scispace - formally typeset
Search or ask a question
Journal ArticleDOI

5-Fluorouracil degradation rate as a predictive biomarker of toxicity in breast cancer patients treated with capecitabine.

TL;DR: The pre-treatment evaluation of 5-fluorouracil degradation rate allows to identify breast cancer patients at high risk for severe 5-FU toxicity, as well as predicting G3–4 toxicity occurrence.
Abstract: Capecitabine is an oral prodrug of 5-fluorouracil with a relevant role in the treatment of breast cancer. Severe and unexpected toxicities related to capecitabine are not rare, and the identificati...

Summary (2 min read)

Introduction

  • Capecitabine is an oral prodrug of 5-fluorouracil (5-FU), enzymatically activated by timidina phosphorylase in tumour tissue, that was rationally designed to mimic continuous infusion 5-FU.1 Capecitabine is rapidly absorbed from the gastrointestinal (GI) tract and metabolized by carboxylesterase in liver, and it is converted to 50 deoxy-5-fluorocytidine and then to 50 deoxy-5-fluorouridine (50 DFUR) by cytidine deaminase.
  • In advanced breast cancer (ABC), the efficacy and safety of capecitabine have been demonstrated both as monotherapy and in combination with other drugs.
  • Several trials of adjuvant capecitabine administered in combination with other effective drugs did not show an advantage over regimens without capecitabine.
  • Finally, skin reactions are common: rash and hand&foot syndrome are frequent but also severe skin toxicities such as Stevens– Johnson syndrome and toxic epidermal necrolysis are reported.
  • The authors previously demonstrated that the evaluation of dihydropyrimidine dehydrogenase (DPYD) polymorphisms and the 5-FU degradation rate (5FUDR) can help to distinguish patients prone to develop severe side effects in colorectal and gastric patients treated with 5-FU in both adjuvant and metastatic setting.

Patients

  • Clinical data of 37 patients treated for locally advanced and metastatic breast cancer at their institutions were retrospectively collected.
  • Age >18 years old, diagnosis of metastatic breast cancer, previous treatment with capecitabine for metastatic breast cancer, PS 0-1 at baseline, absence of liver or kidney impairment, also known as Inclusion criteria were.
  • The authors performed a monthly assessment of treatment toxicity according to the National Cancer Institute-Common Terminology Criteria for Adverse Events version 5 (CTCAE v.5, 2017).
  • Patients were instructed to report and were usually asked for common toxicities during follow-up visits.
  • The study was conducted in accordance with the Declaration of Helsinki and the protocol approved by the institutional (Sapienza University) ethical committee.

Methods

  • Peripheral blood samples from all patients enrolled were collected at the baseline as clinical practice in their Institution to perform genotype analysis and evaluate 5FUDR on peripheral blood cells.
  • The individual 5FUDR was assessed by a liquid chromatography–tandem mass spectrometry on peripheral blood mononuclear cells (PBMC).
  • The assay is composed of three steps: (1) PBMC isolation from peripheral blood, (2) PBMC incubation with 5-FU in vitro and (3) determination of 5-FU amount to calculate the degradation rate.
  • 18 5FUDR is the result of the whole intracellular metabolizing process, regardless the presence or not of a single enzyme alterations.

Statistical analysis

  • SPSS statistical software, Version 24 (SPSS Inc. Chicago, Illinois, USA), was used.
  • The v2 test and t test for unpaired data were applied to compare the frequencies and mean, respectively.
  • Genotype variant association with toxicity events was first analysed using univariate logistic regression and further by a multivariate logistic regression including patient age ( 60 vs. <60 years old), comorbidity ( 2 vs. <2) and type of chemotherapy regimen (CAPE alone vs. CAPE plus navelbine/lapatinib).

Results

  • Overall, 37 Caucasian patients with a median age of 58 years old (range 34–79) treated with CAPE-based chemotherapy for stage II–IV breast cancer were included in this study.
  • Their baseline and demographic characteristics are shown in Table 1.
  • The frequency of toxicity (73.7 vs. 77.3% and 26.7 vs. 22.7% for any grade and G3–4 toxicity, respectively) did not differ between patients who received CAPE alone or in combination with other drugs.

Pharmacogenetic variant analyses

  • Homozygous DPYD IVS14þ 1G>A SNP nor heterozygous DPYD was not observed in the cohort.
  • No association was found between 5FUDR and either TSER or MTHFR genotypes (Table 3).

Discussion

  • Several enzymes are involved in the capecitabine metabolism.
  • The dihydropyrimidine dehydrogenase enzyme (DPD) metabolizes about 80% of the administered 5-FU into the inactive metabolite 5,6-dihydro-5fluorouracil.
  • 1G>A polymorphism in the DPYD gene, which leads to the production of an inactive protein and severe toxicity in about one-half of carrier patients.
  • 33,34 Besides, the authors showed that 5FUDR is associated with progression-free survival in metastatic colorectal patients with an advantage for ultra/poor metabolizers versus normal metabolizers.
  • Hence, the authors carried out a retrospective study aimed to evaluate the impact of each of the following gene polymorphisms MTHFR C667T, MTHFR A1298C, DPYD IVS14þ 1G>A, TSER and the 5-fluorouracil degradation rate (5-FUDR) on toxicities in breast cancer patients treated with capecitabine.

MTHFR

  • Suggesting a predictive value of 5FU-degradation regardless of cancer origin and stage.
  • 1G>A polymorphism in the DPYD gene, but this alteration has a low frequency, and it is not present in some of the patients with severe toxicity.
  • In the adjuvant setting, a test able to predict severe toxicities in these particular setting is crucial in order to avoid reduced dose, delayed administration or interruption of therapy and to maintain dose intensity.
  • Finally, their study has some relevant limitations: it is a retrospective and monocentric study and the population is limited.
  • The authors highlight the importance of conducting prospective studies on larger sample size and on a homogeneous population in order to evaluate the 5FUDR impact on both toxicities and outcome.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

Original Article
5-Fluorouracil degradation rate as a
predictive biomarker of toxicity in breast
cancer patients treated with capecitabine
Andrea Botticelli
1
, Simone Scagnoli
1
, Michela Roberto
2
,
Luana Lionetto
2
, Bruna Cerbelli
2
, Maurizio Simmaco
3
and
Paolo Marchetti
4
Abstract
Capecitabine is an oral prodrug of 5-fluorouracil with a relevant role in the treatment of breast cancer. Severe and
unexpected toxicities related to capecitabine are not rare, and the identification of biomarkers is challenging.
We evaluate the relationship between dihydropyrimidine dehydrogenase, thymidylate synthase enhancer region and
methylenetetrahydrofolate reductase polymorphisms, 5-fluorouracil degradation rate and the onset of G3–4 toxicities in
breast cancer patients. Genetic polymorphisms and the 5-fluorouracil degradation rate of breast cancer patients treated
with capecitabine were retrospectively studied. Genetic markers and the 5-fluorouracil degradation rate were corre-
lated with the reported toxicities. Thirty-seven patients with a median age of 58 years old treated with capecitabine for
stages II–IV breast cancer were included in this study. Overall, 34 (91.9%) patients suffered from at least an episode of
any grade toxicity while nine patients had G3–4 toxicity. Homozygous methylenetetrahydrofolate reductase 677TT was
found to be significantly related to haematological toxicity (OR ¼ 6.5 [95% IC 1.1–37.5], P ¼ 0.04). Three patients had
a degradation rate less than 0.86 ng/mL/106 cells/min and three patients greater than 2.1 ng/mL/106 cells/min. At a
univariate logistic regression analysis, an altered value of 5-fluorouracil degradation rate (values < 0.86 or >2.10 ng/mL/
106 cells/min) increased the risk of G3–4 adverse events (OR ¼ 10.40 [95% IC: 1.48–7.99], P ¼ 0.02). A multivariate logistic
regression analysis, adjusted for age, comorbidity and CAPE-regimen, confirmed the role of 5-fluorouracil degradation rate
as a predictor of G3–4 toxicity occurrence (OR ¼ 10.9 [95% IC 1.2–96.2], P ¼ 0.03). The pre-treatment evaluation of
5-fluorouracil degradation rate allows to identify breast cancer patients at high risk for severe 5-FU toxicity.
Keywords
5-FU degradation rate , breast cancer, capecitabine, chemotherapy toxicity, polymorphism
Date received: 26 May 2019; revised: 13 December 2019; accepted: 15 January 2020
Introduction
Capecitabine is an oral prodrug of 5-fluorouracil
(5-FU), enzymatically activated by timidina phosphor-
ylase in tumour tissue, that was rationally designed
to mimic continuous infusion 5-FU.
1
Capecitabine is
rapidly absorbed from the gastrointestinal (GI) tract
and metabolized by carboxylesterase in liver, and it is
converted to 5
0
deoxy-5-fluorocytidine and then to
5
0
deoxy-5-fluorouridine (5
0
DFUR) by cytidine deam-
inase. Finally, the enzyme thymidine phosphorylase
converts 5
0
DFUR to 5-FU both in normal and
tumour tissues. However, after an oral dose of capeci-
tabine, the concentration of 5-FU in tumor tissue is
higher than in adjacent healthy tissue, as result of an
higher activity of Thymidine phosphorylase (TP).
2
Oral
administration seems to be preferred by patients and
allows to avoid complications and costs linked to 5-FU
IV infusion.
3
1
Azienda Policlinico Umberto I Roma, Roma, Italy
2
Azienda Ospedaliera Sant’Andrea, Roma, Lazio, Italy
3
Department of Neurosciences, Mental Health and Sensory Organs
(NESMOS), Sapienza University of Rome, Roma, Italy
4
Department of Medical Oncology, St Andrea University Hospital, Rome,
Italy
Corresponding author:
Simone Scagnoli, Azienda Policlinico, Umberto I Viale del Policlinico, 155
Roma, Roma 00161, Italy.
Email: simone.scagnoli@hotmail.it
J Oncol Pharm Practice
0(0) 1–7
! The Author(s) 2020
Article reuse guidelines:
sagepub.com/journals-permissions
DOI: 10.1177/1078155220904999
journals.sagepub.com/home/opp

In advanced breast cancer (ABC), the efficacy and
safety of capecitabine have been demonstrated both as
monotherapy and in combination with other drugs.
Several studies have demonstrated the efficacy of cape-
citabine at the dose of 1250 or 1000 mg/m
2
twice
daily for 14 over 21 days in metastatic breast cancer
patients.
4,5
The oral prodrug has also been tested in
an older population of patients (>65 years old) result-
ing effective and well tolerated at the dose of
1000 mg/m
2
.
6
Capecitabine is a landmark treatment
also in HER2þ metastatic disease in association
with lapatinib, after initial progression to first or
second line.
7
Conversely, the role of capecitabine in the adjuvant
setting is still uncertain. Several trials of adjuvant cape-
citabine administered in combination with other effec-
tive drugs did not show an advantage over regimens
without capecitabine.
8,9
Recently, in the CRATE
study, about 900 patients with HER2 negative breast
cancer with residual disease after standard neoadjuvant
chemotherapy have been randomized to receive adju-
vant capecitabine or no treatment (control group).
The results showed improvement in disease-free surviv-
al (74.1% vs. 67.6%r at 5 years, respectively, HR 0.70,
p: 0.01) and overall survival (OS) (89.2% vs. 83.6%
respectively, HR 0.59; P ¼ 0. 01) in treatment group
and even more significant in TNBC subgroup.
10
As
expected, the addition of capecitabine caused
treatment-related toxicities.
11,12
Regardless cancer primary site and setting of treat-
ment, however, capecitabine is related with several high
grade toxicities, some of them with quick onset and
unexpected severity.
13
Diarrhea is the most common,
adverse event that occurs in 55–60% of patients, with a
severe grade 3 or 4 of toxicity in about 12–15%. Other
common GI adverse events are nausea and vomiting.
Moreover, haematological adverse events like neutro-
penia, anaemia and thrombocytopenia are very
common during the treatment. Finally, skin reactions
are common: rash and hand&foot syndrome are fre-
quent but also severe skin toxicities such as Stevens–
Johnson syndrome and toxic epidermal necrolysis are
reported.
14–16
In view of the wide use of capecitabine in breast
cancer, the identification of predictive factors for
capecitabine-related toxicities is a pressing need. Pre-
emptive identification of patients that will develop
severe toxicities is still an open issue. Several tests
have been developed to identify those patients with
alterations in 5-FU metabolism that can lead to
undue and unexpected toxicities. We previously dem-
onstrated that the evaluation of dihydropyrimidine
dehydrogenase (DPYD) polymorphisms and the
5-FU degradation rate (5FUDR) can help to distin-
guish patients prone to develop severe side effects in
colorectal and gastric patients treated with 5-FU in
both adjuvant and metastatic setting.
17
The aim of our study is to explore the association
between 5FUDR, DPYD, thymidylate synthase
enhancer region (TSER) and methylenetet rahydrofo-
late reductase (MTHFR) polymorphisms and toxicities
in breast cancer patients treated with capecitabine.
Patients and methods
Patients
Clinical data of 37 patients treated for locally advanced
and metastatic breast cancer at our institutions were
retrospectively collected. Inclusion criteria were: age
>18 years old, diagnosis of metastatic breast cancer,
previous treatment with capecitabine for metastatic
breast cancer, PS 0-1 at baseline, absence of liver or
kidney impairment. Exclusion criteria were: PS 2 or
more at baseline, less than one month of treatment
with capecitabine and patient lost at follow up during
treatment with capecitabine. As a clinical practice,
CAPE was used at 1000–1250 mg/mq twice daily for
14 days followed by a seven-day rest period. We per-
formed a monthly assessment of treatment toxicity
according to the National Cancer Institute-Common
Terminology Criteria for Adverse Events version 5
(CTCAE v.5, 2017). Patien ts were instructed to
report and were usually asked for common toxicities
during follow-up visits. Total toxicity was defined as
the percentage of patients who suffered from at least
one adverse event irrespective of type and grade. The
study was conducted in accordance with the Declaration
of Helsinki and the protocol approved by the institutional
(Sapienza University) ethical committee.
Methods
Peripheral blood samples from all patients enrolled
were collected at the baseline as clinical practice in
our Institution to perform genotype analysis and eval-
uate 5FUDR on peripheral blood cells. Genotyping of
DPYD GIVS14A (rs3918290), MTHFR C677T
(rs1801133) and A1298C (rs1801131) SNPs was per-
formed by pyro-sequencing technology. PCR analysis
was used for genotyping TSER polymorphism
(rs34743033). The individual 5FUDR was assessed by
a liquid chromatography–tandem mass spectrometry
on peripheral blood mononuclear cells (PBMC).
5FUDR is determined in vitro by measuring the
decrease of a fixed amount of 5-FU (10 mg/mL) added
to a solution of PBMC, after 2 h incubation, expressed
as nanogram per millilitre of 5-FU degraded per
minute 10 cells. The assay is composed of three
steps: (1) PBMC isolation from peripheral blood, (2)
2 Journal of Oncology Pharmacy Practice 0(0)

PBMC incubation with 5-FU in vitro and (3) determi-
nation of 5-FU amount to calculate the degradation
rate.
18
5FUDR is the result of the whole intracellular
metabolizing process, regardless the presence or not of
a single enzyme alterations. As previously reported,
patients were categorized in three groups according
to their 5FUDR value: below the fifth centile (poor
metabolizers–PMs), above the 95th centile (ultra-rapid
metabolizers–UMs) and within the 5–95th centile (0.85–
2.2 ng/mL/106 cells/min) (extensive metabolizers–EM).
Statistical analysis
SPSS statistical software, Version 24 (SPSS Inc.
Chicago, Illinois, USA), was used. Each MTHFR gen-
otypes C677T (CC, CT and TT) and A1298C (AA, AC,
CC), TSER genotypes (2 R/2R, 2 R/3R, 3 R/3R) and
DPYD (GG/GA) were tested. The v
2
test and t test
for unpaired data were applied to compare the frequen-
cies and mean, respectively. Genotype variant associa-
tion with toxicity events was first analysed using
univariate logistic regression and further by a multi var-
iate logistic regression including patient age (60 vs.
<60 years old), comorbidity (2 vs. <2) and type of
chemotherapy regimen (CAPE alone vs. CAPE plus
navelbine/lapatinib). A P value < 0.05 was considered
as statistically significant.
Results
Overall, 37 Caucasian patients with a median age of
58 years old (range 34–79) treated with CAPE -based
chemotherapy for stage II–IV breast cancer were
included in this study. Their baseline and demographic
characteristics are shown in Table 1. A 25% dose
reduction was done in nine cases (24.3%), and the
treatment was prematurely stopped in six (16.2%)
patients due to G1–4 GI (40%) and haematological
(60%) adverse events. The most common treatment-
related adverse events are reported in Figure 1.
CAPE was administered with an adjuvant intent in
16 (43.2%) patients and in 21 (56.8%) patients affected
by ABC, and it was administered alone or in combina-
tion with other drugs in 15 (40.5%) and 22 (59.5%)
cases, respectively. Overall, 34 (91.9%) patients suf-
fered from at least an episode of any grade toxicity
while 9 (24.3%) patients had G3–4 toxicity. No toxic
death was reported. The frequency of toxicity (73.7 vs.
77.3% and 26.7 vs. 22.7% for any grade and G3–4
toxicity, respectively) did not differ between patients
who received CAPE alone or in combination with
other drugs. Besides, capecitabine plus navelbine regi-
men showed a higher incidence of any grade haemato-
logical toxicity and G1–2 GI toxicity than capecitabine
alone or in combination with lapatinib (Figure 2).
Pharmacogenetic variant analyses
The distribution of the analysed genotypes did not
deviate from Hardy–Weinberg equilibrium (DPYD,
P ¼ 0.9, MTHFR 677, P ¼ 0.86; MTHFR 1298,
P ¼ 0.50; TSER, P ¼ 0.87). Homozygous DPYD
Table 1. Clinicopathological parameters of patients.
Parameter Number %
Total 37 100
Median age years (range) 58 (34–79)
Comorbidity
<2 32 86.5
2 5 13.5
Stage
II–III 16 43.2
IV 21 56.8
Estrogen receptor
Median (range) 80 (0–100)
Negative 11 30.6
Progesteron receptor
Median (range) 49.5 (0–90)
Negative 10 27.8
Her2/neu
Positive 10 27.0
Negative 27 73.0
Grading
1 2 5.4
2 6 16.2
3 22 59.5
Missing 7 18.9
Ki 67 expression
Median (range) 32 (7–89)
Capecitabine-based therapy
Mono-chemotherapy 15 40.5
Plus navelbine 17 46.0
Plus lapatinib 5 13.5
TSER
2R/2R 7 18.9
3R/3R 14 37.8
2R/3R 14 37.8
Missing 2 5.4
MTHFR 677
CC 11 29.7
CT 19 51.4
TT 6 16.2
Missing 1 2.7
MTHFR 1298
AA 17 45.9
AC 15 40.5
CC 5 13.5
5FU degradation value
EM 31 83.8
PM 3 8.1
UP 3 8.1
5FU: 5-fluorouracil; EM: extensive metabolizer; PM: poor metabolizer,
UM: ultra-rapid metabolizer.
Botticelli et al. 3

IVS14 þ 1G > A SNP nor heterozygous DPYD was
not observed in the cohort. Homozygous MTHFR
677TT was found to be significantly related to haema-
tological toxicity (OR ¼ 6.5 [95% IC 1.1–37.5],
P ¼ 0.04). However, no association was detected
between each other SNPs and toxicity (Table 2).
Overall, the mean value SD of 5FUDR was 1.45
0.45 (range: 0.49–2.50) ng/mL/106 cells/min. Three
patients had a degradation rate less than 0.86 ng/mL/
106 cells/min (PMs) and three patients greater than
2.1 ng/mL/106 cells/min (UMs). No association was
found between 5FUDR and either TSER or MTHFR
genotypes (Table 3). At a univariate logistic regression
analysis, an altered value of 5FUDR (values < 0.86 or
>2.10 ng/mL/106 cells/min) increased the risk of G3–4
adverse events (OR ¼ 10.40 [95% IC: 1.48–7.99],
P ¼ 0.02) (Table 2). A multivariate logistic regression
analysis, adjusted for age, comorbidity and CAPE reg-
imen, confirmed the role of 5FUDR as a predictor
of G3–4 toxicity occurrence (OR ¼ 10.9 [95% IC 1.2–
96.2], P ¼ 0.03.
Discussion
Several enzymes are involved in the capecitabine
metabolism.
19
The dihydropyrimidine dehydrogenase
enzyme (DPD) metabolizes about 80% of the admin-
istered 5-FU into the inactive metabolite 5,6-dihydro-5-
fluorouracil. The remaining 20% is converted into
active metabolites that cause the inhibition of thymidy-
late synthase (TYMS) and RNA/DNA damage.
20
Several genotypes of the DPD have been associated
Leucopenia
Neutropenia
Anaemia Nausea Vomit Constipation Diarrhoea Mucositis
Hand-foot
syndrome
G1–G2
2661772944
G3G4
042110402
0
2
4
6
8
10
12
14
16
18
N. of patients
Figure 1. Most common toxicities in study population.
G1–G2 G3–G4 G1–G2 G3–G4 G1–G2 G3–G4
Haematological toxicity Gastrointestinal toxicity Hand-foot syndrome
CAPE
408422
CAPE plus Nvb
7410210
CAPE plus lapatinib
003110
0
2
4
6
8
10
12
N. of patients
Figure 2. Type and severity of toxicity according to the therapeutic scheme administered.
4 Journal of Oncology Pharmacy Practice 0(0)

with reduced enzyme activity that could lead to severe
toxic adverse events of capecitabine or fluoropyrimi-
dine.
21
The most used pharmacogenetic test to predict
DPD activity is base d on the detection of IVS14 þ
1G > A polymorphism in the DPYD gene, which
leads to the production of an inactive protein and
severe toxicity in about one-half of carrier patients.
22
Moreover, a decreased value of 5FUDR is linked to
DPYD haplotype, and it could be related to adverse
events development;
23
however, this polymorphism has
a low frequency. Other enzymes are involved in 5-FU
metabolism, and their polymorphisms could result in
increased and unexpected toxicities such as MTHFR,
one of the most relevant enzyme that regulates intra-
cellular folate levels that affect DNA synthesis and
methylation and TYMS.
24,25
The single-nucleotide polymorphisms (SNPs) of
MTHFR 677 C > T and 1298 A > C are clinically
relevant and have been associated with the toxicity
of 5-FU.
26
Moreover, variations of the TSER in the
promoter of TYMS gene have been related to both
survival/response outcomes and toxicities in patients
affected by colorectal cancer treated with 5-FU-based
chemotherapy.
27–30
Finally, we previously described
a non-genomic assay that seems to be able to predict
5-FU toxicity by the assessment of the 5FUDR in the
PBMC.
31
This parameter indicates the amount of drug
consumed by cells in a time unit and reflexes the result
of the entire 5-FU degradation metabolism, not only a
single enzyme activity.
32
Applying the assay on colo-
rectal patients, we previously described two different
classes of patients with a higher risk to develop 5-FU
unexpected toxicities: poor metabolizers and ultrarapid
metabolizers.
33,34
Besides, we showed that 5FUDR is
associated with progression-free survival in metastatic
colorectal patients with an advantage for ultra/poor
metabolizers versus normal metabolizers.
35
Up to now, no data were available on the correla-
tion between 5FUDR and DPD/MTHFR/TSER poly-
morphism and cape citabine-related toxicity specifically
in breast cancer patients. Hence, we carried out a ret-
rospective study aimed to evaluate the impact of each
of the fol lowing gene polymorphisms MTHFR C667T,
MTHFR A1298C, DPYD IVS14 þ 1G > A, TSER and
the 5-fluorouracil degradation rate (5-FUDR) on
toxicities in breast cancer patients treated with capeci-
tabine. Our hypothesis was that ultra/poor metabolizer
patients have a higher percentage of total adverse
events.
Our results suggest that 5FUDR is a possible
predictor of G3–4 toxicity in both metastatic and
non-metastatic breast cancer patients treated with
capecitabine. UMs and PMs patients developed
higher rate of severe toxicities compared with NM,
and these results are similar to our previous findings
on colorectal cancer patients treated with capecitabine,
Table 3. Incidence of toxicity by patients genotype and 5FUDR.
Biomarker Genotype N
Haematoxicity
(G1–4)
(%)
OR
(95% CI) P
GI toxicity
(G1–4)
(%)
OR
(95% CI) P
HFS
(G1–4)
(%)
OR
(95% CI) P
G3–4
Toxicity
(%)
OR
(95% CI) P
MTHFR
C677T
CC 11 18 91 35 36
CT 16 56 75 6 21
TT 6 66 6.5 (1.1–37.5) 0.04 83 0.3 (0.1–3.3) 0.35 20 0.2 (0.1–1.2) 0.08 17 0.4 (0.1–2.1) 0.30
MTHFR
A1298C
AA 15 53 73 21 29
AC 14 43 85 7 13
CC 5 20 0.5 (0.1–2.0) 0.33 100 3.1 (0.5–19.8) 0.23 40 0.7 (0.1–4.0) 0.67 40 0.6 (0.1–2.7) 0.50
TSER 3R/3R 7 43 86 0 29
2R/3R 12 42 83 25 29
2R/2R 13 54 1.2 (0.2–6.6) 0.81 77 0.6 (0.1–6.8) 0.73 23 1.5 (0.2–8.9) 0.65 14 0.7 (0.1–4.4) 0.68
5-FU
dRate
NM 28 43 1.3 (0.2–7.8) 86 22 16 10.4 (1.5–72.9)
PM 3 33 67 0.3 (0.1–2.4) 0 67
UM 3 67 0.75 67 0.28 0 0.0 0.99 67 0.02
Bold- significant difference in G3-4 toxicities between normal metabolizers (NM) and ultra/poor metabolizers (PM/UM) (p ¼ 0.02).
Table 2. 5FUDR descriptive statistics by demographic and
genetic characteristics.
N
5FUDR
(mean value SD) P
Age
<60 years 19 1.39 0.51 0.26
70 years 18 1.56 0.37
MTHFR C677T
CC 11 1.56 0.63 0.72
CT 19 1.42 0.39
TT 6 1.48 0.28
MTHFR A1298C
AA 17 1.57 0.45 0.14
AC 15 1.47 0.36
CC 5 1.12 0.57
TSER
3R3R 7 1.30 0.47 0.60
2R3R 14 1.51 0.41
2R2R 14 1.46 0.43
Botticelli et al. 5

Citations
More filters
Journal ArticleDOI
TL;DR: A comprehensive review of engineered nanoparticles and their payloads as promising tools for the treatment of breast cancer is presented in this article, which provides insights on the importance of engineered NPs and their methodology for validation as a next-generation platform with preventive and therapeutic effects against BC.
Abstract: Breast cancer (BC) is the second most common cancer in women globally after lung cancer. Presently, the most important approach for BC treatment consists of surgery, followed by radiotherapy and chemotherapy. The latter therapeutic methods are often unsuccessful in the treatment of BC because of their various side effects and the damage incurred to healthy tissues and organs. Currently, numerous nanoparticles (NPs) have been identified and synthesized to selectively target BC cells without causing any impairments to the adjacent normal tissues or organs. Based on an exploratory study, this comprehensive review aims to provide information on engineered NPs and their payloads as promising tools in the treatment of BC. Therapeutic drugs or natural bioactive compounds generally incorporate engineered NPs of ideal sizes and shapes to enhance their solubility, circulatory half-life, and biodistribution, while reducing their side effects and immunogenicity. Furthermore, ligands such as peptides, antibodies, and nucleic acids on the surface of NPs precisely target BC cells. Studies on the synthesis of engineered NPs and their impact on BC were obtained from PubMed, Science Direct, and Google Scholar. This review provides insights on the importance of engineered NPs and their methodology for validation as a next-generation platform with preventive and therapeutic effects against BC.

19 citations

Journal ArticleDOI
TL;DR: Issues and barriers to implementing precision dosing approaches for 5FU and capecitabine are identified and discussed with possible solutions proposed.
Abstract: Despite advances in targeted cancer therapy, the fluoropyrimidines 5-fluorouracil (5FU) and capecitabine continue to play an important role in oncology. Historically, dosing of these drugs has been based on body surface area. This approach has been demonstrated to be an imprecise way to determine the optimal dose for a patient. Evidence in the literature has demonstrated that precision dosing approaches, such as DPD enzyme activity testing and, in the case of intravenous 5FU, pharmacokinetic guided dosing, can reduce toxicity and yield better patient outcomes. However, despite the evidence, there has not been uniform adoption of these approaches in the clinical setting. When a drug such as 5FU has been used clinically for many decades, it may be difficult to change clinical practice. With the aim of facilitating change of practice, issues and barriers to implementing precision dosing approaches for 5FU and capecitabine are identified and discussed with possible solutions proposed.

10 citations


Cites background from "5-Fluorouracil degradation rate as ..."

  • ...is warranted.(76) Setting up processes for collection of samples and routine analysis would facilitate further work to expand monitoring beyond the current recommended dosage regimens detailed by IATDMCT....

    [...]

Journal ArticleDOI
TL;DR: The present review discussed the recent in vivo and in vitro studies for exploring the overexpression of oncogenes in the case of BC and the current status of newly discovered natural products in clinical investigations.
Abstract: Breast cancer (BC) is the second leading cause of death among women, and it has become a global health issue due to the increasing number of cases. Different treatment options, including radiotherapy, surgery, chemotherapy and anti-estrogen therapy, aromatase inhibitors, anti-angiogenesis drugs, and anthracyclines, are available for BC treatment. However, due to its high occurrence and disease progression, effective therapeutic options for metastatic BC are still lacking. Considering this scenario, there is an urgent need for an effective therapeutic strategy to meet the current challenges of BC. Natural products have been screened as anticancer agents as they are cost-effective, possess low toxicity and fewer side effects, and are considered alternative therapeutic options for BC therapy. Natural products showed anticancer activities against BC through the inhibition of angiogenesis, cell migrations, proliferations, and tumor growth; cell cycle arrest by inducing apoptosis and cell death, the downstream regulation of signaling pathways (such as Notch, NF-κB, PI3K/Akt/mTOR, MAPK/ERK, and NFAT-MDM2), and the regulation of EMT processes. Natural products also acted synergistically to overcome the drug resistance issue, thus improving their efficacy as an emerging therapeutic option for BC therapy. This review focused on the emerging roles of novel natural products and derived bioactive compounds as therapeutic agents against BC. The present review also discussed the mechanism of action through signaling pathways and the synergistic approach of natural compounds to improve their efficacy. We discussed the recent in vivo and in vitro studies for exploring the overexpression of oncogenes in the case of BC and the current status of newly discovered natural products in clinical investigations.

8 citations

Journal ArticleDOI
TL;DR: The synergistic interactions observed in MDA-MB-231 correlated with an activation of p38 MAPK, inhibition of FAK, increased expression of apoptogenic markers, prolongation of S-phase of cell cycle, and destabilization of actin network.
Abstract: Background: The combination effect of 5-fluorouracil (5-FU) with either CX-4945 or a new inhibitor of protein kinase CK2, namely 14B (4,5,6,7-tetrabromo-1-(3-bromopropyl)-2-methyl-1H-benzimidazole), on the viability of MCF-7 and triple-negative MDA-MB-231 breast cancer cell lines was studied. Methods: Combination index (CI) values were determined using an MTT-based assay and the Chou-Talalay model. The effect of the tested drug combinations on pro-apoptotic properties and cell cycle progression was examined using flow cytometry. The activation of FAK, p38 MAPK, and ERK1/2 kinases and the expression of selected pro-apoptotic markers in MDA-MB-231 cell line after the combined treatment were evaluated by the western blot method. Confocal microscopy was used to examine actin network in MDA-MB-231. Results: Our results showed that a synergistic effect (CI < 1) occurred in MDA-MB-231 after treatment with both combinations of 5-FU with 14B or CX-4945, whereas the combination of 5-FU and 14B evoked an antagonistic effect in MCF-7. We conclude that the synergistic interactions (CI < 1) observed for both the combinations of 5-FU and 14B or CX-4945 in MDA-MB-231 correlated with an activation of p38 MAPK, inhibition of FAK, increased expression of apoptogenic markers, prolongation of S-phase of cell cycle, and destabilization of actin network. Conclusions: The obtained results support the recent observation that CK2 inhibitors can improve 5-FU-based anticancer therapy and FAK kinase can be an attractive molecular target in breast cancer therapy.

8 citations


Cites background from "5-Fluorouracil degradation rate as ..."

  • ...Due to its cytotoxicity [8] and resistance occurrence, 5-FU is one of the most commonly used TS-directed anticancer drugs applied in combination with other chemotherapy compounds for the treatment of various cancers, including breast cancer [9,10]....

    [...]

Journal ArticleDOI
TL;DR: In this article, the authors used Drug-PIN® (Personalized Interactions Network) software in colorectal cancer patients to evaluate drug-drug-gene interactions (DDGIs), defined as the combination of DDIs and individual genetic polymorphisms.
Abstract: Drug–drug interactions (DDIs) can affect both treatment efficacy and toxicity. We used Drug-PIN® (Personalized Interactions Network) software in colorectal cancer (CRC) patients to evaluate drug–drug–gene interactions (DDGIs), defined as the combination of DDIs and individual genetic polymorphisms. Inclusion criteria were: (i) stage II-IV CRC; (ii) ECOG PS (Performance status sec. Eastern coperative oncology group) ≤2; (iii) ≥5 concomitant drugs; and (iv) adequate renal, hepatic, and bone marrow function. The Drug-PIN® system analyzes interactions between active and/or pro-drug forms by integrating biochemical, demographic, and genomic data from 110 SNPs. We selected DDI, DrugPin1, and DrugPin2 scores, resulting from concomitant medication interactions, concomitant medications, and SNP profiles, and DrugPin1 added to chemotherapy drugs, respectively. Thirty-four patients, taking a median of seven concomitant medications, were included. The median DrugPin1 and DrugPin2 scores were 42.6 and 77.7, respectively. In 13 patients, the DrugPin2 score was two-fold higher than the DrugPin1 score, with 7 (54%) of these patients experiencing severe toxicity that required hospitalization. On chi-squared testing for any toxicity, a doubled DrugPin2 score (p = 0.001) was significantly related to G3–G4 toxicity. Drug-PIN® software may prevent severe adverse events, decrease hospitalizations, and improve survival in cancer patients.

7 citations

References
More filters
Journal ArticleDOI
TL;DR: This research presents a novel and scalable approach to personalized medicine called “SmartPharm,” which combines cell reprograming, nanofiltration, and “computational medicine” to improve the quality and efficiency of individualized therapy treatment for cancer patients.
Abstract: Departments of Genetics, Bioengineering, Stanford University Medical Center, Stanford, California, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, North Carolina, USA and Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada, UK Correspondence to Dr Teri E. Klein, PhD, Department of Genetics, Stanford University Medical Center, 300 Pasteur D. Lane L301, Mail Code 5120, Stanford, CA 94305-5120, USA Tel: + 1 650 725 0659; fax: + 1 650 725 3863; e-mail: feedback@pharmgkb.org

99 citations


"5-Fluorouracil degradation rate as ..." refers background in this paper

  • ...Several enzymes are involved in the capecitabine metabolism.(19) The dihydropyrimidine dehydrogenase enzyme (DPD) metabolizes about 80% of the administered 5-FU into the inactive metabolite 5,6-dihydro-5fluorouracil....

    [...]

Journal ArticleDOI
TL;DR: This study suggests that common genetic variation in MTHFR but not TYMS may be useful for predicting toxicity from capecitabine in patients with advanced colorectal cancer.
Abstract: Purpose: To evaluate the effect of thymidylate synthase ( TYMS ) and methylenetetrahydrofolate reductase ( MTHFR ) genotypes on toxicity in patients treated with capecitabine for advanced colorectal cancer and to determine the effect of these polymorphisms on the pretreatment levels of serum folate and plasma homocysteine. Experimental Design: Fifty-four patients with a diagnosis of metastatic colorectal cancer were treated with fixed-dose capecitabine. Germ line DNA from patients was genotyped for TYMS TSER, TSER*3G>C, and 3′-untranslated 6 bp insertion/deletion (3′ untranslated region insertion/deletion), and MTHFR c.677C>T and c.1298A>C using PCRs and RFLP. Toxicity was graded by National Cancer Institute Common Toxicity Criteria version 2.0. Response was assessed by Response Evaluation Criteria in Solid Tumors. Results: MTHFR c.677C>T and c.1298A>C genotypes and diplotypes predicted for grade 2/3 toxicities, whereas the TYMS genotypes had no influence. MTHFR c.677 genotype tended to predict overall survival ( P = 0.08). MTHFR c.677 influenced pretreatment homocysteine ( P < 0.05) and serum folate levels ( P < 0.05). Multivariate analysis suggests that MTHFR c.1298 is an independent predictor of toxicity. Conclusions: This study suggests that common genetic variation in MTHFR but not TYMS may be useful for predicting toxicity from capecitabine in patients with advanced colorectal cancer. In addition, MTHFR single nucleotide polymorphisms predicted serum folate and plasma homocysteine levels, and, combined, these factors may be important predictors of capecitabine-induced toxicity.

93 citations


"5-Fluorouracil degradation rate as ..." refers background in this paper

  • ...Other enzymes are involved in 5-FU metabolism, and their polymorphisms could result in increased and unexpected toxicities such as MTHFR, one of the most relevant enzyme that regulates intracellular folate levels that affect DNA synthesis and methylation and TYMS.(24,25) The single-nucleotide polymorphisms (SNPs) of MTHFR 677C>T and 1298A>C are clinically relevant and have been associated with the toxicity of 5-FU....

    [...]

  • ...Other enzymes are involved in 5-FU metabolism, and their polymorphisms could result in increased and unexpected toxicities such as MTHFR, one of the most relevant enzyme that regulates intracellular folate levels that affect DNA synthesis and methylation and TYMS.24,25 The single-nucleotide polymorphisms (SNPs) of MTHFR 677C>T and 1298A>C are clinically relevant and have been associated with the toxicity of 5-FU.26 Moreover, variations of the TSER in the promoter of TYMS gene have been related to both survival/response outcomes and toxicities in patients affected by colorectal cancer treated with 5-FU-based chemotherapy.27–30 Finally, we previously described a non-genomic assay that seems to be able to predict 5-FU toxicity by the assessment of the 5FUDR in the PBMC.31 This parameter indicates the amount of drug consumed by cells in a time unit and reflexes the result of the entire 5-FU degradation metabolism, not only a single enzyme activity.32 Applying the assay on colorectal patients, we previously described two different classes of patients with a higher risk to develop 5-FU unexpected toxicities: poor metabolizers and ultrarapid metabolizers.33,34 Besides, we showed that 5FUDR is associated with progression-free survival in metastatic colorectal patients with an advantage for ultra/poor metabolizers versus normal metabolizers.35 Up to now, no data were available on the correlation between 5FUDR and DPD/MTHFR/TSER polymorphism and capecitabine-related toxicity specifically in breast cancer patients....

    [...]

Journal ArticleDOI
TL;DR: It is reported that clinical response to FOLFOX is significantly related to methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms, which corroborate the observation that response was related to the cumulative FU dose, whereas progression-free survival was relatedto the cumulative oxaliplatin dose.
Abstract: WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT • Numerous clinical studies, including a few prospective ones, have reported conflicting results on the impact of gene polymorphisms related to fluorouracil (FU) and oxaliplatin pharmacodynamics. WHAT THIS STUDY ADDS • This prospective study is the first to report that clinical response to FOLFOX is significantly related to methylenetetrahydrofolate reductase (MTHFR) gene polymorphisms (677CT and 1298AC), with a response rate of 37, 53, 63 and 80% in patients harbouring no, one, two or three favourable MTHFR alleles, respectively. • Only polymorphisms of genes related to oxaliplatin pharmacodynamics (GSTπ 105IleVal and XPD 751LyGln) influenced progression-free survival. • These results corroborate the observation that response was related to the cumulative FU dose, whereas progression-free survival was related to the cumulative oxaliplatin dose. AIMS To test prospectively the predictive value of germinal gene polymorphisms related to fluorouracil (FU) and oxaliplatin (Oxa) pharmacodynamics on toxicity and responsiveness of colorectal cancer (CRC) patients receiving FOLFOX therapy. METHODS Advanced CRC patients (n= 117) receiving FOLFOX 7 therapy were enrolled. Gene polymorphisms relevant for FU [thymidylate synthase (TYMS, 28 bp repeats including the GC mutation + 6 bp deletion in 3'UTR), methylenetetrahydrofolate reductase (MTHFR, 677CT, 1298AC), dihydropyrimidine deshydrogenase (IVS14+1GA) and Oxa: glutathione S-transferase (GST) π (105IleVal, 114AlaVal), excision repair cross-complementing group 1 (ERCC1) (118AATAAC), ERCC2 (XPD, 751LysGln) and XRCC1 (399ArgGln)] were determined (blood mononuclear cells). RESULTS None of the genotypes was predictive of toxicity. Response rate (54.7% complete response + partial response) was related to FU pharmacogenetics, with both 677CT (P= 0.042) and 1298AC (P= 0.004) MTHFR genotypes linked to clinical response. Importantly, the score of favourable MTHFR alleles (677T and 1298C) was positively linked to response, with response rates of 37.1, 53.3, 62.5 and 80.0% in patients bearing no, one, two or three favourable alleles, respectively (P= 0.040). Polymorphisms of genes related to Oxa pharmacodynamics showed an influence on progression-free survival, with a better outcome in patients bearing GSTπ 105 Val/Val genotype or XPD 751Lys-containing genotype (P= 0.054). CONCLUSIONS These results show that response to FOLFOX therapy in CRC patients may be driven by MTHFR germinal polymorphisms.

92 citations


"5-Fluorouracil degradation rate as ..." refers background in this paper

  • ...Other enzymes are involved in 5-FU metabolism, and their polymorphisms could result in increased and unexpected toxicities such as MTHFR, one of the most relevant enzyme that regulates intracellular folate levels that affect DNA synthesis and methylation and TYMS.(24,25) The single-nucleotide polymorphisms (SNPs) of MTHFR 677C>T and 1298A>C are clinically relevant and have been associated with the toxicity of 5-FU....

    [...]

  • ...Other enzymes are involved in 5-FU metabolism, and their polymorphisms could result in increased and unexpected toxicities such as MTHFR, one of the most relevant enzyme that regulates intracellular folate levels that affect DNA synthesis and methylation and TYMS.24,25 The single-nucleotide polymorphisms (SNPs) of MTHFR 677C>T and 1298A>C are clinically relevant and have been associated with the toxicity of 5-FU.26 Moreover, variations of the TSER in the promoter of TYMS gene have been related to both survival/response outcomes and toxicities in patients affected by colorectal cancer treated with 5-FU-based chemotherapy.27–30 Finally, we previously described a non-genomic assay that seems to be able to predict 5-FU toxicity by the assessment of the 5FUDR in the PBMC.31 This parameter indicates the amount of drug consumed by cells in a time unit and reflexes the result of the entire 5-FU degradation metabolism, not only a single enzyme activity.32 Applying the assay on colorectal patients, we previously described two different classes of patients with a higher risk to develop 5-FU unexpected toxicities: poor metabolizers and ultrarapid metabolizers.33,34 Besides, we showed that 5FUDR is associated with progression-free survival in metastatic colorectal patients with an advantage for ultra/poor metabolizers versus normal metabolizers.35 Up to now, no data were available on the correlation between 5FUDR and DPD/MTHFR/TSER polymorphism and capecitabine-related toxicity specifically in breast cancer patients....

    [...]

Journal ArticleDOI
TL;DR: TYMS and UGT1A polymorphisms influence on tumour response and toxicities derived from irinotecan/5FU treatment in CRC patients is investigated and a genetic-based algorithm to optimise treatment individualisation is proposed.
Abstract: The impact of thymidylate synthase (TYMS) and UDP-glucoronosyltransferase 1A (UGT1A) germline polymorphisms on the outcome of colorectal cancer (CRC) patients treated with irinotecan plus 5-fluorouracil (irinotecan/5FU) is still controversial. Our objective was to define a genetic-based algorithm to select patients to be treated with irinotecan/5FU. Genotyping of TYMS (5′TRP and 3′UTR), UGT1A1*28, UGT1A9*22 and UGT1A7*3 was performed in 149 metastatic CRC patients treated with irinotecan/5FU as first-line chemotherapy enrolled in a randomised phase 3 study. Their association with response, toxicity and survival was investigated by univariate and multivariate statistical analysis. TYMS 3TRP/3TRP genotype was the only independent predictor of tumour response (OR=5.87, 95% confidence interval (CI)=1.68–20.45; P=0.005). UGT1A1*28/*28 was predictive for haematologic toxicity (OR=6.27, 95% CI=1.09–36.12; P=0.04), specifically for neutropenia alone (OR=6.40, 95% CI=1.11–37.03; P=0.038) or together with diarrhoea (OR=18.87, 95% CI=2.14–166.67; P=0.008). UGT1A9*1/*1 was associated with non-haematologic toxicity (OR=2.70, 95% CI=1.07–6.82; P=0.035). Haplotype VII (all non-favourable alleles) was associated with non-haematologic toxicity (OR=2.11, 95% CI=1.12–3.98; P=0.02). TYMS and UGT1A polymorphisms influence on tumour response and toxicities derived from irinotecan/5FU treatment in CRC patients. A genetic-based algorithm to optimise treatment individualisation is proposed.

85 citations

Journal ArticleDOI
TL;DR: It is concluded that patients prefer the regimen with less toxicity and that it is of minor importance whether the medication is administrated orally at home or i.v. at the hospital.

75 citations


"5-Fluorouracil degradation rate as ..." refers background in this paper

  • ...Oral administration seems to be preferred by patients and allows to avoid complications and costs linked to 5-FU IV infusion.(3)...

    [...]

Frequently Asked Questions (1)
Q1. What are the contributions in "5-fluorouracil degradation rate as a predictive biomarker of toxicity in breast cancer patients treated with capecitabine" ?

Genetic polymorphisms and the 5-fluorouracil degradation rate of breast cancer patients treated with capecitabine were retrospectively studied. Genetic markers and the 5-fluorouracil degradation rate were correlated with the reported toxicities. Thirty-seven patients with a median age of 58 years old treated with capecitabine for stages II–IV breast cancer were included in this study.