Side effects of tyrosine kinase inhibitors therapy in patients with non‑small cell lung cancer and associations with EGFR polymorphisms: A systematic review and meta‑analysis

  • Authors:
    • Jasmina Obradovic
    • Jovana Todosijevic
    • Vladimir Jurisic
  • View Affiliations

  • Published online on: December 23, 2022     https://doi.org/10.3892/ol.2022.13649
  • Article Number: 62
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rash and diarrhea are common side effects of tyrosine kinase inhibitor (TKI) therapy administered to patients with non‑small cell lung cancer (NSCLC). The polymorphisms of the epidermal growth factor receptor (EGFR) gene may be a potential predictor of these side effects. The aim of the present meta‑analysis was to examine the association of EGFR polymorphisms and TKI‑associated toxicities. Electronic databases (PubMed, Scopus and ISI Web of Science) were searched for relevant studies. According to the inclusion and exclusion criteria, a search of the databases identified 4,918 results, among which 6 clinical trials were obtained with 1,318 patients with NSCLC. A total of 9 EGFR single nucleotide polymorphisms (SNPs) associated with TKI toxicity were identified including, rs11568315, rs712829, rs712830, rs2227983, rs2075102, rs2293347, rs11977388, rs4947492 and rs884225. The data associated with skin toxicity from rs11568315, rs712829 and rs712830 were analyzed in the present meta‑analysis. Data from rs11568315 were also analyzed in relation to diarrhea. Among all the examined SNPs, statistically significant results were obtained under the dominant genetic model for CA repeats in rs11568315 (SS vs. SL+LL) with skin toxicity. The long CA repeat (SL+LL) carriers were more likely to experience skin toxicity associated with TKIs (P=0.005). By contrast, there was no significant result for diarrhea (P=0.661) under dominant genetic model for CA repeats.

Introduction

As the most prevalent form of lung cancer, non-small cell lung cancer (NSCLC) is reported to be one of the deadliest types of cancer in the world, with 2,206,771 newly diagnosed cases and 1,796,144 new deaths recorded in 2020 (1,2). In patients with advanced NSCLC, platinum-based chemotherapy is the first line treatment, but it is usually cytotoxic and has a short progression-free survival (PFS) time of 3–5 months and an overall survival (OS) time of ≤10 months (3). Targeted therapy has been developed to prevent epidermal growth factor receptor (EGFR) activation (36). EGFR is a transmembrane protein and a potent transducer of altered signals in tumor cells. There are two ways of blocking the EGFR: Either by blocking the ligand from binding to the receptor extracellular domain with anti-EGFR monoclonal antibodies (cetuximab) or by reversibly binding the small molecule tyrosine kinase inhibitors (TKIs) to the receptor intracellular tyrosine kinase domain (4,5). Thus, the introduction of the first TKI generation drugs, gefitinib and erlotinib, resulted in markedly higher treatment response rates (73.7% for TKI compared with 30.7% for chemotherapy), and the median PFS time increased to 10–13 months for patients with NSCLC (68).

For accurate therapeutic decisions to be made in the management of patients with NSCLC, it is essential to find molecular markers that can identify patients who will respond most effectively to treatment. Promising molecular identifiers include mutations in the EGFR gene. In patients carrying exon 19 deletions and point mutations in exon 21, a significant clinical benefit following treatment with TKIs was observed (9,10). However, acquired resistance in connection with EGFR T790 mutations limited the efficacy of the EGFR-TKI (11,12). The role of polymorphisms [including single nucleotide polymorphisms (SNPs) and short tandem CA repeats] of the EGFR gene as another potential molecular target that improved clinical outcomes is well-established (1316). Recently, a meta-analysis elucidated that among rs712829 (−216G>T), rs11568315 (CA repeat), rs2293347 (D994D) and rs4947492, −216G>T and variable CA repeat polymorphisms significantly affected OS and PFS time in gefitinib- or erlotinib-treated patients with NSCLC (17).

EGFR-TKI therapy is associated with side effects, primarily in the form of skin or gastrointestinal toxicities (e.g., skin rash or diarrhea). Although skin toxicities are not lethal or dose-limited, they frequently occur with EGFR-TKIs and affect patient quality of life (18). Among usual skin toxicities, such as xerosis, pruritus, paronychia, mucositis and increased growth of eyelashes or facial hair, skin rash is the most prevalent (1922). Notably, patients with NSCLC that develop skin rashes are better responders to EGFR-TKI therapy and have a longer median overall survival time (1823). EGFR SNPs have been examined in association with survival in NSCLC (17,18); they may provide insight into therapy outcomes, particularly the potential side effects associated with TKIs (2328). Literature analysis discovered notable inconsistency in previously published reports. While some studies found associations with EGFR genotypes and TKI toxicity (2326), others did not (27). Additionally, previous meta-analyses investigated EGFR mutations, but not EGFR polymorphisms and therapy side effects in patients with NSCLC (29,30), or toxicity in relation to radiotherapy (31). With regards to these discrepancies and the role of EGFR SNPs as potential determinants of treatment outcome, the aim of the present meta-analysis was to determine whether the molecular mechanisms involving EGFR SNPs were associated with EGFR-TKI therapy side effects.

Materials and methods

Search strategy and study selection

The present study was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines (PRISMA) (32). The systematic search for the relevant studies was performed using electronic databases, PubMed, Scopus and ISI Web of Science. Searches were performed considering EGFR polymorphisms and side effects of TKI therapy in patients with NSCLC. The search had the following retrieval strategy for the PubMed database: [(‘receptor, epidermal growth factor’ (MeSH Terms) OR EGFR (All Fields)) AND (gene(tiab) OR ‘polymorphism, genetic’(MeSH Terms)) AND (‘carcinoma, non-small-cell lung’ (MeSH Terms) OR NSCLC (All Fields)) AND (‘drug therapy’ (Subheading) OR treatment (All Fields) OR ‘erlotinib hydrochloride’ (MeSH Terms) OR ‘gefitinib’ (MeSH Terms) OR TKI OR ‘TK inhibitors’ OR ‘tyrosine kinase inhibitors’ OR ‘Tyrosine-kinase inhibitor’) AND response (All Fields)) OR Prognosis (MeSH)) OR toxic (MeSH)) OR toxicity (MeSH)) OR side effect (MeSH)) AND (humans (MeSH))]. The Scopus and ISI Web of Science databases were also searched with necessary modifications to the PubMed search query. The full search string is available from the corresponding author upon request. Finally, additional studies were searched for in the bibliographies of the selected eligible studies or reviews.

Selection criteria

All studies fulfilling the following inclusion criteria were eligible: i) Studies published from January 1, 2009, to February 13, 2019; ii) studies published in English; iii) studies involving human subjects; iv) patients >18 years old with histopathologically confirmed NSCLC who received EGFR-TKI therapy and v) clinical trials or observational studies that investigated associations between EGFR polymorphisms and any side effects of TKI therapy. In the systematic review, studies were excluded based on the following criteria: i) Meta-analyses, editorials, letters, commentaries, systematic or narrative reviews; ii) not in the English language; iii) duplicate publications or studies involving animal or cell experimental models; iv) studies investigating EGFR polymorphisms and TKI adverse effects but not reporting their associations; v) single study reports of EGFR polymorphisms associated with TKI toxicities (skin toxicity or diarrhea), or other side effects (such as hepatotoxicity) due to being unable to make comparisons due to the lack of data from other studies and vi) randomized control trial (RCT) studies that did not report genotype numbers data, even though the odds ratio (OR) was reported.

Data extraction

Extracted studies from the electronic databases were first merged and duplicates were removed. A total of 2 authors (JO and JT) independently performed a manual search of titles and abstracts of potentially eligible studies according to the inclusion and exclusion criteria. Any discrepancies were resolved by discussion or by consulting the third author (VJ). Finally, the following data were extracted from the full texts based on the prior determined datasheet: The first author, year of publication, country, study type, study period, number of patients, median age, sex and ethnicity of patients, percentage of smokers, clinical stage, histology, median follow-up (in months), TKI treatment dosage, additional therapy, toxicity assessment, adverse effects of treatment, available EGFR genotype, variant location, SNP database identifier and number of patients/genotype.

Quality assessment

The Newcastle-Ottawa Quality Assessment Scale (NOS) (33) for cohort studies and the Jadad Scale for RCTs (34) were used to assess the methodological quality of the studies included. For the NOS scale, the overall maximum quality score was 9 points; for the Jadad Scale, the score was 5 points. The reviewers (JO and JT) independently evaluated the quality of the studies with discrepancies resolved by consensus.

Statistical analysis

When ≥2 studies had available EGFR polymorphic genotypes associated with TKI therapy side effects, meta-analysis was conducted. To examine heterogeneity between the eligible studies, Cochran's Q statistics and I2 statistics were applied. I2 was interpreted as follows: 0, no heterogeneity; 25, low heterogeneity; 50, moderate heterogeneity and 75%, high heterogeneity (35). The random effect model was used when there was significant heterogeneity between studies (P<0.05; I2>50%), otherwise, the fixed effect model was applied (36). Galbraith's plot was used to identify potential sources of heterogeneity (37). If heterogeneity was present, subgroup analyses of OR were conducted according to the available EGFR SNPs. The dominant genetic model (wild-type homozygote vs. heterozygote + mutant homozygote) of all three EGFR SNPs (rs11568315, rs712829 and rs712830) was used to calculate OR. The available adverse effects for the analysis were skin toxicity (skin rash) and gastrointestinal toxicity (diarrhea). For comparison, the adverse effects were combined and used as any grade vs. the absence of adverse effects. Sensitivity analysis was also performed to determine whether the results would be affected by excluding the study with the smallest sample size. The publication bias of the enrolled studies was tested with Begg's and Egger's tests, as well as funnel plots (38,39). P<0.05 was considered to indicate a statistically significant difference. STATA software package v.15 (StataCorp LP) was used for all statistical analysis.

Results

Study selection

The initial search of databases identified 4,918 results (PubMed, 881; ISI Web of Science, 395; Scopus, 3,642; Fig. 1). An additional study was included after reading the bibliographies of the full-text articles. After merging into the single datasheet and removing duplicates, 4,036 studies remained. Of these, 3,980 were excluded and 56 full-text articles were used to assess eligibility. Of these 56 articles, 50 were excluded due to not fulfilling the inclusion criteria. Finally, 6 clinical trials were included in the systematic review which contained 1,318 patients with NSCLC. A total of 4 studies were included in the meta-analysis.

Characteristics of the studies

The six studies from the search included four cohort studies (23,25,26,28) containing 316 patients and 2 RCTs (24,27) containing 1,002 patients. The studies were published from 2009–2017, with sample sizes ranging from 52–760 patients. A total of two studies were from Asia (Taiwan and China), two from Europe (Germany and Italy), one from Canada and one RCT was from a consortium of counties (Canada, Italy, South Korea and Brazil). The number of male patients in the studies was 33–67%. The percentage of smokers was 12–76%, while the median age was 56–68 years. Most of the patients had adenocarcinoma histology and were in clinical stages IV, IIIB and IIIA (2328). Only one study reported a median follow-up of 12 months (23). The EGFR-TKI therapy type for patients with NSCLC in all examined reports was gefitinib (250 mg/day) and erlotinib (150 mg/day), except in one study where cetuximab (250 or 500 mg/m2) or panitumumab (6 mg/kg) was prescribed (25). Additionally, four studies reported patients that had been previously treated with cisplatin (2427). Adverse effects were skin (rash) and gastrointestinal toxicity (diarrhea and hepatotoxicity). Toxicity assessment was conducted using the National Cancer Institute's Common Terminology Criteria for Adverse Events (40). The quality of the studies was rated acceptable using the NOS and the Jadad scale (33,34). Adequacy of follow-up was the lowest rated aspect. The characteristics and quality assessment of the included studies are presented in Table I.

Table I.

Characteristics of the included studies.

Table I.

Characteristics of the included studies.

First author, yearCountryStudy typeStudy periodPatients, nMedian age, years (range)Males, %Ethnicity, %Smokers, %Clinical stage (%)aHistology (%)Median follow-up, monthsTKI treatment (dose)Additional therapyToxicity assessmentAdverse effectOverall quality score(Refs.)
Huang et al, 2009TaiwanCohortMay 2005-September 1, 20065266 (39–86)33NRNRIIIB (14), IV (87)Adenoc-arcinoma (96), Other (4)12Gefitinib (250 mg/day)NRNCI CTCA E v3.0cSkinNOSh: 7(23)
Giovannetti et al, 2010ItalyCohortNR9664 (NR)57NR69IIIB (9), V (91)Adenoc-arcinoma (56), Other (44)NRGefitinib (250 mg/day)85% of patients were previously treated with chemot-herapybNCI CTC manual versiondSkin, diarrheaNOS: 7(26)
Liu et al, 2012CanadaRCT Phase IIINR24260 (NR)64East Asian (6), Other (94)76IIIB (NR), IV (NR)Adenoc-arcinoma (54), Other (46)NRErlotinib (NR)93% patients were previously treated with chemot-herapyNCI v2.0.eSkinJadadi: 4(24)
Parmar et al, 2013GermanyCohortSeptember 2008-November 201010968 (43–86)67NR12; 49.5 formerIIIA (2); IIIB (11), IV (83), Unknown (5)NRNRGefitinib (250 mg/day), erlotinib (150 mg/day), cetuximab (250 mg/m2, with weekly or biweekly or31% patients were previously treated with chemot-herapy panitumumab (6 mg/kg) biweeklyNCI CTC v3.0fSkinNOS: 6(25)
Kim et al, 2017Canada, Italy, South Korea and BrazilRCT Phase IIINR76062 (56–66)66Caucasian (96), East Asian (3), Other (1)26; 53 formerIIIB (11), IV (89)Adenoc-arcinoma (56), Other (44)NRErlotinib (NR)50% of patients were treated with erlotinib, followed by cisplatin and gemcita bine at progression; 50% were treated with cisplatin and gemcita bine, followed by erlotinib at progressionNRSkin, diarrheaJadad: 3(27)
Ma et al, 2017ChinaCohort2011-20145956 (31–77)49NR61IIIB or IV (64)Adenoc-arcinoma (90), Other (10)NRGefitinib (250 mg/day)NRCTCA E v4.0gSkin, diarrhea, hepat otoxicityNOS: 7(28)

{ label (or @symbol) needed for fn[@id='tfn1-ol-25-02-13649'] } Data is provided to 2 significant places.

a (2328),

b additional 127 chemotherapy-treated/gefitinib-non-treated patients with NSCLC were used as a comparison,

c (23),

d (26),

e (24),

f (25),

g (28),

h (33),

i (34). CTC, Common Toxicity Criteria; CTCAE, Common Terminology Criteria for Adverse Events; NCI, National Cancer Institute; NOS, Newcastle-Ottawa Quality Assessment Scale; NR, not reported; RCT, randomized clinical trial; TKI, tyrosine kinase inhibitor.

A total of nine EGFR SNPs (rs11568315, rs712829, rs712830, rs2227983, rs2075102, rs2293347, rs11977388, rs4947492 and rs884225) relative to TKI toxicity was identified in the literature search, which were provided by seven studies (2328,41). Of these, four studies reported exact numbers of patients/genotype of EGFR SNPs (rs11568315, rs712829 and rs712830) associated with TKI-caused toxicity and were included in quantitative synthesis (23,25,26,28). Genotypes for all EGFR SNPs for the meta-analysis were merged according to the dominant genetic model. The data for the EGFR SNPs genotype and skin toxicity, diarrhea or hepatotoxicity caused by EGFR TKI therapy are presented in Table II. Certain studies or sets of data were excluded from further analyses. The reasons are outlined below.

Table II.

EGFR genotypes and adverse effects of tyrosine kinase inhibitor therapy.

Table II.

EGFR genotypes and adverse effects of tyrosine kinase inhibitor therapy.

A, Patients with skin toxicity (n=880, 78.36%)

dbSNP-IDVariant type, location, and/or consequenceFirst author, yearGenotyping platform usedGenotypeTotal number of patients, nPatients with skin toxicity, nORa95% CIaza P-valuea(Refs.)
rs11568315Intron variant,Huang et al, 2009PCR andSS75Ref. 2.130.032(23)
g.55020560_5502 directSL2686.871.1734-706
0561AC[n] sequencingLL194540.2797
Parmar et al, 2013Real-TimeSS2219Ref. 1.230.217(25)
PCR andSL58412.270.6156-307
16-capillary electrophoresisLL292368.4150
or KASPar
Giovannetti et al, 2010TaqManSS2718Ref. 1.710.087(26)
PCRSL-LL60282.280.8863-002
575.8947
Ma et al, 2017SequenomSS66NANANANA(28)
MassarraySL2014
systemLL1610
Kim et al, 2017SangerSS3828Ref. 0.150.874(27)
sequencing andSL-LL96720.930.3961-807
Taqman PCR 2.1994
Liu et al, 2012PCR and direct sequencingLL-S-NRNR0.600.2–1.9NANA(24)
rs7128295′ UTR variant,Huang et al, 2009PCR and directGG4514Ref. 0.610.540(23)
g.5031G>T, - sequencingGT510.600.1186-206
216G>T TT22223.0567
Parmar et al, 2013Real-TimeGG4933Ref. 1.920.054(25)
PCR andGT48390.410.1670-009
16-capillary electrophoresisTT1211251.0188
or KASPar
Giovannetti et al, 2010TaqMan PCRGG3019Ref. 1.410.158(26)
GT-TT57271.910.7752-007
924.7513
Kim et al, 2017Sanger sequencing andGG4032Ref. 0.910.358(27)
Taqman PCRGT-TT83601.530.62–3.82902
Liu et al, 2012PCR and direct sequencingGT-GGNRNR2.000.5–8.3NANA(24)
rs7128305′ UTR variant,Parmar et al, 2013Real-TimeCC7959Ref. 0.580.561(25)
g.5056A>C, - PCR andCA30240.730.2637-007
191C>A 16-capillary electrophoresis or KASParAA00752.0624
Giovannetti et al, 2010TaqManCC7236Ref. 1.160.245(26)
PCRCA-AA15100.50.1554-201
1.6089
Kim et al, 2017Sanger sequencing andCC10072Ref. 1.440.147(27)
Taqman PCRCA- AA23200.380.1062-807
571.4007
Liu et al, 2012PCR and direct sequencingCA-CCNRNR1.000.1–6.8NANA(24)
rs2227983bMissense variant,Parmar et al, 2013Real-Time PCR andGG5749Ref. 2.450.014(25)
(1562G>A,16-capillary electrophoresisGA47293.241.2657-102
R497K) or KASParAA55268.3073
Giovannetti et al, 2010TaqManGG-GA75380.580.1585-0.790.425(26)
PCR 692.1734800
AA117Ref.
Huang et al,PCR andGG124Ref. 0.160.867(23)
2009directGA2881.120.2832-701
sequencingAA12554.4695

B, Patients with diarrhea (n=233, 20.75%)

Patients
Variant type, Totalwith
location, and/or number of diarrhea
dbSNP-ID consequenceFirst author, yearGenotyping platform used Genotypepatients, ntoxicity, nORa95% CIaza P-valuea(Refs.)

rs11568315Intron variant,Giovannetti et al, 2010TaqMan assaySS2613Ref. 0.760.446(26)
g.55020560_550 SS-LL49201.450.5569-106
20561AC[n] 3.7751
Ma et al, 2017SequenomSS62Ref. 0.500.612(28)
MassarraySL2090.620.1012-608
systemLL16753.8585
Kim et al, 2017Sanger sequencing andSS3815Ref. 0.120.902(27)
Taqman PCRSL-LL96390.950.4424-205
2.0535
rs7128295′ UTR variant,Kim et al, 2017Sanger sequencing andGG4018Ref. 0.940.346(27)
g.5031G>T, - Taqman PCRGT-TT83301.440.6711-106
216G>T 553.1131
rs7128305′ UTR variant,Kim et al, 2017Sanger sequencing andCC10037Ref. 0.950.339(27)
g.5056A>C, - Taqman PCRCA-AA23110.640.2570-504
191C>A 1.597
rs2227983bMissense variant,Giovannetti et al, 2010TaqMan PCRGG-GA74262.760.7163-1.470.139(26)
(1562G>A, 9210.7057608
R497K) AA106Ref.

C, Patients with hepatotoxicity (n=10, 0.89%)

Patients
Variant type, Totalwith
location, and/or Genotyping number of hepatoto
dbSNP-ID consequenceAuthor, yearplatform used Genotypepatients, nxicity, nORa95% CIaza P-valuea(Refs.)

rs1156Intron variant,Ma et al, 2017Sequenom MassarraySS61Ref. 0.400.682(28)
8315g.55020560_550 systemSL2160.620.0640-907
20561AC[n] LL163226.0507

a Pre-calculated values according to available data for EGFR SNP genotypes,

b rs11543848 was merged with rs2227983. CI, confidence interval; dbSNP-ID, single nucleotide polymorphism database identifier); rs11568315 SS, rs712829 GG, rs712830 CC, rs2227983 GG: wild-type homozygotes; rs11568315 SL, rs712829 GT, rs712830 CA, rs2227983 GA: heterozygotes; rs11568315 LL, rs712829 TT, rs712830 AA, rs2227983 AA: mutant homozygotes; NA, not applicable; Ref., reference value; NR, not reported; OR, odds ratio; EGFR, epidermal growth factor receptor; UTR, untranslated region.

Due to lack of data from other studies for comparison, one study was excluded from further analysis, although the study did identify rs884225, a 3′-untranslated region variant c.*774T>C associated with EGFR TKI toxicity (41). Similarly, data for the hepatotoxicity, as well as for four EGFR SNPs were not included (rs2075102, rs2293347, rs11977388 and rs4947492) (27). An RCT study reported pre-calculated ORs for three examined EGFR SNPs, but without precise numbers of patients per genotype (24), and was therefore included in the qualitative, but not the quantitative, analysis. Consequently, another RCT study was excluded from quantitative analysis (27) to avoid comparison between observational and RCT studies, and prevent potential heterogeneity. If zeros present in patient genotype numbers for rs2227983 and rs11568315 interfered with computation or if there were insufficient data for analysis, studies were excluded from quantitative synthesis (2325,28). In the literature search, three other studies explored EGFR TKI toxicity, as well as EGFR SNPs, but failed to find any associations between them (4244).

Side effects of EGFR-TKIs

There was notable inconsistency in the scientific reports describing the association between EGFR SNPs and TKI toxicity in patients with NSCLC. While some articles reported evidence of association with skin toxicity (2326) or severe diarrhea (26), one article found no association with skin or gastrointestinal toxicities (27).

In patients treated with gefitinib, there was a significant association between SS genotype in CA repeat polymorphism and early G2/3 skin rash (P=0.031), meaning these patients were more likely to develop early G2/3 rash (23). Despite this, the EGFR polymorphisms −216G>T and R521K were not associated with early G2/3 rash (P=0.104 and P=0.720, respectively) (23). Another study on patients treated with erlotinib found a similar result for three EGFR polymorphisms and skin rash: −191C>A, −216G>T and CA repeats (P=1.00, P=0.13 and P=0.34, respectively) (24). Only the EGFR −216/-191GC haplotype was associated with the appearance of skin rash (P=0.029) (25). Nevertheless, the absence of association with skin rash was evidenced for the single EGFR SNPs −191C>A (P=0.62), −216G>T (P=0.147) and CA repeats (P=0.36) (25). Diarrhea was a less frequent toxicity and no significant association between any of the EGFR SNPs or haplotypes with diarrhea was observed (25). However, in another study, severe diarrhea occurred in patients with NSCLC treated with gefitinib, most frequently in carriers of −191C>A, −191A>A (P<0.0001) and −216G>G genotypes (P<0.01) (26). There was no significant association between EGFR CA repeat polymorphisms and skin or gastrointestinal toxicity, nor any association between EGFR polymorphism and skin toxicity (26).

Toxicity

The most common adverse effects associated with TKIs in treating advanced NSCLC were skin toxicity (78.36%) and diarrhea (20.75%; Table II). One study reported hepatotoxicity (0.89%) (27), but the study was excluded since there were no data from other studies for comparison. Among the studies available for the meta-analysis, gefitinib (250 mg/day) or erlotinib (150 mg/day) were predominant. For data available for genotypes relative to skin toxicity, the OR and 95% confidence interval (CI) were calculated and their effect was summarized in the quantitative synthesis (Fig. 2). This involved three EGFR SNPs (rs11568315, rs712829 and rs712830) obtained from three studies for skin toxicity (23,25,26). Of these, two examined rs11568315 and diarrhea (26,28). The pooled OR for skin toxicity and rs11568315, rs712829 and rs712830 was 1.17 (95% CI, 0.63–2.18; P=0.616) with moderate heterogeneity (I2=57.4%; P=0.022; Fig. 2).

To test heterogeneity, random effect model and subgroup analyses were performed. Subgroup analysis for skin toxicity showed that the OR for rs11568315 was 2.72 (95% CI, 1.34–5.49; P=0.005) without heterogeneity (I2=0.0%; P=0.533). A statistically significant result for skin toxicity (z=2.785 and P=0.005) were obtained under the dominant genetic model for rs11568315 (SS vs. SL + LL). OR for rs712829 was 0.81 (95% CI, 0.28–2.36; P=0.700) with moderate heterogeneity (I2=65.1%; P=0.057) and OR for rs712830 was 0.62 (95% CI, 0.29–1.35; P=0.229) with no heterogeneity (I2=0.0%, P=0.625; Fig. 3). Data for diarrhea was only available for rs11568315 (data not shown). It was tested in two studies using the fixed effects model (OR, 1.21; 95% CI, 0.52–2.82), with no evidence of heterogeneity (I2=0.0%; P=0.422) and without statistically significant association (P=0.661) (26,28).

Publication bias and sensitivity analysis

The results of the sensitivity analysis regarding toxicity were relatively stable. The overall effective size was not affected by exclusion of each of the studies, even by a study with a smaller sample size (OR, 6.87; 95% CI, 1.17–2.28; Fig. 4A) (23). The funnel plot for EGFR SNPs and TKI skin toxicity in patients with NSCLC was roughly symmetric (Fig. 4B). Begg's funnel plot and Egger's regression test (P=0.545) were used to test the publication bias, but no significantly different results were obtained (Fig. 4C and D). Similarly, the funnel plot revealed no potential bias of rs11568315 (CA repeat) and TKI-caused diarrhea (data not shown). Galbraith's plot identified no source of heterogeneity relative to skin toxicity (data not shown).

Discussion

The present systematic review involved the analysis of two RCTs and four cohort studies to test the association of EGFR polymorphisms with the potential toxicity of TKI therapy regimens in patients with NSCLC. A total of 1,123 patients per genotype were observed with any TKI-associated toxicity. A total of four studies provided data for the meta-analysis (23,25,26,28), while six were involved in quality analysis (2328). In the literature search, nine EGFR SNPs relative to TKI toxicity were identified: rs11568315, rs712829, rs712830, rs2227983, rs2075102, rs2293347, rs11977388, rs4947492 and rs884225 (2328,41). Of these, enough data was available for three (rs11568315, rs712829 and rs712830) to be included in the meta-analysis.

Our recent meta-analysis showed that CA repeat polymorphism and −216G>T significantly affected survival in patients with NSCLC treated with TKI (17). In light of the inconsistency of previous reports (2331), the present meta-analysis was performed to extend our previous findings and to analyze the effect of the EGFR polymorphisms and TKIs on NSCLC.

A number of studies in the present review founded an association between some EGFR polymorphisms and TKI-related skin toxicity (23,25,26) or diarrhea (26,27). Contradictory results were also detected in previous studies published before 2009 (4547). The most common TKI adverse effects in the present meta-analysis were skin toxicity and diarrhea, which were 78.36% and 20.75%, respectively (concerning any grade of toxicity vs. no toxicity). They were separately analyzed in the meta-analysis. The pooled OR for three EGFR SNPs (rs11568315, rs712829 and rs712830) was 1.17 (95% CI, 0.63–2.18) without a statistically significant overall effect on skin toxicity (P=0.616). In further analysis, a moderate overall heterogeneity (I2=57.4%; P=0.022) was observed. To explore the heterogeneity further, a subgroup analysis was performed and the random effect model was applied. The subgroup analysis involved three EGFR SNPs (rs11568315, rs712829 and rs712830) concerning skin toxicity (23,25,26). The source of heterogeneity (I2=65.1%; P=0.057) was likely due to the −216G>T (rs712829) polymorphism (26). The CIs were overlapping the line of no effect for all three studies, suggesting the result was not statistically significant. A total of two studies favored the GG genotype for −216G>T (rs712829) and skin toxicity (23,25), which contrasts the GT+TT genotype favored by Giovannetti et al (26). Most importantly, there was no heterogeneity for the other two SNPs examined (rs11568315, I2=0.0%; P=0.533; rs712830, I2=0.0%; P=0.625).

Chemotherapy is the first-line treatment for patients with NSCLC, but notable improvements in the response rate have been observed following the application of the TKIs gefitinib and erlotinib (68,48). However, resistance, as well as adverse effects, is common in this therapy regimen. Typical side effects of the drugs used in NSCLC treatment (for both monoclonal antibodies and small molecule TKIs) are skin rash and diarrhea (49,50). Since the EGFR is commonly affected by somatic mutations in altered neoplastic cells and the EGFR gene is highly polymorphic, the potential cause of those toxic manifestations of drugs may be EGFR genetic variability (11,1316). SNPs or microsatellite tandem repeats are typically found in the EGFR promoter region and intron 1. These notably affect EGFR gene expression and may mediate response to TKI therapy. A CA single sequence repeat polymorphism (rs11568315) is located in EGFR intron 1 and it usually comprises 14–21 variable short tandem repeats. The shorter allele is associated with increased EGFR expression and carriers of this polymorphism are better responders to TKI therapy and have prolonged overall survival time (13,14,47,5154).

Among side effects of TKI therapy, typical skin rash manifestations were in the form of papules and pustules on the scalp, face, neck and upper trunk. To the best of our knowledge, the mechanism of skin rash development has not yet been elucidated. One hypothesis is that there is a genetic susceptibility for rash development, where altered EGFR expression alters the TKI response (11,1316,28). Another is that poor vascularization of the tumor tissue and drug concentrations at a level that does not inhibit tumor growth may cause a skin rash by over-saturation of EGFR (18,55). There is evidence of a significant association between skin rash and an improved outcome in patients with NSCLC (18,56). Skin rash has been reported to be a predictor of tumor response (25) and EGFR CA repeat is a valuable predictor of early G2/3 rash (23). Previous studies have reported that lower number CA repeat carriers develop skin toxicity when treated with gefitinib (13,23,57), while other studies did not (2428,45,47,53). In another study where patients with NSCLC were treated with erlotinib, SL allele length was associated with a higher risk of diarrhea (46). In the present meta-analysis, the pooled OR values for CA repeats (rs11568315) and skin toxicity were 2.72 (95% CI, 1.34–5.49). A significant association with skin toxicity was evident under the dominant genetic model. Namely, heterozygote and long alleles (SL + LL) or prevalently long CA repeat carriers were more likely to develop TKI-related skin toxicity (P=0.005). However, it is probable that short CA carriers would be less likely develop skin rash. There was no association between CA repeats and diarrhea (P=0.661).

The other well-examined SNPs, −191C>A (rs712830) and −216G>T (rs712829) polymorphisms, are located in the EGFR promoter region and are associated with enhanced EGFR mRNA expression (14,53,58). A previous meta-analysis revealed that any genotype with T allele for −216G>T showed an association with higher response and disease control rates and longer PFS and OS times than GG homozygote carriers (59). Another meta-analysis elucidated that the −216G>T polymorphism significantly affected OS and PFS times in patients with NSCLC treated with gefitinib or erlotinib (17). Both of the aforementioned polymorphisms are reported to be in linkage disequilibrium (D'=1.0) (25). Considering their association with toxicities, a study reported the haplotypes showing association with the appearance of skin rash (25). Other studies reported that the T allele of −216G>T was significantly associated with high-risk of TKI-induced skin rash (24) or diarrhea (14). An association between −216G>T and −191C>A with grade >1 diarrhea has also been reported (26). Contrary to these findings, the present meta-analysis observed no significant association for EGFR SNPs −216G>T and −191C>A with skin toxicity (P=0.700 and P=0.229, respectively), which agreed with the findings from previous studies (23,24,27,45).

The advantage of the present meta-analysis over previous meta-analyses is the examination of commonly used TKIs (such as gefitinib and erlotinib) and their toxicity, while other studies involved a single therapeutic agent (60,61). Other analyses investigated EGFR mutations, not EGFR polymorphisms (29,30) or their association with toxicity (59), or they only investigated toxicity in relation to radiotherapy (31). The present meta-analysis has certain limitations. Firstly, some studies included in the analysis had small sample sizes so consistent conclusions could not be obtained, as with the RCTs that have larger sample sizes. A total of two RCTs were excluded from the meta-analysis (24,27), since one study alone did not provide enough data to be tested. In particular, exact numbers of patients with NSCLC with each EGFR SNP genotype were not reported and the study presented only pre-calculated data for OR (24). The aforementioned RCTs obtained low NOS scores in the quality analysis, whereas the other studies included in the present meta-analysis had relatively good scores. Also, the results of the present meta-analysis were not adjusted for other factors (i.e., demographic factors), although the majority of the studies did not report ethnicity for the examined subjects. Potential bias in the results may be due to the absence of a consensus in the literature of an exact number of CA repeats when reporting short vs. long alleles. The linkage disequilibrium between examined SNPs was not taken into account and selection bias may be present.

In conclusion, the results of the present meta-analysis revealed that out of nine EGFR SNPs related to TKI side effects, rs11568315, rs712829 and rs712830 were associated with skin toxicity. NSCLC carriers of long CA repeats (rs11568315, SL + LL) were more likely to develop TKI-associated skin toxicity than short CA repeats (rs11568315, SS). To establish clear inter-individual benefits of TKI therapy, future RCTs that include a broader genetic panel are required to determine genetic susceptibility to TKI-induced toxicity in patients with NSCLC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Ministry of Education, Science and Technological Development, Serbia (no. OI 175056 under agreement number 451-03-68/2022-14/200378).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

Conceptualization and supervision of the study were conducted by VJ. The selection of papers, formal analysis, investigation and writing were conducted by JO. The acquisition of data was conducted by JT. All authors have read and approved the final manuscript. JO, JT and VJ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Yang ZY, Liu L, Mao C, Wu XY, Huang YF, Hu XF and Tang JL: Chemotherapy with cetuximab versus chemotherapy alone for chemotherapy-naive advanced non-small cell lung cancer. Cochrane Database Syst Rev. 17:CD0099482014.PubMed/NCBI

4 

Hirsch FR, Herbst RS, Olsen C, Chansky K, Crowley J, Kelly K, Franklin WA, Bunn PA Jr, Varella-Garcia M and Gandara DR: Increased EGFR gene copy number detected by fluorescent in situ hybridization predicts outcome in non-small-cell lung cancer patients treated with cetuximab and chemotherapy. J Clin Oncol. 26:3351–3357. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, Park K, de Marinis F, Eberhardt WEE, Paz-Ares L, Störkel S, et al: EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: Analysis of data from the phase 3 FLEX study. Lancet Oncol. 13:33–42. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, Gemma A, Harada M, Yoshizawa H, Kinoshita I, et al: Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 362:2380–2388. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, Zhang S, Wang J, Zhou S, Ren S, et al: Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): A multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 12:735–742. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Lee CK, Davies L, Wu YL, Mitsudomi T, Inoue A, Rosell R, Zhou C, Nakagawa K, Thongprasert S, Fukuoka M, et al: Gefitinib or erlotinib vs chemotherapy for EGFR mutation-positive lung cancer: Individual patient data meta-analysis of overall survival. J Natl Cancer Inst. 109:djw2792017. View Article : Google Scholar

9 

Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, et al: Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 350:2129–2139. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, et al: EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science. 304:1497–1500. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Yun CH, Mengwasser KE, Toms AV, Woo MS, Greulich H, Wong KK, Meyerson M and Eck MJ: The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc Natl Acad Sci U S A. 105:2070–2075. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Goldberg SB, Oxnard GR, Digumarthy S, Muzikansky A, Jackman DM, Lennes IT and Sequist LV: Chemotherapy with Erlotinib or chemotherapy alone in advanced non-small cell lung cancer with acquired resistance to EGFR tyrosine kinase inhibitors. Oncologist. 18:1214–1220. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Amador ML, Oppenheimer D, Perea S, Maitra A, Cusatis G, Iacobuzio-Donahue C, Baker SD, Ashfaq R, Takimoto C, Forastiere A and Hidalgo M: An epidermal growth factor receptor intron 1 polymorphism mediates response to epidermal growth factor receptor inhibitors. Cancer Res. 64:9139–9143. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Liu G, Gurubhagavatula S, Zhou W, Wang Z, Yeap BY, Asomaning K, Su L, Heist R, Lynch TJ and Christiani DC: Epidermal growth factor receptor polymorphisms and clinical outcomes in non-small-cell lung cancer patients treated with gefitinib. Pharmacogenomics J. 8:129–138. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Jung M, Cho BC, Lee CH, Park HS, Kang YS, Kim SK, Chang J, Kim DJ, Rha SY, Kim JH and Lee JH: EGFR polymorphism as a predictor of clinical outcome in advanced lung cancer patients treated with EGFR-TKI. Yonsei Med J. 53:1128–1135. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Winther-Larsen A, Ebert EB, Meldgaard P and Sorensen BS: EGFR gene polymorphism predicts improved outcome in patients with EGFR mutation-positive non-small cell lung cancer treated with erlotinib. Clin Lung Cancer. 20:161–166.e161. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Jurisic V, Vukovic V, Obradovic J, Gulyaeva LF, Kushlinskii NE and Djordjević N: EGFR polymorphism and survival of NSCLC patients treated with TKIs: A systematic review and meta-analysis. J Oncol. 2020:19732412020. View Article : Google Scholar : PubMed/NCBI

18 

Peréz-Soler R and Saltz L: Cutaneous adverse effects with HER1/EGFR-targeted agents: Is there a silver lining? J Clin Oncol. 23:5235–5246. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Segaert S and Van Cutsem E: Clinical signs, pathophysiology and management of skin toxicity during therapy with epidermal growth factor receptor inhibitors. Ann Oncol. 16:1425–1433. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Agero AL, Dusza SW, Benvenuto-Andrade C, Busam KJ, Myskowski P and Halpern AC: Dermatologic side effects associated with the epidermal growth factor receptor inhibitors. J Am Acad Dermatol. 55:657–670. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Hu JC, Sadeghi P, Pinter-Brown LC, Yashar S and Chiu MW: Cutaneous side effects of epidermal growth factor receptor inhibitors: Clinical presentation, pathogenesis, and management. J Am Acad Dermatol. 56:317–326. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Bianchini D, Jayanth A, Chua YJ and Cunningham D: Epidermal growth factor receptor inhibitor-related skin toxicity: Mechanisms, treatment, and its potential role as a predictive marker. Clin Colorectal Cancer. 7:33–43. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Huang CL, Yang CH, Yeh KH, Hu FC, Chen KY, Shih JY, Lin ZZ, Yu CJ, Cheng AL and Yang PC: EGFR intron 1 dinucleotide repeat polymorphism is associated with the occurrence of skin rash with gefitinib treatment. Lung Cancer. 64:346–351. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Liu G, Cheng D, Ding K, Maitre AL, Liu N, Patel D, Chen Z, Seymour L, Shepherd FA and Tsao MS: Pharmacogenetic analysis of BR.21, a placebo-controlled randomized phase III clinical trial of erlotinib in advanced non-small cell lung cancer. J Thorac Oncol. 7:316–322. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Parmar S, Schumann C, Rüdiger S, Boeck S, Heinemann V, Kächele V, Seeringer A, Paul T, Seufferlein T and Stingl JC: Pharmacogenetic predictors for EGFR-inhibitor-associated skin toxicity. Pharmacogenomics J. 13:181–188. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Giovannetti E, Zucali PA, Peters GJ, Cortesi F, D'Incecco A, Smit EF, Falcone A, Burgers JA, Santoro A, Danesi R, et al: Association of polymorphisms in AKT1 and EGFR with clinical outcome and toxicity in non-small cell lung cancer patients treated with gefitinib. Mol Cancer Ther. 9:581–593. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Kim L, Saieg M, Di Maio M, Gallo C, Butts C, Ciardiello F, Feld R, Cheng D, Gebbia V, Burgio MA, et al: Biomarker analysis of the phase 3 TORCH trial for first line erlotinib versus chemotherapy in advanced non-small cell lung cancer patients. Oncotarget. 8:57528–57536. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Ma Y, Xin S, Huang M, Yang Y, Zhu C, Zhao H, Zhang Y, Chen L, Zhao Y, Li J, et al: Determinants of Gefitinib toxicity in advanced non-small cell lung cancer (NSCLC): A pharmacogenomic study of metabolic enzymes and transporters. Pharmacogenomics J. 17:325–330. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Petrelli F, Borgonovo K, Cabiddu M, Lonati V and Barni S: Relationship between skin rash and outcome in non-small-cell lung cancer patients treated with anti-EGFR tyrosine kinase inhibitors: A literature-based meta-analysis of 24 trials. Lung Cancer. 78:8–15. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Wo H, He J, Zhao Y, Yu H, Chen F and Yi H: The efficacy and toxicity of gefitinib in treating non-small cell lung cancer: A meta-analysis of 19 randomized clinical trials. J Cancer. 9:1455–1465. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Wang X, Xu Y, Tang W and Liu L: Efficacy and safety of radiotherapy plus EGFR-TKIs in NSCLC patients with brain metastases: A meta-analysis of published data. Transl Oncol. 11:1119–1127. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Moher D, Liberati A, Tetzlaff J and Altman DG: Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. BMJ. 339:b25352009. View Article : Google Scholar : PubMed/NCBI

33 

Ottawa Hospital Research Institute, . NOS Manual. Available from:. http://www.ohri.ca/programs/clinical_epidemiology/nos_manual.pdfMarch 8–2021

34 

Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ and McQuay HJ: Assessing the quality of reports of randomized clinical trials: Is blinding necessary? Control Clin Trials. 17:1–12. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Higgins JPT, Thompson SG, Deeks JJ and Altman DG: Measuring inconsistency in meta-analyses. BMJ. 327:557–560. 2003. View Article : Google Scholar : PubMed/NCBI

36 

DerSimonian R and Laird N: Meta-analysis in clinical trials revisited. Contemp Clin Trials. 45:139–145. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Galbraith RF: A note on graphical presentation of estimated odds ratios from several clinical trials. Stat Med. 7:889–894. 1988. View Article : Google Scholar : PubMed/NCBI

38 

Begg CB and Mazumdar M: Operating characteristics of a rank correlation test for publication bias. Biometrics. 50:1088–1101. 1994. View Article : Google Scholar : PubMed/NCBI

39 

Egger M, Smith GD, Schneider M and Minder C: Bias in meta-analysis detected by a simple, graphical test. BMJ. 315:629–634. 1997. View Article : Google Scholar : PubMed/NCBI

40 

National Cancer Institue, . Common Terminology Criteria for Adverse Events (CTCAE) Version 4. Available at. https://stacks.stanford.edu/file/druid:nw036fx4646/CTCAE_4.03_2010-06-14_QuickReference_5×7.pdfFebruary 12–2021

41 

Ruan Y, Jiang J, Guo L, Li Y, Huang H, Shen L, Luan M, Li M, Du H, Ma C, et al: Genetic association of curative and adverse reactions to tyrosine kinase inhibitors in chinese advanced non-small cell lung cancer patients. Sci Rep. 6:233682016. View Article : Google Scholar : PubMed/NCBI

42 

Nie Q, Yang XN, An SJ, Zhang XC, Yang JJ, Zhong WJ, Liao RQ, Chen ZH, Su J, Xie Z and Wu YL: CYP1A1*2A polymorphism as a predictor of clinical outcome in advanced lung cancer patients treated with EGFR-TKI and its combined effects with EGFR intron 1 (CA)n polymorphism. Eur J Cancer. 47:1962–1970. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Tiseo M, Rossi G, Capelletti M, Sartori G, Spiritelli E, Marchioni A, Bozzetti C, De Palma G, Lagrasta C, Campanini N, et al: Predictors of gefitinib outcomes in advanced non-small cell lung cancer (NSCLC): Study of a comprehensive panel of molecular markers. Lung Cancer. 67:355–360. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Chilingirova N, Hammoudeh Z, Balabanski L, Ivanov S, Vazharova R, Nikolova D, Kurteva G, Toncheva D and Chilingirov P: TruSight cancer sequencing panel reveals pharmacogenetic variants associated with sensitivity to chemotherapy in lung cancer. Memo-Magazine of European Medical Oncology. 9:30–38. 2016. View Article : Google Scholar

45 

Cusatis G, Gregorc V, Li J, Spreafico A, Ingersoll RG, Verweij J, Ludovini V, Villa E, Hidalgo M, Sparreboom A and Baker SD: Pharmacogenetics of ABCG2 and adverse reactions to gefitinib. J Natl Cancer Inst. 98:1739–1742. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Rudin CM, Liu W, Desai A, Karrison T, Jiang X, Janisch L, Das S, Ramirez J, Poonkuzhali B, Schuetz E, et al: Pharmacogenomic and pharmacokinetic determinants of erlotinib toxicity. J Clin Oncol. 26:1119–1127. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Tiseo M, Capelletti M, De Palma G, Franciosi V, Cavazzoni A, Mozzoni P, Alfieri RR, Goldoni M, Galetti M, Bortesi B, et al: Epidermal growth factor receptor intron-1 polymorphism predicts gefitinib outcome in advanced non-small cell lung cancer. J Thorac Oncol. 3:1104–1111. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Cappuzzo F, Ligorio C, Jänne PA, Toschi L, Rossi E, Trisolini R, Paioli D, Holmes AJ, Magrini E, Finocchiaro G, et al: Prospective study of gefitinib in epidermal growth factor receptor fluorescence in situ hybridization-positive/phospho-Akt-positive or never smoker patients with advanced non-small-cell lung cancer: The ONCOBELL trial. J Clin Oncol. 25:2248–2255. 2007. View Article : Google Scholar : PubMed/NCBI

49 

O'Byrne KJ, Bondarenko I, Barrios C, Eschbach C, Martens U, Kortsik YH, Celik I, Stroh C and Pirker R: Molecular and clinical predictors of outcome for cetuximab in non-small cell lung cancer (NSCLC): Data from the FLEX study. J Clin Oncol. 27:8007. 2009. View Article : Google Scholar

50 

Su X, Lacouture ME, Jia Y and Wu S: Risk of high-grade skin rash in cancer patients treated with cetuximab-an antibody against epidermal growth factor receptor: Systemic review and meta-analysis. Oncology. 77:124–133. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Dubey S, Stephenson P, Levy DE, Miller JA, Keller SM, Schiller JH, Johnson DH and Kolesar JM; Eastern Cooperative Oncology Group, : EGFR dinucleotide repeat polymorphism as a prognostic indicator in non-small cell lung cancer. J Thorac Oncol. 1:406–412. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Gebhardt F, Bürger H and Brandt B: Modulation of EGFR gene transcription by secondary structures, a polymorphic repetitive sequence and mutations-a link between genetics and epigenetics. Histol Histopathol. 15:929–936. 2000.PubMed/NCBI

53 

Han SW, Jeon YK, Lee KH, Keam B, Hwang PG, Oh DY, Lee SH, Kim DW, Im SA, Chung DH, et al: Intron 1 CA dinucleotide repeat polymorphism and mutations of epidermal growth factor receptor and gefitinib responsiveness in non-small-cell lung cancer. Pharmacogenet Genomics. 17:313–319. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Ichihara S, Toyooka S, Fujiwara Y, Hotta K, Shigematsu H, Tokumo M, Soh J, Asano H, Ichimura K, Aoe K, et al: The impact of epidermal growth factor receptor gene status on gefitinib-treated Japanese patients with non-small-cell lung cancer. Int J Cancer. 120:1239–1247. 2007. View Article : Google Scholar : PubMed/NCBI

55 

Dienstmann R, Braña I, Rodon J and Tabernero J: Toxicity as a biomarker of efficacy of molecular targeted therapies: Focus on EGFR and VEGF inhibiting anticancer drugs. Oncologist. 16:1729–1740. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Wacker B, Nagrani T, Weinberg J, Witt K, Clark G and 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. 13:3913–3921. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Perea S, Oppenheimer D, Amador M, Cusati G, Baker S, Takimoto C, Maitra A, Iocobuzio-Donahue C and Hidalgo M: Genotypic bases of EGFR inhibitors pharmacological actions. J Clin Oncol. 22:3005. 2004. View Article : Google Scholar

58 

Liu W, Innocenti F, Wu MH, Desai AA, Dolan EM, Cook EH Jr and Ratain MJ: A functional common polymorphism in a Sp1 recognition site of the epidermal growth factor receptor gene promoter. Cancer Res. 65:46–53. 2005. View Article : Google Scholar : PubMed/NCBI

59 

Zhang HX, Tang Y, Wang L, Wei SX, Liu QX, Li F and Yuan XL: EGFR-216 G/T polymorphism as a predictor of clinical outcomes in advanced non-small cell lung cancer patients treated with EGFR-TKIs: A meta-analysis. Int J Clin Exper Med. 9:10273–10280. 2016.

60 

Biaoxue R, Hua L, Wenlong G and Shuanying Y: Efficacy and safety of icotinib in treating non-small cell lung cancer: A systematic evaluation and meta-analysis based on 15 studies. Oncotarget. 7:86902–86913. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Yi L, Fan J, Qian R and Luo P: Efficacy and safety of osimertinib in treating EGFR-mutated advanced NSCLC: A meta-analysis. Int J Cancer. 145:284–294. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2023
Volume 25 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Obradovic J, Todosijevic J and Jurisic V: Side effects of tyrosine kinase inhibitors therapy in patients with non‑small cell lung cancer and associations with <em>EGFR</em> polymorphisms: A systematic review and meta‑analysis. Oncol Lett 25: 62, 2023
APA
Obradovic, J., Todosijevic, J., & Jurisic, V. (2023). Side effects of tyrosine kinase inhibitors therapy in patients with non‑small cell lung cancer and associations with <em>EGFR</em> polymorphisms: A systematic review and meta‑analysis. Oncology Letters, 25, 62. https://doi.org/10.3892/ol.2022.13649
MLA
Obradovic, J., Todosijevic, J., Jurisic, V."Side effects of tyrosine kinase inhibitors therapy in patients with non‑small cell lung cancer and associations with <em>EGFR</em> polymorphisms: A systematic review and meta‑analysis". Oncology Letters 25.2 (2023): 62.
Chicago
Obradovic, J., Todosijevic, J., Jurisic, V."Side effects of tyrosine kinase inhibitors therapy in patients with non‑small cell lung cancer and associations with <em>EGFR</em> polymorphisms: A systematic review and meta‑analysis". Oncology Letters 25, no. 2 (2023): 62. https://doi.org/10.3892/ol.2022.13649