• No results found

ABCB1 Variation Affects Myelosuppression, Progression-free Survival and Overall Survival in Paclitaxel/Carboplatin-treated Ovarian Cancer Patients

N/A
N/A
Protected

Academic year: 2021

Share "ABCB1 Variation Affects Myelosuppression, Progression-free Survival and Overall Survival in Paclitaxel/Carboplatin-treated Ovarian Cancer Patients"

Copied!
53
0
0

Loading.... (view fulltext now)

Full text

(1)

ABCB1 Variation Affects Myelosuppression,

Progression-free Survival and Overall Survival in

Paclitaxel/Carboplatin-treated Ovarian Cancer

Patients

Niclas Björn, Ingrid Jakobsen Falk, Ignace Vergote and Henrik Green

The self-archived postprint version of this journal article is available at Linköping

University Institutional Repository (DiVA):

http://urn.kb.se/resolve?urn=urn:nbn:se:liu:diva-150852

N.B.: When citing this work, cite the original publication.

Björn, N., Jakobsen Falk, I., Vergote, I., Green, H., (2018), ABCB1 Variation Affects Myelosuppression, Progression-free Survival and Overall Survival in Paclitaxel/Carboplatin-treated Ovarian Cancer Patients, Basic & Clinical Pharmacology & Toxicology, 123(3), 277-287.

https://doi.org/10.1111/bcpt.12997

Original publication available at:

https://doi.org/10.1111/bcpt.12997

Copyright: Wiley (12 months)

http://eu.wiley.com/WileyCDA/

(2)

ABCB1 variation affects myelosuppression, progression-free

survival, and overall survival in paclitaxel/carboplatin-treated

ovarian cancer patients

Niclas Björna*, Ingrid Jakobsen Falka, Ignace Vergoteb, Henrik Gréena 4 

a Clinical Pharmacology, Division of Drug Research, Department of

Medical and Health Sciences, Linköping University, SE-58185 6 

Linköping, Sweden. 7 

b Department of Obstetrics and Gynecology, University Hospital Leuven,

Leuven Cancer Institute, B-3000 Leuven, Belgium. 9 

* Corresponding author: 10 

Niclas Björn, Clinical Pharmacology, Division of Drug Research, 11 

Department of Medical and Health Sciences, Linköping University, SE-12  58185 Linköping, Sweden. 13  E-mail: niclas.bjorn@liu.se 14  Telephone: +46101032029 15  Fax: +4613104195 16 

Running title: ABCB1 in paclitaxel-treated ovarian cancer 17 

Keywords: ABCB1, CYP2C8, adverse drug reactions, toxicity, 18 

myelosuppression, ovarian cancer, paclitaxel. 19 

Disclosure statements (conflicts of interest): The authors declare no 20 

conflicts of interest. However, the study was conducted in collaboration 21 

with Oasmia Pharmaceuticals AB, Uppsala, Sweden, who let us perform 22 

this study on their phase III cohort and gave financial support to perform 23 

the pharmacogenetics part of the study. 24 

(3)

Abstract

25 

The standard chemotherapy for ovarian cancer is paclitaxel/carboplatin. 26 

Patients often exhibit myelosuppressive toxicity, and the treatment 27 

response varies considerably. In this study, we investigated the 28 

previously reported SNPs 1199G>A (rs2229109), 1236C>T (rs1128503), 29 

2677G>T/A (rs2032582), 3435C>T (rs1045642) in ABCB1, and 30 

1196A>G (rs10509681) in CYP2C8 and their association with treatment-31 

induced myelosuppression, progression-free survival (PFS), and overall 32 

survival (OS). From the Phase III study, OAS-07OVA, 525 patients (All) 33 

treated with carboplatin and paclitaxel administered as Paclical (Arm A, 34 

n=260) or Taxol® (Arm B, n=265), were included and genotyped using 35 

pyrosequencing. Genotype associations with myelosuppression, PFS, and 36 

OS were investigated using ANOVA, Kaplan-Meier analysis, and Cox 37 

proportional hazard models. The most prominent finding was for the 38 

ABCB1 variant 3435TT, which was significantly associated with 39 

increased PFS in All (hazard ratio (HR) = 0.623), in Arm A (HR=0.590), 40 

and in Arm B (HR=0.627), as well as increased OS in All (HR=0.443) 41 

and in Arm A (HR=0.372) compared to the wildtype, 3435CC. For 42 

toxicity, the most interesting finding concerned the haplotype, including 43 

1236TT, 2677TT, and 3435TT, which was associated with higher 44 

neutrophil values in Arm B (P=0.039) and less neutrophil decrease in All 45 

(P=0.048) and in Arm B (P=0.021). It is noteworthy that the results 46 

varied depending on the treatment arm which indicates that the effects of 47 

ABCB1 variants vary with the treatment regimen. Our results reflect the 48 

contradictory results of previous studies, confirming that small variations 49 

(4)

in the composition of treatment regimens and patient populations may 50 

influence the interpretation of SNPs effects on treatment outcome. 51 

Introduction

52 

Ovarian cancer is one of the leading causes of cancer-related deaths 53 

among women in Europe [1]. The standard chemotherapy for primary 54 

and recurrent ovarian cancer is paclitaxel in combination with 55 

carboplatin [2-4]. Patients initially respond well to this treatment, but the 56 

recurrence rate is around 70%, and the relative 5-year survival rate for 57 

stages III and IV is only 25-30% [5]. Myelosuppression is a common 58 

toxicity of paclitaxel and carboplatin therapy: neutrophil depression is 59 

the most acute hematological toxicity, but thrombocytopenia, leukopenia, 60 

and neutropenia are all commonly acquired by patients undergoing the 61 

treatment [2, 6-9]. Neuropathy is another, often dose-limiting, side-effect 62 

of paclitaxel [2, 10, 11]. 63 

Paclitaxel is insoluble in water, and the solvent Cremophor® EL (CrEL) 64 

is used in the traditional formulation of paclitaxel, Taxol®, to circumvent 65 

this [12]. However, the use of CrEL can lead to hypersensitivity 66 

reactions. Therefore, newer paclitaxel formulations that apply 67 

nanotechnology to form water-soluble micelles, such as Paclical and 68 

Abraxane® [12, 13], have been developed. 69 

Currently, dosages are adapted to the body surface area and Calvert’s 70 

formula for paclitaxel and carboplatin, respectively. However, there 71 

remains considerable variation in both treatment response and toxicities. 72 

There is a need for better means of individualization of the therapy. The 73 

(5)

inter-individual differences in treatment response and toxicity are 74 

thought to be partly attributable to genetic differences. 75 

The two proteins, ABCB1 and CYP2C8, are involved in the 76 

transportation and hepatic metabolism of paclitaxel, respectively [14]. 77 

ABCB1 expression affects paclitaxel resistance in ovarian cancer cell 78 

lines [15] and has for a long time been known to affect paclitaxel drug 79 

disposition [16]. CYP2C8 is the main metabolizing enzyme of paclitaxel, 80 

and genetic variations can alter this metabolism [17]. The effect of 81 

variations in these genes on treatment response and toxicity in cancers 82 

treated with paclitaxel has been extensively studied. However, the 83 

results, from both our group and others have yielded inconclusive and 84 

contradictory results [6, 7, 18-28], reviewed by Krens et al. [29] and 85 

Fredericks et al. [30]. 86 

This inconsistency and the inability to replicate previously reported 87 

results for genetic variants in ABCB1 and CYP2C8 in relation to toxicity 88 

and response can partly be blamed on the use of small sample sizes, 89 

differences in treatment regimens, concomitant medications, the ethnicity 90 

of patients, and the inclusion of various cancer types. This study was 91 

designed to evaluate and validate the association between the SNPs 92 

ABCB1 1199G>A (rs2229109), 1236C>T (rs1128503), 2677G>T/A 93 

(rs2032582), 3435C>T (rs1045642), CYP2C8 1196A>G (rs10509681) 94 

and paclitaxel/carboplatin-induced myelosuppression, progression-free 95 

survival (PFS), and overall survival (OS) in 525 ovarian cancer patients 96 

homogeneously treated with paclitaxel and carboplatin. 97 

(6)

Materials and Methods

98 

Study population

99 

This study was conducted using a subset of patients from the phase III 100 

study OAS-07OVA (EudraCT - 2008-002668-32, ClinicalTrials.gov - 101 

NCT00989131). This phase III study included 789 patients with 102 

recurrent ovarian cancer with the intent to compare the efficacy and 103 

safety of paclitaxel micellar nanoparticles, Paclical, with paclitaxel 104 

CrEL, Taxol®. Eligible patients were randomized to treatment Arm A 105 

consisting of Paclical (250 mg/m2, given as a one-hour IV infusion) and 106 

carboplatin (AUC 5-6) repeated every three weeks for six cycles; or 107 

treatment Arm B consisting of Taxol® (175 mg/m2, given as a three-hour 108 

IV infusion) and carboplatin (AUC 5-6) repeated every three weeks for 109 

six cycles. A total of 525 patients (All; Arm A n=260 and Arm B n=265) 110 

were included in this pharmacogenetics study after giving written 111 

informed consent, following the Helsinki Declaration and with 112 

permission from the regional ethics committees. Peripheral blood 113 

samples were collected and stored at -80°C prior to DNA extraction. 114 

Myelosuppression

115 

Blood status was measured at baseline; at day 1, 8 and 15 through cycles 116 

1 and 2; at day 1 and 8 through cycles 3-6; and at the end of the study. 117 

Myelosuppression was evaluated for thrombocytes, leukocytes, and 118 

neutrophils as nadir values during the first cycle and during the whole 119 

treatment. The nadir values were graded according to the National 120 

Cancer Institute (NCI) Common Terminology Criteria for Adverse 121 

Events (CTCAE) version 4.03. The relative decreases in values from the 122 

(7)

baseline were also evaluated during the first cycle and during the whole 123 

treatment, according to Equation 1. 124 

1:

125 

Progression-free survival and overall survival

126 

Progression-free survival (PFS) and overall survival (OS) were measured 127 

as the time from inclusion to an event, i.e., confirmed progression with 128 

Computed Tomography (CT) or death for PFS and OS, respectively. CT 129 

scans were performed within 6 weeks before treatment, after cycle 3 and 130 

6, every third month during the follow-up period, and when the patient 131 

left the study. 132 

DNA extraction and genotyping

133 

DNA was extracted from blood samples using the Promega Maxwell 16 134 

system (Promega Biotech, Nacka, Sweden) according to the 135 

manufacturer’s protocol. Genotyping of 1199G>A, 1236C>T, 136 

2677G>T/A, and 3435C>T in ABCB1 and 1196A>G in CYP2C8 was 137 

performed using the pyrosequencer PSQ96MD (Qiagen, Uppsala, 138 

Sweden) according to the manufacturer’s protocol, as previously 139 

described [20, 22]. In short, HotStar Taq Master Mixture (VWR 140 

International, Stockholm, Sweden) was used for polymerase chain 141 

reaction (PCR) amplification performed on a Mastercycler gradient 142 

(Eppendorf, Hamburg, Germany) with the total volume 10 µL using 0.4 143 

µmol/L primer, 1.5 mmol/L MgCl2 and annealing temperature of 58°C. 144 

Sequencing primer and biotinylated single-stranded PCR template were 145 

(8)

annealed at 80°C for 2 minutes, enzyme and substrate were added, and 146 

sequencing was carried out by dispensing dNTPs in a predefined order. 147 

ABCB1 haplotype 148 

The haplotype structure of ABCB1 has previously been described by 149 

Kroetz et al. [31]. In this study, haplotypes involving the variants 150 

1236C>T, 2677G>T (excluding the low frequency A variant) and 151 

3435C>T in ABCB1 were investigated. Haplotypes were formed for all 152 

patients that were homozygous wild-type, heterozygous or homozygous 153 

variant at all three variant positions, denoted as CCGGCC, CTGTCT, 154 

and TTTTTT (in order of physical appearance, i.e., 1236C>T, 2677G>T, 155 

3435C>T) in the remainder of this paper. 156 

Statistical analysis

157 

Patient characteristics, baseline blood values, and genotype frequencies 158 

were compared between the treatment arms using logistic regression or 159 

ANOVA. Accordance of genotype distributions with the Hardy-160 

Weinberg equilibrium and the Caucasian population in dbSNP was also 161 

evaluated. 162 

CTCAE grades of thrombocytes, leukocytes, and neutrophils were 163 

compared between the treatment arms using Fisher’s exact test. 164 

The association of baseline thrombocyte, leucocyte, and neutrophil 165 

values with the patient characteristics treatment arm (A or B), age, 166 

relapse (first or second), Eastern Cooperative Oncology Group (ECOG) 167 

performance status, radiotherapy, cytoreductive surgery, previous 168 

treatment with paclitaxel/docetaxel and chemotherapy-free interval (≥1 169 

(9)

or <1 year, defined as the time from last chemotherapy course to 170 

inclusion in this study) all relevant at the time of inclusion (tumor type 171 

and tumor stage were evaluated at primary diagnosis and had likely 172 

changed since; therefore, they were not investigated) were evaluated 173 

using ANOVA. The genotype association with nadir values and relative 174 

decreases of thrombocytes, leucocytes, and neutrophils (during both the 175 

first cycle and the whole treatment) were assessed using ANOVA with 176 

the respective baseline values (only for nadir values, not for relative 177 

decreases) and treatment arm as covariables. 178 

Kaplan-Meier analysis with the log-rank test was used to estimate 179 

differences in PFS and OS between genotypes. For genotypes found to 180 

be significant in the Kaplan-Meier analysis, Cox proportional hazard 181 

models were constructed using the covariables treatment arm, age, 182 

relapse, ECOG performance status, radiotherapy, cytoreductive surgery, 183 

previous treatment with paclitaxel/docetaxel, chemotherapy-free interval 184 

and dose delay/reduction (reduced or delayed dose administration during 185 

treatment). Patients with no registered event (progression or death) were 186 

censored at the date of their last CT scan or last follow-up for PFS and 187 

OS, respectively. 188 

IBM SPSS Statistics version (v.) 23 was used for logistic regressions and 189 

Cox proportional hazard models. The statistical software R [32] (v. 3.2.0) 190 

was used for Fisher’s exact test package stats v. 3.2.0), ANOVA (R-191 

package stats v. 3.2.0), and Kaplan-Meier analysis (R-package survival 192 

v. 2.38-3). All analyses were performed for All, Arm A, and Arm B. 193 

Statistical significance was set as P≤0.05. 194 

(10)

Results

195 

Patient characteristics and baseline blood status

196 

Patient characteristics are presented in Table 1. Logistic regression 197 

showed no significant differences (P>0.05 for all comparisons) in patient 198 

characteristics between the treatment arms, and there were no significant 199 

differences between the treatment arms in terms of baseline blood values. 200 

Furthermore, ANOVA revealed an association between patient 201 

characteristics and baseline blood status (Supplemental Table 1). 202 

However, these associations varied depending on blood cell types. All 203 

patients were of Caucasian ethnicity, except one who was of African 204 

descent. Although all the included patients were previously treated with 205 

platinum-containing chemotherapy, at least six months had passed, so 206 

they were deemed to be platinum sensitive. A clinical paper comparing 207 

the treatment arms using the full study population of the phase III study 208 

OAS-07OVA is under preparation. 209 

Genotyping and genotype distribution

210 

Genotyping was successful, and genotype frequencies are summarized in 211 

Supplemental Table 2. There was no difference in genotype distributions 212 

between the treatment arms (P>0.05 for all comparisons using logistic 213 

regression). The distribution of genotypes corresponded to the 214 

distribution seen for the Caucasian population in dbSNP (data not 215 

shown) and was in accordance with the Hardy-Weinberg distribution 216 

(data not shown). 217 

(11)

The haplotype analysis of ABCB1 categorized 73, 169 and 89 patients as 218 

CCGGCC, CTGTCT, and TTTTTT, respectively. This analysis excluded 219 

294 patients who could not be classified into these haplotypes. 220 

Toxicity

221 

CTCAE grades for nadir values (first cycle and whole treatment) for 222 

thrombocytes, leukocytes, and neutrophils are shown in Supplemental 223 

Table 3. Fisher’s exact test showed significantly higher/greater toxicity 224 

in treatment Arm A for all phenotypes (P<0.05 for all comparisons). 225 

Inherent to the more prevalent toxicity in Arm A, 49 patients received 226 

dose reductions/delay before the second treatment cycle whereas only 23 227 

patients in Arm B required this. Furthermore, during the whole treatment 228 

170 patients in Arm A and 137 in Arm B required dose 229 

delays/reductions. 230 

Results from the ANOVAs of 3435C>T, the haplotype and 231 

myelosuppression are summarized in Table 2. In summary, the variant 232 

alleles of 3435C>T and the haplotype were mainly associated with lower 233 

toxicity (except in Arm A where the variant allele of 3435C>T was 234 

associated with increased neutrophil toxicity). The direction of the 235 

toxicity can be interpreted from boxplots in Supplemental Figure 1, 2 and 236 

3 where mean values and standard deviations (SD) are also displayed for 237 

all significant associations. The genotype 3435C>T was significant for 238 

neutrophil whole treatment nadir in Arm A (P=0.018; mean count/L (SD) 239 

for CC=8.5×108 (9.4×108), CT=6.0×108 (4.1×108) and TT=5.7×108 240 

(4.7×108)), and the neutrophil first cycle relative decrease in All 241 

(P=0.048; mean % (SD) for CC=70.0 (18.3), CT=61.8 (25.6) and 242 

(12)

TT=63.2 (27.0)) and in Arm B (P=0.049; mean relative decrease (SD) for 243 

CC=70.5 (17.5), CT=58.8 (26.6) and TT=60.1 (24.9)). The haplotype 244 

was associated with the thrombocyte first cycle nadir in All (P=0.041; 245 

mean count/L (SD) for CCGGCC=1.5×1011 (6.1×1010), 246 

CTGTCT=1.8×1011 (7.6×1010) and TTTTTT=1.7×1011 (6.4×1010)), the 247 

neutrophil first cycle nadir in Arm B (P=0.039; mean count/L (SD) for 248 

CCGGCC=1.2×109 (8.6×108), CTGTCT= 1.6×109 (9.4×108) and 249 

TTTTTT=1.5×109 (8.9×108)), the leukocyte whole treatment nadir in 250 

Arm B (P=0.029; mean count/L (SD) for CCGGCC=2.1×109 (7.4×108), 251 

CTGTCT=2.3×109 (8.2×108) and TTTTTT=2.6×109 (8.9×108)), and the 252 

neutrophil first cycle relative decrease in All (P=0.048 mean % (SD) for 253 

CCGGCC=70.0 (18.7), CTGTCT=61.7 (25.2) and TTTTTT=65.8 (19.9)) 254 

and in Arm B (P=0.021; mean % (SD) for CCGGCC=71.4 (17.4), 255 

CTGTCT=58.2 (25.2) and TTTTTT=61.1 (21.8)). It should be noted that 256 

the covariable Treatment Arm (A or B) was always significant (data not 257 

shown) in the ANOVAs of toxicity in All. This was expected due to the 258 

harsher treatment regime in Arm A. Also, the covariable baseline blood 259 

status was significant (data not shown) in all but one analysis (Arm A, 260 

leukocytes, P=0.104). For toxicity associations with 1199G>A, 261 

1236C>T, 2677G>T/A, and 1196A>G, see Supplemental Results and 262 

Supplemental Table 4. 263 

Progression-free survival

264 

Kaplan-Meier analysis showed no initial differences in PFS between the 265 

two treatment arms (data not shown). Table 3 lists the results from the 266 

Meier analyses of PFS for 3435C>T and the haplotype. Kaplan-267 

(13)

Meier curves are shown in Supplemental Figure 4 and mean PFS times in 268 

months for all genotypes are listed in Supplemental Table 5. Cox 269 

proportional hazard models for PFS are presented in Table 4. This 270 

showed that the mean PFS in All for patients with 3435TT was 10.8 271 

months compared to 9.6 months in patients with 3435CC, hazard ratio 272 

(HR) = 0.623 (confidence interval (CI) = 0.464–0.835, P=0.002). In Arm 273 

A, the mean PFS for patients with 3435TT was 11.6 months compared to 274 

9.8 months in patients with 3435CC, HR=0.590 (CI=0.390–0.893, 275 

P=0.013). In Arm B, the mean PFS for patients with 3435TT was 10.0 276 

months compared to 9.3 months in patients with 3435CC, HR=0.627 277 

(CI=0.409–0.960, P=0.032). For patients in All with the haplotype 278 

TTTTTT, the mean PFS was 11.0 months compared to 9.9 months in 279 

patients with the haplotype CCGGCC, HR=0.652 (CI=0.452–0.940, 280 

P=0.022). This was also indicated in Arm A (P=0.057). For patients in 281 

Arm B with the heterozygous haplotype CTGTCT, the mean PFS was 282 

10.6 months compared to 10.1 months in patients with the haplotype 283 

CCGGCC, HR=0.625 (CI=0.394–0.991, P=0.046). Kaplan-Meier 284 

analyses of PFS for 1199G>A, 1236C>T, 2677G>T/A, and 1196A>G 285 

are summarized in the Supplemental Results, Supplemental Table 5 and 286 

6, and Supplemental Figure 5 (only 1236C>T). 287 

Overall survival

288 

Kaplan-Meier analysis showed no initial differences in OS between the 289 

two treatment arms (data not shown). Results from the Kaplan-Meier 290 

analyses of OS for 3435C>T and the haplotype are listed in Table 3 and 291 

shown in Supplemental Figure 6, and the mean OS times in days for all 292 

(14)

genotypes are presented in Supplemental Table 5. Cox proportional 293 

hazard models for OS are presented in Table 5. This showed that the 294 

mean OS in All for patients with 3435TT was 431 days compared to 374 295 

days in patients with 3435CC, HR=0.443 (CI=0.264–0.746, P=0.002). In 296 

Arm A, the mean OS for patients with 3435TT was 436 days compared 297 

to 379 days in patients with 3435CC, HR=0.372 (CI=0.186–0.745, 298 

P=0.005). This was not seen in Arm B (P=0.396). Similarly, the mean 299 

OS in All for patients with the haplotype TTTTTT was 435 days 300 

compared to 387 days in patients with the haplotype CCGGCC, 301 

HR=0.467 (CI=0.248–0.878, P=0.018). In Arm A, the mean OS for the 302 

haplotype TTTTTT was 444 days compared to 366 days for the 303 

haplotype CCGGCC, HR=0.336 (CI=0.136–0.827, P=0.018). However, 304 

this was not seen in Arm B (P=0.712). Kaplan-Meier analyses of OS for 305 

1199G>A, 1236C>T, 2677G>T/A, and 1196A>G are summarized in the 306 

Supplemental Results, Supplemental Table 5 and 6, and Supplemental 307 

Figure 7 (for 1236C>T and 2677G>T). 308 

Depiction of significant associations

309 

Figure 1 shows an overall view of the significant associations for the 310 

ABCB1 variants 1199G>A, 3435C>T and the haplotype, as well as the 311 

CYP2C8 variant 1196A>G. 312 

Discussion

313 

The major strengths of this study are its large and clearly defined 314 

population using homogenous treatment in two different regimens. A 315 

potential drawback of the study is that variations in genes involved in 316 

(15)

DNA repair, metabolism, and transportation essential for carboplatin [25, 317 

33, 34] were not investigated. Variation in those genes could also 318 

influence toxicity, PFS, and OS. However, as patients in both treatment 319 

arms were treated with equal doses of carboplatin, the effects should be 320 

similar in both and not a major contributor to the differences seen, 321 

although their impact cannot be ruled out entirely. Also, no correction for 322 

multiple testing was done, which should be borne in mind when 323 

interpreting the results. 324 

Toxicity

325 

For ABCB1, the variant alleles of 1236C>T, 2677G>T, 3435C>T, and 326 

the variant haplotype TTTTTT were associated with less 327 

myelosuppressive toxicity in All and in Arm B. Although this was not 328 

consistent for all blood cell types or for the duration of the treatment, it 329 

indicates that the effects of ABCB1 SNPs on myelosuppression may 330 

differ depending on factors specific to different blood cell types. 331 

However, we did not see the same pattern in Arm A. At the same time, 332 

Arm A had more myelosuppressive toxicity overall, probably due to the 333 

higher concentration of paclitaxel. This indicates that the protective 334 

effect of the variant alleles shown in Arm B can be counteracted by a 335 

harsher treatment regimen like the one used in Arm A. 336 

Hoffmeyer et al. [35] have previously shown decreased mRNA 337 

expression for the variant allele of 3435C>T, followed by increased 338 

digoxin plasma concentrations. This is in accordance with the results of 339 

Wang et al. [36] which showed decreased expression and lowered 340 

mRNA stability, which were subsequently supported by the findings of 341 

(16)

Sissung et al. [26] describing increased neutropenia in patients carrying 342 

3435C>T variant alleles. On the contrary, Baldissera et al. [37] describe 343 

higher ABCB1 expression for 3435T allele carriers in hepatocellular 344 

carcinoma samples. Furthermore, cell populations in peripheral blood 345 

and bone marrow express ABCB1 [38]. The idea is that lowered 346 

expression of ABCB1 leads to decreased cellular clearance of paclitaxel, 347 

higher intracellular concentrations and thus more cell death and greater 348 

toxicity. This infers that a low expression of ABCB1 in tumor cells would 349 

be beneficial to the treatment of cancer, and high expression of ABCB1 350 

in blood cells would be helpful for preventing toxicity. Our findings that 351 

ABCB1 variant alleles decrease myelosuppressive toxicity cannot be 352 

explained by the results of Hoffmeyer et al. [35], Wang et al. [36] and 353 

Sissung et al. [26]. However, the expression of ABCB1 in different cell 354 

types and the effects of the genetic variants on gene expression and 355 

toxicity is not fully known. 356 

The CYP2C8 variant 1196A>G was associated with higher leukocyte 357 

toxicity in Arm A. 1196A>G is included in the CYP2C8*3 haplotype and 358 

is in linkage with rs1113129 (D’=1.0 evaluated using SNAP [39]) in 359 

CYP2C8-HapC. Previous studies have associated CYP2C8-HapC [40] 360 

and CYP2C8*3 [18, 22] with decreased paclitaxel clearance and 361 

CYP2C8-HapC [7] with lower leukocyte and neutrophil values. This 362 

means that CYP2C8 variation could mediate an increased risk of 363 

paclitaxel/carboplatin-induced leukopenia. 364 

(17)

Survival

365 

Chemotherapy-free interval ≥ 1 year was the biggest contributor to 366 

longer PFS and OS. This is understandable since a more aggressive 367 

cancer would result in a faster recurrence, and thus shorter 368 

chemotherapy-free interval, and a worse prognosis. When PFS and OS 369 

were investigated, 1236TT, 3435TT, and the haplotype TTTTTT were 370 

associated with longer PFS; and 1236TT, 2677TT, 3435TT, and the 371 

haplotype TTTTTT were associated with longer OS compared to 372 

wildtypes. The most prominent finding was that 3435TT showed 373 

increased PFS in All, Arm A, and Arm B as well as enhanced OS in All 374 

and Arm A. This adds up to an indication of overall better prognosis for 375 

patients being 3435TT. The meta-analysis of 4616 ovarian cancer 376 

patients by Johnatty et al. [23] marginally associated 1236TT 377 

(rs1128503) with improved OS and showed a tendency for a worse 378 

prognosis in patients with higher tumor expression of ABCB1. Also, 379 

ABCB1 2677T/A (allele carriers), and patients with 1236TT and 3435CC 380 

have been associated with longer PFS [24, 27]. However, the latter is not 381 

supported by our findings as we found the 3435TT variants to be 382 

beneficial. Furthermore, some studies using different treatment regimens 383 

have not seen any differences between genotypes and PFS or OS [19, 384 

25], which might be explained by the fact that the different treatment 385 

regimens in our study yielded different associations of genotypes for PFS 386 

and OS (and myelosuppression). The effects observed are small and 387 

likely not only dependent on the genotypes but also on treatment 388 

conditions. 389 

(18)

Another critical factor that could affect differences in survival is the 390 

tumor expression of ABCB1. High ABCB1 tumor expression has been 391 

associated with poor prognosis and chemotherapy resistance by Sun et 392 

al. [41]. This was further strengthened by the results of Hoffmeyer et al. 393 

[35] and Wang et al. [36] showing that 3435TT is associated with lower 394 

ABCB1 expression, which supports our findings of 3435TT being 395 

favorable for PFS and OS (unlike the association seen with toxicity, as 396 

previously discussed). However, Gao et al. [42] have suggested that an 397 

ABCB1-related survival difference in ovarian cancer patients is more 398 

likely to be an effect of whole body paclitaxel clearance variations rather 399 

than an effect of the variant on cancer cells. Since we do not know about 400 

the somatic ABCB1 mutations or tumor expression of ABCB1 in our 401 

samples, it is impossible to draw definite conclusions. 402 

Lastly, the previously shown short progression-free survival of patients 403 

with the ABCB1 variant 1199G>A [19, 21] was not seen in the presented 404 

study. One explanation for this could be the presence of different 405 

haplotypes in different study populations, yet another contributor to the 406 

inconclusive body of literature published concerning ABCB1 genetic 407 

variants and their importance for treatment effects. 408 

Conclusion

409 

Combining the results from myelosuppression, PFS and OS the study 410 

indicates a toxicity protective effect of variant alleles in ABCB1, 411 

especially for the variant allele of 3435C>T. The effects of ABCB1 412 

variants in this study, using a large homogeneous patient sample, are 413 

however small, and varied with the dose of paclitaxel. This currently 414 

(19)

limits the predictive value of ABCB1 genotyping before the start of 415 

paclitaxel treatment in clinical practice, based on our results. Our results 416 

are reflected in the contradictory results of previous studies [6, 7, 18-30], 417 

confirming that small variations in the composition of treatment 418 

regimens, and the selection of patient populations could influence the 419 

interpretation of the SNP effect on treatment outcomes. Furthermore, the 420 

investigated variants might need different consideration depending on 421 

whether the aim is to minimize toxicity or maximize the anti-tumor 422 

effect of the treatment. Therefore, more thorough investigations of 423 

paclitaxel treatments and genotypes are needed in combination with 424 

ABCB1 expression analysis, and PK/PD studies in vivo and in vitro using 425 

different cell types and both normal and tumor tissues to get a clear idea 426 

of how to implement ABCB1 variation into the individualization of 427 

treatment regimens containing paclitaxel. 428 

Acknowledgements

429 

The research presented in this article was supported by grants from the 430 

Swedish Cancer Society, the Swedish Research Council, Linköping 431 

University, and ALF grants Region Östergötland. Also, we would like to 432 

thank Oasmia Pharmaceuticals AB, Uppsala, Sweden, for letting us 433 

perform this pharmacogenetics study on their phase III cohort and for 434 

their financial support of the pharmacogenetics part of the study. 435 

(20)

References

436 

1. Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer 437 

incidence and mortality in Europe in 2008. European journal of cancer. 438 

2010;46:765-81. 439 

2. du Bois A, Luck HJ, Meier W, Adams HP, Mobus V, Costa S, et 440 

al. A randomized clinical trial of cisplatin/paclitaxel versus 441 

carboplatin/paclitaxel as first-line treatment of ovarian cancer. J Natl 442 

Cancer Inst. 2003;95:1320-9. 443 

3. Parmar MK, Ledermann JA, Colombo N, du Bois A, Delaloye 444 

JF, Kristensen GB, et al. Paclitaxel plus platinum-based chemotherapy 445 

versus conventional platinum-based chemotherapy in women with 446 

relapsed ovarian cancer: the ICON4/AGO-OVAR-2.2 trial. Lancet. 447 

2003;361:2099-106. 448 

4. Stuart GC, Kitchener H, Bacon M, duBois A, Friedlander M, 449 

Ledermann J, et al. 2010 Gynecologic Cancer InterGroup (GCIG) 450 

consensus statement on clinical trials in ovarian cancer: report from the 451 

Fourth Ovarian Cancer Consensus Conference. Int J Gynecol Cancer. 452 

2011;21:750-5. 453 

5. Cannistra SA. Cancer of the ovary. The New England journal of 454 

medicine. 2004;351:2519-29. 455 

6. Bergmann TK, Brasch-Andersen C, Green H, Mirza MR, 456 

Skougaard K, Wihl J, et al. Impact of ABCB1 variants on neutrophil 457 

depression: a pharmacogenomic study of paclitaxel in 92 women with 458 

ovarian cancer. Basic & clinical pharmacology & toxicology. 459 

2012;110:199-204. 460 

(21)

7. Green H, Khan MS, Jakobsen-Falk I, Avall-Lundqvist E, 461 

Peterson C. Impact of CYP3A5*3 and CYP2C8-HapC on 462 

paclitaxel/carboplatin-induced myelosuppression in patients with ovarian 463 

cancer. Journal of pharmaceutical sciences. 2011;100:4205-9. 464 

8. Langer CJ, Leighton JC, Comis RL, O'Dwyer PJ, McAleer CA, 465 

Bonjo CA, et al. Paclitaxel and carboplatin in combination in the 466 

treatment of advanced non-small-cell lung cancer: a phase II toxicity, 467 

response, and survival analysis. J Clin Oncol. 1995;13:1860-70. 468 

9. Baird RD, Tan DS, Kaye SB. Weekly paclitaxel in the treatment 469 

of recurrent ovarian cancer. Nat Rev Clin Oncol. 2010;7:575-82. 470 

10. Argyriou AA, Polychronopoulos P, Koutras A, Xiros N, Petsas 471 

T, Argyriou K, et al. Clinical and electrophysiological features of 472 

peripheral neuropathy induced by administration of cisplatin plus 473 

paclitaxel-based chemotherapy. Eur J Cancer Care (Engl). 2007;16:231-474 

7. 475 

11. Kudlowitz D, Muggia F. Defining risks of taxane neuropathy: 476 

insights from randomized clinical trials. Clin Cancer Res. 2013;19:4570-477 

7. 478 

12. Ma P, Mumper RJ. Paclitaxel Nano-Delivery Systems: A 479 

Comprehensive Review. J Nanomed Nanotechnol. 2013;4:1000164. 480 

13. Joerger M. Treatment regimens of classical and newer taxanes. 481 

Cancer Chemother Pharmacol. 2016;77:221-33. 482 

14. Oshiro C, Marsh S, McLeod H, Carrillo MW, Klein T, Altman 483 

R. Taxane pathway. Pharmacogenet Genomics. 2009;19:979-83. 484 

15. Vaidyanathan A, Sawers L, Gannon AL, Chakravarty P, Scott 485 

AL, Bray SE, et al. ABCB1 (MDR1) induction defines a common 486 

(22)

resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer 487 

cells. Br J Cancer. 2016;115:431-41. 488 

16. Gottesman MM, Pastan I, Ambudkar SV. P-glycoprotein and 489 

multidrug resistance. Curr Opin Genet Dev. 1996;6:610-7. 490 

17. Dai D, Zeldin DC, Blaisdell JA, Chanas B, Coulter SJ, 491 

Ghanayem BI, et al. Polymorphisms in human CYP2C8 decrease 492 

metabolism of the anticancer drug paclitaxel and arachidonic acid. 493 

Pharmacogenetics. 2001;11:597-607. 494 

18. Bergmann TK, Brasch-Andersen C, Green H, Mirza M, Pedersen 495 

RS, Nielsen F, et al. Impact of CYP2C8*3 on paclitaxel clearance: a 496 

population pharmacokinetic and pharmacogenomic study in 93 patients 497 

with ovarian cancer. Pharmacogenomics J. 2011;11:113-20. 498 

19. Bergmann TK, Green H, Brasch-Andersen C, Mirza MR, 499 

Herrstedt J, Holund B, et al. Retrospective study of the impact of 500 

pharmacogenetic variants on paclitaxel toxicity and survival in patients 501 

with ovarian cancer. Eur J Clin Pharmacol. 2011;67:693-700. 502 

20. Green H, Soderkvist P, Rosenberg P, Horvath G, Peterson C. 503 

mdr-1 single nucleotide polymorphisms in ovarian cancer tissue: 504 

G2677T/A correlates with response to paclitaxel chemotherapy. Clin 505 

Cancer Res. 2006;12:854-9. 506 

21. Green H, Soderkvist P, Rosenberg P, Horvath G, Peterson C. 507 

ABCB1 G1199A polymorphism and ovarian cancer response to 508 

paclitaxel. Journal of pharmaceutical sciences. 2008;97:2045-8. 509 

22. Green H, Soderkvist P, Rosenberg P, Mirghani RA, Rymark P, 510 

Lundqvist EA, et al. Pharmacogenetic studies of Paclitaxel in the 511 

(23)

treatment of ovarian cancer. Basic & clinical pharmacology & 512 

toxicology. 2009;104:130-7. 513 

23. Johnatty SE, Beesley J, Gao B, Chen X, Lu Y, Law MH, et al. 514 

ABCB1 (MDR1) polymorphisms and ovarian cancer progression and 515 

survival: a comprehensive analysis from the Ovarian Cancer Association 516 

Consortium and The Cancer Genome Atlas. Gynecologic oncology. 517 

2013;131:8-14. 518 

24. Johnatty SE, Beesley J, Paul J, Fereday S, Spurdle AB, Webb 519 

PM, et al. ABCB1 (MDR 1) polymorphisms and progression-free 520 

survival among women with ovarian cancer following 521 

paclitaxel/carboplatin chemotherapy. Clin Cancer Res. 2008;14:5594-522 

601. 523 

25. Marsh S, Paul J, King CR, Gifford G, McLeod HL, Brown R. 524 

Pharmacogenetic assessment of toxicity and outcome after platinum plus 525 

taxane chemotherapy in ovarian cancer: the Scottish Randomised Trial in 526 

Ovarian Cancer. J Clin Oncol. 2007;25:4528-35. 527 

26. Sissung TM, Mross K, Steinberg SM, Behringer D, Figg WD, 528 

Sparreboom A, et al. Association of ABCB1 genotypes with paclitaxel-529 

mediated peripheral neuropathy and neutropenia. European journal of 530 

cancer. 2006;42:2893-6. 531 

27. Johnatty SE, Beesley J, Paul J, Fereday S, Spurdle AB, Webb 532 

PM. ABCB1 (MDR1) Polymorphisms and Progression-Free Survival 533 

among Women with Ovarian Cancer following Paclitaxel/Carboplatin 534 

Chemotherapy (vol 14, pg 5594, 2008). Clinical Cancer Research. 535 

2012;18:319-20. 536 

(24)

28. Lambrechts S, Lambrechts D, Despierre E, Van Nieuwenhuysen 537 

E, Smeets D, Debruyne PR, et al. Genetic variability in drug transport, 538 

metabolism or DNA repair affecting toxicity of chemotherapy in ovarian 539 

cancer. BMC Pharmacol Toxicol. 2015;16:2. 540 

29. Krens SD, McLeod HL, Hertz DL. Pharmacogenetics, enzyme 541 

probes and therapeutic drug monitoring as potential tools for 542 

individualizing taxane therapy. Pharmacogenomics. 2013;14:555-74. 543 

30. Frederiks CN, Lam SW, Guchelaar HJ, Boven E. Genetic 544 

polymorphisms and paclitaxel- or docetaxel-induced toxicities: A 545 

systematic review. Cancer Treat Rev. 2015;41:935-50. 546 

31. Kroetz DL, Pauli-Magnus C, Hodges LM, Huang CC, 547 

Kawamoto M, Johns SJ, et al. Sequence diversity and haplotype structure 548 

in the human ABCB1 (MDR1, multidrug resistance transporter) gene. 549 

Pharmacogenetics. 2003;13:481-94. 550 

32. RCoreTeam. R: A language and environment for statistical 551 

computing. R Foundation for Statistical Computing, Vienna, Austria; 552 

2015. 553 

33. Perez-Ramirez C, Canadas-Garre M, Alnatsha A, Villar E, 554 

Delgado JR, Faus-Dader MJ, et al. Pharmacogenetic predictors of 555 

toxicity to platinum based chemotherapy in non-small cell lung cancer 556 

patients. Pharmacol Res. 2016;111:877-84. 557 

34. Wu G, Jiang B, Liu X, Shen Y, Yang S. Association of GSTs 558 

gene polymorphisms with treatment outcome of advanced non-small cell 559 

lung cancer patients with cisplatin-based chemotherapy. Int J Clin Exp 560 

Pathol. 2015;8:13346-52. 561 

(25)

35. Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brockmoller J, 562 

Johne A, et al. Functional polymorphisms of the human multidrug-563 

resistance gene: multiple sequence variations and correlation of one 564 

allele with P-glycoprotein expression and activity in vivo. Proceedings of 565 

the National Academy of Sciences of the United States of America. 566 

2000;97:3473-8. 567 

36. Wang D, Johnson AD, Papp AC, Kroetz DL, Sadee W. 568 

Multidrug resistance polypeptide 1 (MDR1, ABCB1) variant 3435C>T 569 

affects mRNA stability. Pharmacogenet Genomics. 2005;15:693-704. 570 

37. Baldissera VD, de Mattos AA, Coral GP, de Araujo FB, Marroni 571 

CA, de Mello Brandao AB, et al. Evaluation of the C3435T 572 

polymorphism in the MDR1 gene in patients with hepatocellular 573 

carcinoma. Ann Hepatol. 2012;11:899-906. 574 

38. Drach D, Zhao S, Drach J, Mahadevia R, Gattringer C, Huber H, 575 

et al. Subpopulations of normal peripheral blood and bone marrow cells 576 

express a functional multidrug resistant phenotype. Blood. 577 

1992;80:2729-34. 578 

39. Johnson AD, Handsaker RE, Pulit SL, Nizzari MM, O'Donnell 579 

CJ, de Bakker PIW. SNAP: a web-based tool for identification and 580 

annotation of proxy SNPs using HapMap. Bioinformatics. 2008;24:2938-581 

9. 582 

40. Rodriguez-Antona C, Niemi M, Backman JT, Kajosaari LI, 583 

Neuvonen PJ, Robledo M, et al. Characterization of novel CYP2C8 584 

haplotypes and their contribution to paclitaxel and repaglinide 585 

metabolism. Pharmacogenomics J. 2008;8:268-77. 586 

(26)

41. Sun S, Cai J, Yang Q, Zhu Y, Zhao S, Wang Z. Prognostic Value 587 

and Implication for Chemotherapy Treatment of ABCB1 in Epithelial 588 

Ovarian Cancer: A Meta-Analysis. PLoS One. 2016;11:e0166058. 589 

42. Gao B, Russell A, Beesley J, Chen XQ, Healey S, Henderson M, 590 

et al. Paclitaxel sensitivity in relation to ABCB1 expression, efflux and 591 

single nucleotide polymorphisms in ovarian cancer. Scientific reports. 592 

2014;4:4669. 593 

(27)

Figure

595 

596 

Figure 1. Overall view of the significant associations in the study. All significant associations of the ABCB1 variants 1199G>A, 3435C>T, and

597 

the haplotype# as well as the CYP2C8 SNP 1196A>G with thrombocytes, leukocytes, neutrophils, progression-free survival, and overall survival

598 

for All, Arm A, and Arm B. Green indicates that the variant was associated with an advantage (lower toxicity, longer PFS, or longer OS) and 599 

orange indicates that the variant was associated with a disadvantage (higher toxicity, shorter PFS, or shorter OS). 600 

* p < 0.05. 601 

** p < 0.01. 602 

# Haplotype combining all homozygous wildtype (CCGGCC), heterozygous (CTGTCT), and homozygous variant (TTTTTT) at all three positions

603 

1236C>T, 2677G>T, and 3435C>T. 604 

(28)

Tables

605 

Table 1: Patient characteristics for All patients, treatment Arm A and treatment Arm B. 606 

All Arm A Arm B

N 525 260 265

Median age (range), at inclusion 56 (26-81) 56 (26-81) 56 (37-81)

Tumor type, primary diagnosis

Epithelial ovarian cancer 504 96.0% 253 97.3% 251 94.7%

Fallopian tube cancer 12 2.3% 4 1.5% 8 3.0%

Primary peritoneal cancer 9 1.7% 3 1.2% 6 2.3%

Tumor stage, primary diagnosis

Stage I 48 9.1% 25 9.6% 23 8.7%

Stage II 52 9.9% 26 10.0% 26 9.8%

Stage III 341 65.0% 171 65.8% 170 64.2%

Stage IV 83 15.8% 38 14.6% 45 17.0%

Missing 1 0.2% 0 0.0% 1 0.4%

Number of relapses, at inclusion

First relapse 399 76.0% 192 73.8% 207 78.1%

Second relapse 126 24.0% 68 26.2% 58 21.9%

ECOG Performance Status, at inclusion

0 291 55.4% 145 55.8% 146 55.1%

1 218 41.5% 106 40.8% 112 42.3%

2 16 3.0% 9 3.5% 7 2.6%

Radiotherapy, before inclusion

Yes 29 5.5% 14 5.4% 15 5.7%

No 496 94.5% 246 94.6% 250 94.3%

Cytoreductive surgery, before inclusion

Yes 482 91.8% 240 92.3% 242 91.3%

No 43 8.2% 20 7.7% 23 8.7%

Paclitaxel and/or docetaxel, before inclusion

Yes 270 51.4% 133 51.2% 137 51.7%

No 255 48.6% 127 48.8% 128 48.3%

Chemotherapy-free interval, time from last administrated

chemotherapy to inclusion

≥1 year 279 53.1% 139 53.5% 140 52.8%

<1 year 246 46.9% 121 46.5% 125 47.2%

Patient status, end of study

Dead 135 25.7% 67 25.8% 68 25.7%

Alive 390 74.3% 193 74.2% 197 74.3%

Abbreviation: ECOG, Eastern Cooperative Oncology Group. 607 

Note: Logistic regression to assess differences in baseline characteristics between treatment Arm A and 608 

treatment Arm B found no significant differences (P>0.05). 609 

(29)

Table 2: P-values from ANOVA showing 3435C>T and ABCB1 haplotype effects on nadir values and relative decrease in thrombocytes,

610 

leukocytes, and neutrophils during both the first cycle and the whole course of the treatment for All, Arm A, and Arm B.   611 

All Arm A Arm B

Response variable Thrombocytes Leukocytes Neutrophils Thrombocytes Leukocytes Neutrophils Thrombocytes Leukocytes Neutrophils

Cycle 1 nadir 3435C>T 0.248 0.499 0.490 0.927 0.910 0.698 0.141 0.280 0.106 Haplotype# 0.041 0.263 0.275 0.363 0.979 0.856 0.102 0.094 0.039 Total nadir 3435C>T 0.444 0.597 0.466 0.705 0.487 0.018 0.343 0.305 0.297 Haplotype# 0.471 0.314 0.912 0.814 0.333 0.151 0.309 0.029 0.096

Cycle 1 relative decrease

3435C>T 0.417 0.109 0.048 0.943 0.364 0.558 0.182 0.290 0.049

Haplotype# 0.224 0.100 0.048 0.655 0.593 0.580 0.276 0.134 0.021

Total relative decrease

3435C>T 0.479 0.359 0.934 0.754 0.810 0.646 0.240 0.425 0.354

Haplotype# 0.563 0.094 0.660 0.850 0.462 0.353 0.272 0.060 0.055

Note: Significant P-values, P ≤ 0.05, are marked in bold. To understand the direction of toxicity for the significant associations see Supplemental 612 

Figures 1, 2, and 3 with boxplots and mean values. 613 

# Haplotype combining all homozygous wildtype (CCGGCC), heterozygous (CTGTCT), and homozygous variant (TTTTTT) at all three positions

614 

1236C>T, 2677G>T, and 3435C>T. 615 

(30)

Table 3: P-values for the Kaplan-Meier analyses of both progression-free survival and overall 616 

survival for 3435C>T and the ABCB1 haplotype for All, Arm A, and Arm B. An overall P-value of 617 

the difference as well as P-values comparing the individual genotypes with each other are shown. 618 

Progression-free survival Overall survival Genotype P-value comparing All Arm A Arm B All Arm A Arm B 3435C>T overall difference 0.018 0.031 0.066 0.011 0.017 0.215

CC TT 0.006 0.032 0.019 0.003 0.006 0.299

CC CT 0.215 0.939 0.057 0.199 0.069 0.919

CT TT 0.039 0.012 0.761 0.025 0.171 0.103 Haplotype# overall difference 0.099 0.084 0.341 0.091 0.077 0.584

CCGGCC TTTTTT 0.052 0.125 0.134 0.043 0.025 0.585 CCGGCC CTGTCT 0.599 0.766 0.206 0.659 0.486 0.841

CTGTCT TTTTTT 0.057 0.021 0.910 0.033 0.088 0.313

Note: Significant P-values, P ≤ 0.05, are marked in bold. Mean progression-free survival and mean overall 619 

survival are presented in Supplemental Table 5. 620 

# Haplotype combining all homozygous wildtype (CCGGCC), heterozygous (CTGTCT), and homozygous 621 

variant (TTTTTT) at all three positions 1236C>T, 2677G>T, and 3435C>T. 622 

(31)

Table 4: Cox proportional hazard models of progression-free survival for the genotypes 1199G>A, 1236C>T, 3435C>T, and haplotype in

623 

All, Arm A, and Arm B. The analysis was split into separate models for each significant genotype from the previous Kaplan-Meier

624 

analyses of progression-free survival.

625 

All Arm A Arm B

P HR 95% CI P HR 95% CI P HR 95% CI

Model with 1199G>A

Treatment Arm A 1 - - - B 0.025 1.270 1.031 - 1.564 - - - - Age 0.016 0.986 0.975 - 0.997 0.009 0.978 0.962 - 0.994 0.524 0.995 0.979 - 1.011 ECOG 0 1 1 1 1 0.447 1.086 0.878 - 1.342 0.951 1.01 0.731 - 1.396 0.524 1.097 0.824 - 1.461 2 0.013 2.243 1.187 - 4.240 0.477 1.372 0.574 - 3.278 <0.001 5.851 2.283 - 14.991 Relapse First 1 1 1 Second 0.590 1.070 0.836 - 1.371 0.279 1.226 0.847 - 1.774 0.996 0.999 0.710 - 1.405 Radiotherapy No 1 1 1 Yes 0.942 0.982 0.603 - 1.599 0.353 1.441 0.666 - 3.117 0.600 0.843 0.445 - 1.597 Cytoreductive surgery No 1 1 1 Yes 0.874 0.969 0.661 - 1.422 0.516 0.825 0.461 - 1.476 0.894 0.966 0.580 - 1.610 Paclitaxel/docetaxel No 1 1 1 Yes 0.884 0.984 0.792 - 1.222 0.363 0.855 0.611 - 1.197 0.753 1.048 0.783 - 1.402

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.548 0.446 - 0.674 0.003 0.631 0.465 - 0.857 <0.001 0.452 0.335 - 0.610 Dose delay/reduction No 1 1 1 Yes 0.233 0.880 0.714 - 1.085 0.384 0.864 0.622 - 1.200 0.204 0.832 0.627 - 1.105 1199G>A G/G 1 1 1 G/A + A/A 0.169 1.348 0.880 - 2.064 0.553 1.176 0.688 - 2.008 0.02 2.389 1.146 - 4.979 Model with 1236C>T Treatment Arm A 1 - - - B 0.026 1.265 1.028 - 1.557 - - - - Age 0.034 0.988 0.976 - 0.999 0.018 0.980 0.964 - 0.996 0.504 0.994 0.979 - 1.011

(32)

ECOG 0 1 1 1 1 0.440 1.087 0.879 - 1.343 0.883 0.976 0.706 - 1.349 0.562 1.089 0.817 - 1.451 2 0.020 2.132 1.125 - 4.040 0.655 1.223 0.506 - 2.953 <0.001 6.061 2.352 - 15.615 Relapse First 1 1 1 Second 0.567 1.075 0.839 - 1.377 0.395 1.177 0.809 - 1.713 0.923 1.017 0.724 - 1.429 Radiotherapy No 1 1 1 Yes 0.766 1.078 0.659 - 1.763 0.358 1.437 0.664 - 3.112 0.760 0.903 0.470 - 1.736 Cytoreductive surgery No 1 1 1 Yes 0.786 0.948 0.644 - 1.395 0.352 0.755 0.418 - 1.363 0.712 0.907 0.541 - 1.521 Paclitaxel/docetaxel No 1 1 1 Yes 0.838 0.978 0.788 - 1.214 0.243 0.818 0.584 - 1.146 0.764 1.046 0.781 - 1.401

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.534 0.434 - 0.657 0.001 0.6 0.440 - 0.820 <0.001 0.423 0.312 - 0.573 Dose delay/reduction No 1 1 1 Yes 0.118 0.845 0.683 - 1.044 0.372 0.863 0.624 - 1.193 0.100 0.784 0.588 - 1.047 1236C>T C/C 1 1 1 C/T 0.069 0.806 0.638 - 1.017 0.784 0.954 0.684 - 1.332 0.011 0.645 0.460 - 0.904 T/T 0.006 0.660 0.491 - 0.889 0.012 0.563 0.360 - 0.882 0.079 0.690 0.457 - 1.043 Model with 3435C>T Treatment Arm A 1 - - - B 0.031 1.256 1.021 - 1.546 - - - - Age 0.020 0.987 0.975 - 0.998 0.012 0.979 0.964 - 0.996 0.621 0.996 0.980 - 1.012 ECOG 0 1 1 1 1 0.335 1.110 0.898 - 1.372 0.985 1.003 0.723 - 1.392 0.585 1.083 0.812 - 1.445 2 0.017 2.180 1.147 - 4.145 0.725 1.173 0.482 - 2.851 <0.001 5.710 2.229 - 14.627 Relapse First 1 1 1 Second 0.609 1.066 0.834 - 1.362 0.376 1.180 0.818 - 1.704 0.981 1.004 0.716 - 1.407 Radiotherapy No 1 1 1 Yes 0.783 1.071 0.656 - 1.749 0.432 1.364 0.629 - 2.958 0.743 0.897 0.467 - 1.721 Cytoreductive surgery No 1 1 1 Yes 0.822 0.957 0.651 - 1.406 0.412 0.781 0.433 - 1.408 0.934 0.978 0.586 - 1.634 Paclitaxel/docetaxel No 1 1 1

(33)

Yes 0.772 0.969 0.780 - 1.202 0.237 0.817 0.584 - 1.143 0.805 1.037 .775 - 1.390

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.541 0.440 - 0.664 0.001 0.606 0.447 - 0.822 <0.001 0.438 0.324 - 0.593 Dose delay/reduction No 1 1 1 Yes 0.206 0.873 0.708 - 1.077 0.398 0.870 0.630 - 1.202 0.150 0.810 0.608 - 1.079 3435C>T C/C 1 1 1 C/T 0.039 0.756 0.580 - 0.986 0.684 0.926 0.638 - 1.344 0.009 0.599 0.407 - 0.881 T/T 0.002 0.623 0.464 - 0.835 0.013 0.590 0.390 - 0.893 0.032 0.627 0.409 - 0.960

Model with Haplotype#

Treatment Arm A 1 - - - B 0.155 1.210 0.930 - 1.575 - - - - Age 0.145 0.990 0.976 - 1.004 0.028 0.978 0.959 - 0.998 0.853 1.002 0.983 - 1.021 ECOG 0 1 1 1 1 0.238 1.171 0.901 - 1.520 0.298 1.246 0.824 - 1.885 0.925 1.017 0.713 - 1.450 2 0.391 1.510 0.590 - 3.867 0.662 0.714 0.157 - 3.235 0.012 4.785 1.416 - 16.175 Relapse First 1 1 1 Second 0.809 1.037 0.775 - 1.388 0.843 0.953 0.590 - 1.540 0.545 1.127 0.766 - 1.658 Radiotherapy No 1 1 1 Yes 0.977 1.008 0.577 - 1.762 0.62 1.241 0.529 - 2.913 0.610 0.817 0.375 - 1.777 Cytoreductive surgery No 1 1 1 Yes 0.584 1.134 0.722 - 1.782 0.942 0.973 0.458 - 2.064 0.598 1.171 0.650 - 2.110 Paclitaxel/docetaxel No 1 1 1 Yes 0.665 0.943 0.724 - 1.229 0.473 0.85 0.546 - 1.324 0.924 0.983 0.690 - 1.400

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.585 0.455 - 0.754 0.006 0.562 0.374 - 0.845 <0.001 0.503 0.348 - 0.728 Dose delay/reduction No 1 1 1 Yes 0.790 0.965 0.745 - 1.251 0.917 0.978 0.648 - 1.477 0.385 0.853 0.597 - 1.220 Haplotype# CCGGCC 1 1 1 CTGTCT 0.343 0.856 0.621 - 1.180 0.525 1.166 0.726 - 1.874 0.046 0.625 0.394 - 0.991 TTTTTT 0.022 0.652 0.452 - 0.940 0.057 0.594 0.347 - 1.015 0.125 0.662 0.391 - 1.121 Abbreviation: ECOG, Eastern Cooperative Oncology Group, HR, Hazard Ratio, 95% CI, 95% Confidence Interval.

(34)

Note: Significant P-values, P ≤ 0.05, are marked in bold. 627 

#Haplotype combining all homozygous wildtype (CCGGCC), heterozygous (CTGTCT), and homozygous variant (TTTTTT) at all three positions

628 

1236C>T, 2677G>T, and 3435C>T.

(35)

Table 5: Cox proportional hazard models of overall survival for the genotypes 1236C>T, 2677G>T, 3435C>T, and haplotype in All, Arm

630 

A, and Arm B. The analysis was split into separate models for each significant genotype from the previous Kaplan-Meier analyses of

631 

overall survival.

632 

All Arm A Arm B

P HR 95% CI P HR 95% CI P HR 95% CI Model with 1236C>T Treatment Arm A 1 - - - B 0.636 0.919 0.647 - 1.304 - - - - - - Age 0.952 0.999 0.981 - 1.018 0.723 1.005 0.978 - 1.033 0.847 0.997 0.969 - 1.026 ECOG 0 1 1 1 1 0.096 1.372 0.946 - 1.989 0.525 1.196 0.688 - 2.081 0.139 1.484 0.880 - 2.502 2 0.215 1.822 0.706 - 4.703 0.567 1.450 0.406 - 5.184 0.303 2.307 0.470 - 11.329 Relapse First 1 1 1 Second 0.581 1.136 0.723 - 1.786 0.209 1.528 0.788 - 2.961 0.759 0.900 0.460 - 1.762 Radiotherapy No 1 1 1 Yes 0.832 1.121 0.392 - 3.205 0.406 1.860 0.431 - 8.020 0.725 0.760 0.165 - 3.494 Cytoreductive surgery No 1 1 1 Yes 0.476 0.794 0.422 - 1.496 0.962 0.975 0.335 - 2.834 0.347 0.670 0.290 - 1.544 Paclitaxel/docetaxel No 1 1 1 Yes 0.815 1.048 0.708 - 1.552 0.929 1.026 0.579 - 1.819 0.905 1.035 0.585 - 1.833

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.378 0.263 - 0.545 0.001 0.425 0.254 - 0.711 <0.001 0.336 0.192 - 0.587 Dose delay/reduction No 1 1 1 Yes 0.198 0.789 0.550 - 1.132 0.286 0.753 0.446 - 1.269 0.493 0.821 0.468 - 1.441 1236C>T C/C 1 1 1 C/T 0.448 0.857 0.576 - 1.277 0.365 0.776 0.448 - 1.344 0.677 0.872 0.458 - 1.660 T/T 0.007 0.487 0.288 - 0.823 0.019 0.390 0.178 - 0.855 0.189 0.585 0.263 - 1.301 Model with 2677G>T Treatment Arm A 1 - - -

(36)

B 0.526 0.891 0.622 - 1.274 - - - - - - Age 0.942 1.001 0.982 - 1.020 0.694 1.006 0.978 - 1.034 0.762 1.005 0.976 - 1.034 ECOG 0 1 1 1 1 0.113 1.362 0.930 - 1.995 0.512 1.215 0.679 - 2.173 0.138 1.493 0.879 - 2.536 2 0.239 1.909 0.651 - 5.598 0.608 1.517 0.309 - 7.452 0.525 1.689 0.335 - 8.508 Relapse First 1 1 1 Second 0.553 1.148 0.728 - 1.811 0.274 1.448 0.746 - 2.813 0.719 0.882 0.446 - 1.745 Radiotherapy No 1 1 1 Yes 0.796 1.150 0.400 - 3.308 0.496 1.666 0.384 - 7.227 0.774 0.796 0.168 - 3.763 Cytoreductive surgery No 1 1 1 Yes 0.742 0.896 0.466 - 1.722 0.956 1.031 0.350 - 3.038 0.66 0.823 0.346 - 1.960 Paclitaxel/docetaxel No 1 1 1 Yes 0.859 0.964 0.642 - 1.448 0.931 0.974 0.532 - 1.782 0.905 0.964 0.533 - 1.745

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.363 0.248 - 0.532 0.003 0.432 0.250 - 0.747 <0.001 0.316 0.177 - 0.564 Dose delay/reduction No 1 1 1 Yes 0.533 0.888 0.610 - 1.292 0.435 0.804 0.465 - 1.390 0.991 1.003 0.561 - 1.796 2677G>T G/G 1 1 1 G/T 0.672 0.913 0.598 - 1.393 0.531 0.830 0.464 - 1.487 0.809 1.089 0.547 - 2.166 T/T 0.006 0.472 0.275 - 0.810 0.018 0.382 0.172 - 0.850 0.292 0.637 0.276 - 1.473 Model with 3435C>T Treatment Arm A 1 - - - B 0.629 0.917 0.645 - 1.303 - - - - - - Age 0.867 0.998 0.980 - 1.017 0.925 1.001 0.976 - 1.028 0.880 0.998 0.971 - 1.026 ECOG 0 1 1 1 1 0.073 1.411 0.968 - 2.056 0.282 1.363 0.775 - 2.395 0.182 1.428 0.846 - 2.411 2 0.273 1.699 0.658 - 4.386 0.599 1.402 0.398 - 4.944 0.346 2.121 0.444 - 10.132 Relapse First 1 1 1 Second 0.496 1.165 0.750 - 1.809 0.184 1.537 0.815 - 2.900 0.735 0.891 0.459 - 1.732 Radiotherapy No 1 1 1 Yes 0.816 1.133 0.396 - 3.238 0.367 1.964 0.454 - 8.498 0.751 0.780 0.168 - 3.617 Cytoreductive surgery No 1 1 1

(37)

Yes 0.626 0.855 0.454 - 1.608 0.965 1.023 0.364 - 2.875 0.429 0.714 0.310 - 1.646

Paclitaxel/docetaxel No 1 1 1

Yes 0.852 1.038 0.699 - 1.542 0.887 1.043 0.587 - 1.852 0.84 1.062 0.592 - 1.904

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.404 0.282 - 0.579 0.001 0.413 0.247 - 0.690 0.001 0.375 0.214 - 0.657 Dose delay/reduction No 1 1 1 Yes 0.219 0.797 0.556 - 1.144 0.326 0.769 0.455 - 1.300 0.623 0.866 0.489 - 1.534 3435C>T C/C 1 1 1 C/T 0.079 0.674 0.434 - 1.047 0.033 0.530 0.295 - 0.951 0.868 0.933 0.414 - 2.103 T/T 0.002 0.443 0.264 - 0.746 0.005 0.372 0.186 - 0.745 0.396 0.672 0.268 - 1.684

Model with Haplotype#

Treatment Arm A 1 - - - B 0.598 0.890 0.576 - 1.374 - - - - - - Age 0.834 1.003 0.979 - 1.026 0.776 1.005 0.972 - 1.039 0.861 1.003 0.969 - 1.038 ECOG 0 1 1 1 1 0.255 1.299 0.827 - 2.041 0.332 1.422 0.699 - 2.892 0.374 1.319 0.716 - 2.430 2 0.421 1.681 0.475 - 5.949 0.823 1.307 0.126 - 13.604 0.769 1.292 0.235 - 7.111 Relapse First 1 1 1 Second 0.623 0.878 0.524 - 1.474 0.867 0.931 0.406 - 2.138 0.45 0.751 0.357 - 1.579 Radiotherapy No 1 1 1 Yes 0.738 0.782 0.184 - 3.315 0.332 2.127 0.463 - 9.778 0.978 0.000 0.000 - inf Cytoreductive surgery No 1 1 1 Yes 0.835 1.083 0.511 - 2.294 0.936 0.948 0.257 - 3.494 0.808 1.136 0.405 - 3.187 Paclitaxel/docetaxel No 1 1 1 Yes 0.690 1.102 0.684 - 1.776 0.436 1.352 0.634 - 2.885 0.648 0.853 0.430 - 1.692

Chemotherapy-free interval <1 year 1 1 1

≥1 year <0.001 0.406 0.261 - 0.631 0.013 0.418 0.210 - 0.833 0.006 0.380 0.190 - 0.760 Dose delay/reduction No 1 1 1 Yes 0.414 0.830 0.531 - 1.297 0.667 0.857 0.424 - 1.730 0.619 0.838 0.418 - 1.682 Haplotype# CCGGCC 1 1 1 CTGTCT 0.629 0.874 0.507 - 1.508 0.356 0.703 0.332 - 1.487 0.916 1.050 0.424 - 2.602 TTTTTT 0.018 0.467 0.248 - 0.878 0.018 0.336 0.136 - 0.827 0.712 0.826 0.298 - 2.286

(38)

Abbreviation: ECOG, Eastern Cooperative Oncology Group, HR, Hazard Ratio, 95% CI, 95% Confidence Interval, inf, infinite. 633 

Note: Significant P-values, P ≤ 0.05, are marked in bold. 634 

# Haplotype combining all homozygous wildtype (CCGGCC), heterozygous (CTGTCT) and homozygous variant (TTTTTT) at all three positions

635 

1236C>T, 2677G>T, and 3435C>T. 636 

(39)

Supplemental results, figures, and tables

Title:

ABCB1 variation affects myelosuppression, progression-free survival, and overall survival in paclitaxel/carboplatin-treated ovarian cancer patients

Journal:

Basic & Clinical Pharmacology & Toxicology

Authors:

Niclas Björna*, Ingrid Jakobsen Falka, Ignace Vergoteb and Henrik Gréena

Affiliations:

a Clinical Pharmacology, Division of Drug Research, Department of Medical and Health Sciences, Linköping University, SE-58185 Linköping, Sweden

b Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven Cancer Institute, B-3000 Leuven, Belgium

* Corresponding author:

Niclas Björn, Clinical Pharmacology, Division of Drug Research, Department of Medical and Health Sciences, Linköping University, SE-58185 Linköping, Sweden.

(40)

Supplemental results

Results for ABCB1 SNPs 1199G>A, 1236C>T, and 2677G>T/A and the CYP2C8 SNP 1196A>G are here presented. Supplemental Figures 1-7 and Supplemental Tables 1-6 will follow the summarizing text below.

Genotyping and genotype distribution

All SNPs were successfully genotyped in all patients and genotype frequencies are summarized in Supplemental Table 2. Due to the low number of A/A genotypes for 1199G>A these were analyzed together with heterozygous genotypes G/A in a dominant manner. Likewise, the G/G genotype of 1196A>G was analyzed together with the A/G genotypes. For 2677G>T/A genotypes including the A allele was excluded from further studies because of the low genotype frequency, leaving 491 patients for analyzes of 2677G>T.

Toxicity

The association of 1199G>A, 1236C>T, 2677G>T/A, and 1196A>G with nadir values and relative decrease of thrombocytes, leukocytes, and neutrophils during the first cycle and the whole treatment were evaluated with ANOVA and the results are listed in Supplemental Table 4. Supplemental Figure 1, 2, and 3 can be used to determine that the significant associations show lower toxicity for the variant alleles of 1236C>T and 2677G>T whereas the variant alleles of 1199G>A and 1196A>G show increased toxicity.

Progression free survival and overall survival

Supplemental Table 7 lists all P-values from the Kaplan-Meier analysis of PFS and OS for 1199G>A, 1236C>T, 2677G>T/A, and 1196A>G. Variants that had at least one significant P-value (1199G>A, 1236C>T, and 2677G>T) were further analyzed using Cox proportional hazard models, results are listed in Table 4 and 5 in the main article for PFS and OS, respectively. These analyses showed that variant alleles of 1236C>T had prolonged PFS in All, Arm A and Arm B. Kaplan-Meier curves for PFS and 1236C>T are shown in Supplemental Figure 5. Further, patients being 1236TT or 2677TT showed prolonged OS in All and Arm A compared to wildtype. OS Kaplan-Meier curves for 1236C>T and 2677G>T are shown in Supplemental Figure 7. Mean PFS and OS times for the variants are listed in Supplemental Table 5.

(41)

Supplemental Figure 1. The figures show jittering of the nadir values during the first cycle for different genotypes (with significant p-values in the ANOVA displayed in Table 2 and Supplementary Table 4)accompanied by boxplots, mean, and standard deviations (SD): A) Leukocyte count for Arm B shown for each of the 1199G>A genotypes G/A + A/A and G/G; B) Thrombocyte count for All shown for each of the 1236C>T genotypes C/C, C/T and T/T; C) Thrombocyte count for All divided on 2677G>T genotypes G/G, G/T and T/T; D) Thrombocyte count for All shown for each of the ABCB1 haplotypes# CCGGCC, CTGTCT, and TTTTTT; E) Neutrophil count for Arm B shown for each of the ABCB1 haplotypes# CCGGCC, CTGTCT, and TTTTTT.

(42)

Supplemental Figure 2. The figures show a jittering of the nadir values during the whole course of the treatment for different genotypes (with significant p-values in the ANOVA displayed in Table 2 and Supplementary Table 4) accompanied by boxplots, mean, and standard deviations (SD): A) Leukocyte count for Arm B shown for each of the 1199G>A genotypes G/A + A/A and G/G; B) Neutrophil count for Arm B shown for each of the 2677G>T

genotypes G/G, G/T and T/T; C) Neutrophil count for Arm A shown for each of the 3435C>T genotypes C/C, C/T and T/T; D) Leukocyte count for Arm B shown for each of the ABCB1 haplotypes# CCGGCC, CTGTCT and TTTTTT; E) Leukocyte count for Arm A shown for each of the CYP2C8 1196A>G genotypes A/A and A/G + G/G.

(43)

Supplemental Figure 3. The figures show a jittering of the relative decrease in nadir values to the baseline values for either the first cycle (A-D) or the whole course of the treatment (E and F) for different genotypes (with significant p-values in the ANOVA displayed in Table 2 and Supplementary Table 4) accompanied by boxplots, mean, and standard deviations (SD): A) Neutrophil relative decrease first cycle for All shown for each of the 3435C>T genotypes C/C, C/T and T/T; B) Neutrophil relative decrease first cycle for Arm B shown for each of the 3435C>T genotypes C/C, C/T and T/T; C) Neutrophil relative decrease first cycle for All shown for each of the ABCB1 haplotypes# CCGGCC, CTGTCT and TTTTTT; D) Neutrophil relative decrease first cycle for Arm B shown for each of the ABCB1 haplotypes# CCGGCC, CTGTCT and TTTTTT; E) Neutrophil relative decrease during the, whole treatment for Arm B shown for each of the 2677G>T genotypes G/G, G/T and T/T; F) Leukocyte relative decrease during the whole treatment for Arm A shown for each of the CYP2C8 1196A>G genotypes A/A

(44)

Supplemental Figure 4. Kaplan-Meier curves for progression-free survival (PFS), in months, for 3435C>T and the haplotype in All, Arm A and Arm B. Significance was assessed using the log-rank test. In All, patients with 3435TT had significantly longer PFS compared to 3435CC (P = 0.006) (A) this was also seen in Arm A (P = 0.032) (B) and Arm B (P = 0.019) (C). For the haplotype#, PFS was significantly longer for patients with TTTTTT compared to CTGTCT in Arm A (P = 0.021) (E), but not in all (P > 0.05) (D) and Arm B (P > 0.05) (F). Mean PFS times for the genotypes and groups are listed in Supplemental Table 5. # Haplotype combining homozygous wildtype (CCGGCC), heterozygous (CTGTCT), and homozygous variant (TTTTTT) at all three positions 1236C>T, 2677G>T, and 3435C>T.

References

Related documents

För att uppskatta den totala effekten av reformerna måste dock hänsyn tas till såväl samt- liga priseffekter som sammansättningseffekter, till följd av ökad försäljningsandel

Från den teoretiska modellen vet vi att när det finns två budgivare på marknaden, och marknadsandelen för månadens vara ökar, så leder detta till lägre

The increasing availability of data and attention to services has increased the understanding of the contribution of services to innovation and productivity in

Generella styrmedel kan ha varit mindre verksamma än man har trott De generella styrmedlen, till skillnad från de specifika styrmedlen, har kommit att användas i större

Parallellmarknader innebär dock inte en drivkraft för en grön omställning Ökad andel direktförsäljning räddar många lokala producenter och kan tyckas utgöra en drivkraft

Närmare 90 procent av de statliga medlen (intäkter och utgifter) för näringslivets klimatomställning går till generella styrmedel, det vill säga styrmedel som påverkar

I dag uppgår denna del av befolkningen till knappt 4 200 personer och år 2030 beräknas det finnas drygt 4 800 personer i Gällivare kommun som är 65 år eller äldre i

Det har inte varit möjligt att skapa en tydlig överblick över hur FoI-verksamheten på Energimyndigheten bidrar till målet, det vill säga hur målen påverkar resursprioriteringar