mendelian errors, and sex differences in allele frequencies and heterozygosity. A total of 873,829 variants passed quality control (QC). Individual-level QC included removing individuals with <98% call rate, chromosomal anomalies, discordance in self-reported versus genetic sex, and relatedness. A total of 1,209 individuals (98%) passed quality control.

Population structure and imputation

Principal components analysis was implemented using PLINK (v1.9) to investigate population structure. The first seven eigenvectors discriminated individuals based on self-identified race and explained 82% of the genetic variation; these were used as covariates in analysis.

We imputed genotypes to infer unobserved genotypes and increase the genetic variant density. Samples were phased using SHAPEIT2 (24) and imputed using IMPUTE2 (25, 26), with the 1000 Genomes Project phase 3 (27) as the imputation reference panel. After imputation, we converted genotype probabilities to allelic dosages. Poorly imputed variants (info metric < 0.3) and variants with a minor allele frequency less than 1% were excluded. A total of 10,574,903 directly genotyped and imputed variants were included in the final analysis. Only autosomal chromosomes were analyzed.

Statistical analysis

Descriptive statistics comparing demographic variables by skin toxicity were calculated using χ2 tests or t tests, as appropriate. Logistic regression was used to calculate ORs and 95% confidence intervals (CI) for the association of each genetic variant and severe skin toxicity, adjusting for age at diagnosis (continuous), sex, and the first seven eigenvectors of genetic ancestry. Genetic variants were modeled using the log-additive approach, relating genotype dosage (the expected number of risk allele copies) to development of severe skin toxicity. Quantile-quantile (QQ) plots were produced with 95% CIs based on the null distribution of observed P values, as well as genomic control coefficients to assess for possible systemic inflation and bias (28). We produced Manhattan plots and used a threshold of P < 5 × 10−8 to denote genome-wide significance. Analyses were performed using R version 3.5.2 (29).

In silico genomic follow-up

In silico bioinformatics analysis was performed for loci that reached genome-wide significance. The NCI “LDassoc” web tool (https://ldlink.nci.nih.gov) was used to nominate candidate causal variants at each locus by selecting the most statistically significant variant (the lead variant) and all variants in linkage disequilibrium (LD; defined as R2 ≥ 0.8 in 1000 Genomes Phase 3 “EUR” population; ref. 27) with the lead variant. HaploReg v4.1 was then used to evaluate the functional annotation and regulatory chromatin states in different cell lines for each locus variant (30). Using SNP NEXUS (31), locus variants were annotated with genome-wide functional prediction scores [CADD (ref. 32; Combined Annotation Dependent Depletion) and Eigen-PC; ref. 33], prioritizing variants with CADD phred score ≥ 20 or Eigen-PC phred scores ≥ 17. Using the University of California Santa Cruz (UCSC, Santa Cruz, CA) genome browser (http://genome.ucsc.edu/; ref. 34), we visually inspected whether any of the locus variants mapped to (i) regions with predicted regulatory function using the GeneHancer (35) trackhub or (ii) DNaseI hypersensitivity or transcription factor ChIP-seq (chromatin immunoprecipitation followed by sequencing) clusters based on the ENCODE (36, 37) trackhub. Tissue-specific gene expression and overlap of locus variants with expression Quantitative Trait Loci (eQTLs) were evaluated using GTEx data (https://www.gtexportal.org). Tissue-specific gene expression at the mRNA and protein level were further examined using Human Protein Atlas data (https://www.proteinatlas.org/humanproteome/tissue; ref. 38).

Baseline demographic and clinical characteristics of the 1,209 patients with stage III colon cancer fulfilling the eligibility criteria for this study are shown in Table 1. Overall, 243 individuals (20%) had severe (grade 3–4) skin toxicities. The majority of these were grade 3, with only 7 individuals experiencing grade 4 skin toxicities. Compared with patients without severe skin toxicity, individuals with severe skin toxicity were younger (mean age 55 vs. 59 years, P < 0.001), more likely to be male (61% vs. 50%, P = 0.002), and less likely to be underweight (21% vs. 30%, P = 0.032).

The Manhattan plot for the genome-wide analysis is shown in Fig. 1 (QQ plot: Supplementary Fig. S1). Variants on chromosomes 2 and 17 reached genome-wide significance (P < 5 × 10−8; Table 2). The peak on chromosome 17 contained three variants in strong LD (R2 > 0.8), located at 38,433,225–38,470,142 bp (genome build GRCh37/hg19). The lead variant (rs78201730: OR, 3.98; 95% CI, 2.50–6.33; P = 5.6 × 10−9) was within the intronic region of the WIPF2 gene (WAS/WASL-interacting protein family member 2). The other two variants (rs117011100: P = 7.76 × 10−9; rs74453681: P = 7.73 × 10−9; both OR, 3.93; 95% CI, 2.47–6.25) were within the intronic region of the RARA gene (retinoic acid receptor alpha).

The peak on chromosome 2 contained two neighboring intergenic variants (rs7601541 and rs7562256 at 119,942,096–119,942,097bp) in complete LD with each other (R2 = 1.0) (Fig. 2). These were rare variants, with a 5% minor allele frequency (MAF) among affected individuals (OR, 6.78; 95% CI, 3.49–13.15; P = 1.54 × 10−8). Five additional intergenic variants at 119,942,324–119,946,838bp were in LD with the lead variants (R2 > 0.8) but did not reach genome-wide significance.

In silico genomic annotation

On chromosome 17, the RARA intronic variants (rs117011100 and rs74453681) had higher predicted importance (CADD phred score 20.9 and 12.8, respectively) than the WIPF2 intronic variant (rs78201730; CADD phred score 0.04; Supplementary Table S1). According to HaploReg, these variants overlapped with active enhancer (H3K27ac) and promoter (H3K9ac) histone marks in multiple cell lines, including skin (enhancer only). GeneHancer functional annotations indicated both RARA variants are in a promoter. In addition, these variants are located in regions consistent with transcription factor–binding sites and with active regulatory elements for several genes. Evaluation of gene expression and tissue specificity using GTEx and the Human Protein Atlas showed that both RARA and WIPF2 have widespread expression and low tissue specificity, with moderate expression in the skin.

All variants on the chromosome 2 locus were intergenic with low predicted importance (CADD phred score < 1) with the exception of rs7557444 (CADD phred score 15.5; Supplementary Table S1). None of the variants were in active promoters or enhancers for skin cell lines according to HaploReg. Minimal functional annotations were identified using GeneHancer and eQTL data were not available for these loci. ENCODE annotations suggested rs7557444 may be in a transcription factor motif for FOS and MYC, two proto-oncogenes.

Our GWAS identified novel genetic loci within the intronic regions of WIPF2 and RARA that reached genome-wide significance for the association of severe skin toxicity in patients receiving cetuximab for stage III colon cancer treated within the NCCTG (Alliance) N0147 clinical trial. Intergenic variants on chromosome 2 also reached genome-wide significance.

Factors underlying cetuximab-induced skin toxicity occurrence and severity are poorly understood. It is believed that skin toxicity is caused by direct inhibition of EGFR in the skin, as EGFR is normally expressed in epidermal keratinocytes (39). To minimize dose-limiting toxicities while maintaining maximal drug efficacy, management of cetuximab-induced skin toxicity involves treatment with corticosteroids and/or antibiotics, escalating from topical to systemic based on rash severity (4, 5). Recommendations include cetuximab dose reduction or interruption with grade ≥3 rash. Recent randomized clinical trials have shown prophylactic therapy with systemic tetracycline antibiotics, prophylactic use of sunscreens, and low potency topical steroids can reduce rash occurrence and severity (40, 41). However, because this nonspecific approach can be associated with substantive adverse sequelae, it would be beneficial to identify individuals at higher risk of severe rash and tailor prophylactic therapy accordingly. Prior studies have identified EGFR polymorphisms (13–16), gene copy-number variants (18), and number of CA repeats within the EGFR gene are associated with skin toxicity, but this is not currently applied to clinical practice.

Rash severity is strongly associated with cetuximab efficacy (1, 16–18), highlighting a need to identify predictors of rash that can be targeted through alternative pathways. The agnostic GWAS approach has previously been successful in identifying genetic variants involved in chemotherapeutic toxicity for many cancer types, identifying potentially targetable mechanistic and regulatory pathways (19). With only 282 patients with colon cancer, the only prior GWAS to evaluate germline variants predictive of cetuximab-induced skin toxicity did not identify any variants reaching genome-wide significance (20). Ours is the first investigation to identify genetic variants associated with cetuximab-induced severe skin toxicity that are unrelated to EGFR.

On the basis of CADD and functional annotation, the two RARA variants (rs117011100 and rs74453681) have biologically plausible effect. The RARA gene is responsible for producing retinoic acid receptor alpha (RARα), a transcription factor important in retinoid signaling (42). Both the retinoid isotretinoin and topical retinoids, such as adapalene, tazarotene, and tretinoin, have high clinical efficacy for the treatment of acne vulgaris (43). While cetuximab-associated skin toxicity appears visually similar to acne vulgaris, it is histopathologically distinct (44). However, a few case reports have evaluated the treatment of cetuximab-induced skin toxicity with either systemic or topical retinoids (45–49). In general, these reports have shown promising results, with substantial reduction of skin lesions without requiring cetuximab dose reduction, albeit with limited sample sizes. There has been no apparent effect on cetuximab efficacy, but rigorous assessment was not conducted. However, a randomized clinical trial of prophylactic tazarotene in patients with stage IV colorectal cancer receiving cetuximab found no observed clinical benefit and noted 33% of patients discontinued therapy due to significant skin irritation (8). This could be due to differences in formulation, dosing (once vs twice daily), or prophylactic versus reactive use. More research is needed to determine which patients undergoing cetuximab treatment may benefit from retinoid therapy and to establish the most effective formulation and dosing regimen. Currently, low-dose isotretinoin is only recommended for refractory grade ≥3 rash (12).

The mechanism of action of isotretinoin is unknown. While it does not directly bind RARs, it may act as a prodrug that is converted to metabolites that act as agonists for retinoic acid nuclear receptors (50). Adapalene and tazarotene are retinoid prodrugs which bind to all three members of the retinoic acid receptor family (α, β, and γ), but appear to have selectivity for RARβ and RARγ (51, 52). It is unclear how the identified RARA variants may influence rash severity and whether these variants influence response to retinoid therapy. Because of the design of GWAS platforms, the variants identified are likely surrogates for the causal variants and additional sequencing is warranted to gain a detailed understanding of the locus. However, it is plausible that the minority of patients with these variants might benefit from retinoid therapy. Further work is needed to identify the causal variants, determine the utility of retinoids in patients with these germline variants, and ensure there is no effect of retinoid therapy on cetuximab efficacy.

On the basis of the low overall minor allele frequency and minimal predicted biologic effect using in silico bioinformatics, we have less confidence in the significance of our GWAS findings associated with the chromosome 2 locus (rs7601541 and rs7562256). These variants are more common in African populations (MAF 9% vs. 2% in European based on 1000 Genomes phase 3; ref. 27) and our study population was primarily of European descent. Validation of this finding in another ethnically diverse population and in larger sample sizes could help differentiate whether this may be a spurious finding or instead a biologically important rare variant not well represented in our current trial sample set.

To date, this is the largest GWAS evaluating variants associated with cetuximab-induced skin toxicity. In addition to the large sample size, strengths of our study include standardized treatment protocols, uniform assessment of skin toxicity, and controlled treatment administration. Our study also has some limitations. Our analysis was unable to account for the timing of skin toxicity, dose cycle at which the greatest toxicity was reported, total number of skin toxicities experienced, or dose density of cetuximab. Despite randomization, we noted some differences in the age and sex of those experiencing severe skin toxicity, so these variables were adjusted for in our analysis. We also noted some differences in BMI for those experiencing severe skin toxicity but were unable to account for this in our analysis due to low sample sizes within each BMI category. Further research is needed to determine whether BMI influences the observed association between RARA variants and severe skin toxicity, as some laboratory research has shown a link between RARs and obesity (53). We had limited racial and ethnic diversity in our study population and did not assess patients treated with alternate systemic anti-EGFR antibodies (i.e., panitumumab), which may restrict generalizability of our findings.

In conclusion, we identified novel loci in the RARA gene that were associated with severe cetuximab-induced skin toxicity. This finding could represent a potential therapeutic target for prophylactic or reactive treatment in a subset of patients undergoing treatment with this agent. If validated, a precision medicine strategy using these variants could risk stratify cancer patients undergoing cetuximab treatment to direct management of skin toxicity.

Q. Shi reports other from Amgen, Merck & Co., and Johnson & Johnson; personal fees from Yiviva Inc.; grants from Celgene and Roche/Genentech; and personal fees from Boehringer Ingelheim Pharmaceuticals, Inc outside the submitted work. G. Yothers reports grants from NCI during the conduct of the study. F.A. Sinicrope reports a patent for Roche Tissue Diagnostics issued and with royalties paid. R.M. Goldberg reports grants from NCI during the conduct of the study and personal fees from Amgen, Bayer, AstraZeneca, Merck, Novartis, Genentech, and Taiho outside the submitted work. T.J. George reports other from BMS, Merck, AstraZeneca, Genentech, Tesaro/GSK, Ipsen, and Seattle Genetics outside the submitted work. A.T. Chan reports grants and personal fees from Bayer Pharma AG and personal fees from Pfizer Inc. and Boeringer Ingelheim outside the submitted work. No disclosures were reported by the other authors.

The content of this manuscript does not necessarily reflect the views or policies of the NCI or authors' affiliated institutions.

J.D Labadie: Conceptualization, formal analysis, visualization, methodology, writing–original draft, writing–review and editing. X. Hua: Conceptualization, data curation, methodology, writing–review and editing. T.A. Harrison: Methodology, project administration, writing–review and editing. B.L. Banbury: Data curation, methodology, writing–review and editing. J.R. Huyghe: Methodology, writing–review and editing. W. Sun: Resources, methodology, writing–review and editing. Q. Shi: Resources, data curation, project administration, writing–review and editing. G. Yothers: Data curation, funding acquisition, writing–review and editing. S.R. Alberts: Data curation, funding acquisition, writing–review and editing. F.A. Sinicrope: Data curation, funding acquisition, writing–review and editing. R.M. Goldberg: Data curation, funding acquisition, writing–review and editing. T.J. George: Data curation, funding acquisition, writing–review and editing. K.L. Penney: Methodology, writing–review and editing. A.I. Phipps: Methodology, writing–review and editing. S.A. Cohen: Investigation, writing–review and editing. U. Peters: Resources, supervision, funding acquisition, writing–review and editing. A.T. Chan: Funding acquisition, writing–review and editing. P.A. Newcomb: Resources, supervision, funding acquisition, writing–original draft, writing–review and editing.

The authors thank all study participants and staff of the NCCTG N0147 (Alliance) clinical trial. This work was supported by the NCI of the NIH under Award Numbers U10CA180821, U10CA180882, and U24CA196171 (to the Alliance for Clinical Trials in Oncology); UG1CA233180 and UG1CA233339 (https://acknowledgments.alliancefound.org); UG1CA189858 (Southeast Clinical Oncology Research Consortium; and R01CA176272 (to P.A. Newcomb and A.T. Chan). It was also supported in part by funds from Genentech, Sanofi, Pfizer, and Eli Lilly and Company. Fred Hutchinson Cancer Research Center investigators were also supported by NIH T32 CA094880 and NIH K05 CA152715 (to J.D. Labadie and P.A. Newcomb).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Baas
JM
,
Krens
LL
,
Guchelaar
H-J
,
Ouwerkerk
J
,
Jong
FAD
,
Lavrijsen
APM
, et al
Recommendations on management of EGFR inhibitor-induced skin toxicity: a systematic review
.
Cancer Treat Rev
2012
;
38
:
505
14
.
2.
Fabbrocini
G
,
Panariello
L
,
Caro
G
,
Cacciapuoti
S
. 
Acneiform rash induced by EGFR inhibitors: review of the literature and new insights
.
Skin Appendage Disord
2015
;
1
:
31
7
.
3.
Chanprapaph
K
,
Vachiramon
V
,
Rattanakaemakorn
P
. 
Epidermal growth factor receptor inhibitors: a review of cutaneous adverse events and management
.
Dermatol Res Pract
2014
;
2014
:
734249
.
4.
Eaby
B
,
Culkin
A
,
Lacouture
ME
. 
An interdisciplinary consensus on managing skin reactions associated with human epidermal growth factor receptor inhibitors
.
Clin J Oncol Nurs
2008
;
12
:
283
90
.
5.
Gutzmer
R
,
Becker
JC
,
Enk
A
,
Garbe
C
,
Hauschild
A
,
Leverkus
M
, et al
Management of cutaneous side effects of EGFR inhibitors: recommendations from a German expert panel for the primary treating physician
.
J Dtsch Dermatol Ges
2011
;
9
:
195
203
.
6.
Melosky
B
,
Burkes
R
,
Rayson
D
,
Alcindor
T
,
Shear
N
,
Lacouture
M
. 
Management of skin rash during EGFR-targeted monoclonal antibody treatment for gastrointestinal malignancies: Canadian recommendations
.
Curr Oncol
2009
;
16
:
16
26
.
7.
Lacouture
ME
,
Mitchell
EP
,
Piperdi
B
,
Pillai
MV
,
Shearer
H
,
Iannotti
N
, et al
Skin toxicity evaluation protocol with panitumumab (STEPP), a phase II, open-label, randomized trial evaluating the impact of a pre-Emptive Skin treatment regimen on skin toxicities and quality of life in patients with metastatic colorectal cancer
.
J Clin Oncol
2010
;
28
:
1351
7
.
8.
Scope
A
,
Agero
ALC
,
Dusza
SW
,
Myskowski
PL
,
Lieb
JA
,
Saltz
L
, et al
Randomized double-blind trial of prophylactic oral minocycline and topical tazarotene for cetuximab-associated acne-like eruption
.
J Clin Oncol
2007
;
25
:
5390
6
.
9.
Joshi
SS
,
Ortiz
S
,
Witherspoon
JN
,
Rademaker
A
,
West
DP
,
Anderson
R
, et al
Effects of epidermal growth factor receptor inhibitor-induced dermatologic toxicities on quality of life
.
Cancer
2010
;
116
:
3916
23
.
10.
Wagner
LI
,
Lacouture
ME
. 
Dermatologic toxicities associated with EGFR inhibitors: the clinical psychologist's perspective. Impact on health-related quality of life and implications for clinical management of psychological sequelae
.
Oncology
2007
;
21
:
34
6
.
11.
Li
T
,
Perez-Soler
R
. 
Skin toxicities associated with epidermal growth factor receptor inhibitors
.
Target Oncol
2009
;
4
:
107
19
.
12.
Lacouture
ME
,
Anadkat
MJ
,
Bensadoun
R-J
,
Bryce
J
,
Chan
A
,
Epstein
JB
, et al
Clinical practice guidelines for the prevention and treatment of EGFR inhibitor-associated dermatologic toxicities
.
Support Care Cancer
2011
;
19
:
1079
95
.
13.
Pander
J
,
Gelderblom
H
,
Antonini
NF
,
Tol
J
,
Krieken
JHJMV
,
Straaten
TVD
, et al
Correlation of FCGR3A and EGFR germline polymorphisms with the efficacy of cetuximab in KRAS wild-type metastatic colorectal cancer
.
Eur J Cancer
2010
;
46
:
1829
34
.
14.
Saito
R
,
Suzuki
H
,
Yamada
T
,
Endo
S
,
Moriwaki
T
,
Ueno
T
, et al
Predicting skin toxicity according to EGFR polymorphisms in patients with colorectal cancer receiving antibody against EGFR
.
Anticancer Res
2013
;
33
:
4995
8
.
15.
Froelich
MF
,
Stintzing
S
,
Kumbrink
J
,
Grünewald
TGP
,
Mansmann
U
,
Heinemann
V
, et al
The DNA-polymorphism rs849142 is associated with skin toxicity induced by targeted anti-EGFR therapy using cetuximab
.
Oncotarget
2018
;
9
:
30279
88
.
16.
Klinghammer
K
,
Knodler
M
,
Schmittel
A
,
Budach
V
,
Keilholz
U
,
Tinhofer
I
. 
Association of epidermal growth factor receptor polymorphism, skin toxicity, and outcome in patients with squamous cell carcinoma of the head and neck receiving cetuximab-docetaxel treatment
.
Clin Cancer Res
2010
;
16
:
304
10
.
17.
Vallböhmer
D
,
Zhang
Wu
,
Gordon
M
,
Yang
DY
,
Yun
J
,
Press
OA
, et al
Molecular determinants of cetuximab efficacy
.
J Clin Oncol
2005
;
23
:
3536
44
.
18.
Wacker
B
,
Nagrani
T
,
Weinberg
J
,
Witt
K
,
Clark
G
,
Cagnoni
PJ
. 
Correlation between development of rash and efficacy in patients treated with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in two large phase III studies
.
Clin Cancer Res
2007
;
13
:
3913
21
.
19.
Harper
AR
,
Topol
EJ
. 
Pharmacogenomics in clinical practice and drug development
.
Nat Biotechnol
2012
;
30
:
1117
24
.
20.
Baas
J
,
Krens
L
,
Bohringer
S
,
Mol
L
,
Punt
C
,
Guchelaar
H-J
, et al
Genome wide association study to identify predictors for severe skin toxicity in colorectal cancer patients treated with cetuximab
.
PLoS One
2018
;
13
:
e0208080
.
21.
Alberts
SR
,
Sargent
DJ
,
Nair
S
,
Mahoney
MR
,
Mooney
M
,
Thibodeau
SN
, et al
Effect of oxaliplatin, fluorouracil, and leucovorin with or without cetuximab on survival among patients with resected stage III colon cancer: a randomized trial
.
JAMA
2012
;
307
:
1383
93
.
22.
Trotti
A
,
Colevas
A
,
Setser
A
,
Rusch
V
,
Jaques
D
,
Budach
V
, et al
CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment
.
Semin Radiat Oncol
2003
;
13
:
176
81
.
23.
Penney
KL
,
Banbury
BL
,
Bien
S
,
Harrison
TA
,
Hua
X
,
Phipps
AI
, et al
Genetic variant associated with survival of patients with stage II-III colon cancer
.
Clin Gastroenterol Hepatol
2020
;
18
:
2717
23
.
24.
Delaneau
O
,
Marchini
J
,
1000 Genomes Project Consortium
. 
Integrating sequence and array data to create an improved 1000 Genomes Project haplotype reference panel
.
Nat Commun
2014
;
5
:
3934
.
25.
Howie
B
,
Marchini
J
,
Stephens
M
. 
Genotype imputation with thousands of genomes
.
G3
2011
;
1
:
457
70
.
26.
Howie
BN
,
Donnelly
P
,
Marchini
J
. 
A flexible and accurate genotype imputation method for the next generation of genome-wide association studies
.
PLos Genet
2009
;
5
:
e1000529
.
27.
1000 Genomes Project Consortium
,
Auton
A
,
Brooks
LD
,
Durbin
RM
,
Garrison
EP
,
Kang
HM
, et al
A global reference for human genetic variation
.
Nature
2015
;
526
:
68
74
.
28.
Devlin
B
,
Roeder
K
,
LJTpb
W
. 
Genomic control, a new approach to genetic-based association studies
. 
2001
;
60
:
155
66
.
29.
R: A Language and Environment for Statistical Computing [computer program]
.
Vienna, Austria
:
R Foundation for Statistical Computing
; 
2018
.
30.
Ward
LD
,
Kellis
M
HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants
.
Nucleic Acids Res
2012
;
40
:
D930
934
.
31.
Dayem Ullah
AZ
,
Oscanoa
J
,
Wang
J
,
Nagano
A
,
Lemoine
NR
,
Chelala
C
. 
SNPnexus: assessing the functional relevance of genetic variation to facilitate the promise of precision medicine
.
Nucleic Acids Res
2018
;
46
:
W109
W113
.
32.
Kircher
M
,
Witten
DM
,
Jain
P
,
O'Roak
BJ
,
Cooper
GM
,
Shendure
J
. 
A general framework for estimating the relative pathogenicity of human genetic variants
.
Nat Genet
2014
;
46
:
310
15
.
33.
Ionita-Laza
I
,
McCallum
K
,
Xu
B
,
Buxbaum
JD
. 
A spectral approach integrating functional genomic annotations for coding and noncoding variants
.
Nat Genet
2016
;
48
:
214
20
.
34.
Kent
WJ
,
Sugnet
CW
,
Furey
TS
,
Roskin
KM
,
Pringle
TH
,
Zahler
AM
, et al
The human genome browser at UCSC
.
Genome Res
2002
;
12
:
996
1006
.
35.
Fishilevich
S
,
Nudel
R
,
Rappaport
N
,
Hadar
R
,
Plaschkes
I
,
Iny Stein
T
, et al
GeneHancer: genome-wide integration of enhancers and target genes in GeneCards
.
Database
2017
;
2017
:
bax028
.
36.
Rosenbloom
KR
,
Sloan
CA
,
Malladi
VS
,
Dreszer
TR
,
Learned
K
,
Kirkup
VM
, et al
ENCODE data in the UCSC genome browser: year 5 update
.
Nucleic Acids Res
2013
;
41
:
D56
63
.
37.
Consortium
EP
. 
A user's guide to the encyclopedia of DNA elements (ENCODE)
.
PLoS Biol
2011
;
9
:
e1001046
.
38.
Uhlen
M
,
Fagerberg
L
,
Hallstrom
BM
,
Lindskog
C
,
Oksvold
P
,
Mardinoglu
A
, et al
Proteomics. Tissue-based map of the human proteome
.
Science
2015
;
347
:
1260419
.
39.
Lichtenberger
BM
,
Gerber
PA
,
Holcmann
M
,
Buhren
BA
,
Amberg
N
,
Smolle
V
, et al
Epidermal EGFR controls cutaneous host defense and prevents inflammation
.
Sci Transl Med
2013
;
5
:
199ra111
.
40.
Petrelli
F
,
Borgonovo
K
,
Cabiddu
M
,
Coinu
A
,
Ghilardi
M
,
Lonati
V
, et al
Antibiotic prophylaxis for skin toxicity induced by antiepidermal growth factor receptor agents: a systematic review and meta-analysis
.
Br J Dermatol
2016
;
175
:
1166
74
.
41.
Bachet
J‐B
,
Peuvrel
L
,
Bachmeyer
C
,
Reguiai
Z
,
Gourraud
PA
,
Bouché
O
, et al
Folliculitis induced by EGFR inhibitors, preventive and curative efficacy of tetracyclines in the management and incidence rates according to the type of EGFR inhibitor administered: a systematic literature review
.
Oncologist
2012
;
17
:
555
68
.
42.
di Masi
A
,
Leboffe
L
,
De Marinis
E
,
Pagano
F
,
Cicconi
L
,
Rochette-Egly
C
, et al
Retinoic acid receptors: from molecular mechanisms to cancer therapy
.
Mol Aspects Med
2015
;
41
:
1
115
.
43.
Kolli
SS
,
Pecone
D
,
Pona
A
,
Cline
A
,
Feldman
SR
. 
Topical retinoids in acne vulgaris: a systematic review
.
Am J Clin Dermatol
2019
;
20
:
345
65
.
44.
Lacouture
ME
. 
Mechanisms of cutaneous toxicities to EGFR inhibitors
.
Nat Rev Cancer
2006
;
6
:
803
12
.
45.
Gutzmer
R
,
Werfel
T
,
Mao
R
,
Kapp
A
,
Elsner
J
. 
Successful treatment with oral isotretinoin of acneiform skin lesions associated with cetuximab therapy
.
Br J Dermatol
2005
;
153
:
849
51
.
46.
Bidoli
P
,
Cortinovis
DL
,
Colombo
I
,
Crippa
A
,
Cicchiello
F
,
Villa
F
, et al
Isotretinoin plus clindamycin seem highly effective against severe erlotinib-induced skin rash in advanced non-small cell lung cancer
.
J Thorac Oncol
2010
;
5
:
1662
3
.
47.
DeWitt
CA
,
Siroy
AE
,
Stone
SP
. 
Acneiform eruptions associated with epidermal growth factor receptor-targeted chemotherapy
.
J Am Acad Dermatol
2007
;
56
:
500
5
.
48.
Vezzoli
P
,
Marzano
Av
,
Onida
F
,
Alessi
E
,
Galassi
B
,
Tomirotti
M
, et al
Cetuximab-induced acneiform eruption and the response to isotretinoin
.
Acta Derm Venereol
2008
;
88
:
84
6
.
49.
Andrews
ED
,
Garg
N
,
Patel
AB
. 
A retrospective chart review on oral retinoids as a treatment for epidermal growth factor receptor inhibitor and mitogen-activated protein kinase kinase inhibitor induced acneiform eruptions
.
J Am Acad Dermatol
2020
;
82
:
998
1000
.
50.
Layton
A
. 
The use of isotretinoin in acne
.
Dermatoendocrinol
2009
;
1
:
162
9
.
51.
Michel
S
,
Jomard
A
,
Demarchez
M
. 
Pharmacology of adapalene
.
Br J Dermatol
1998
;
139
:
3
7
.
52.
Gaikwad
J
,
Sharma
S
,
Hatware
KV
. 
Review on characteristics and analytical methods of tazarotene: an update
.
Crit Rev Anal Chem
2020
;
50
:
90
6
.
53.
Wolf
G
. 
Retinoic acid activation of peroxisome proliferation-activated receptor delta represses obesity and insulin resistance
.
Nutr Rev
2010
;
68
:
67
70
.