You are on page 1of 10

Pharmacological Research 106 (2016) 27–36

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Review

Cisplatin resistance and opportunities for precision medicine


Lauren Amable ∗
National Institute on Minority Health and Health Disparities, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States

a r t i c l e i n f o a b s t r a c t

Article history: Cisplatin is one of the most commonly used chemotherapy drugs, treating a wide range of cancer
Received 28 December 2015 types. Unfortunately, many cancers initially respond to platinum treatment but when the tumor returns,
Accepted 1 January 2016 drug resistance frequently occurs. Resistance to cisplatin is attributed to three molecular mechanisms:
Available online 22 January 2016
increased DNA repair, altered cellular accumulation, and increased drug inactivation. The use of preci-
sion medicine to make informed decisions on a patient’s cisplatin resistance status and predicting the
Keywords:
tumor response would allow the clinician to tailor the chemotherapy program based on the biology of
Cisplatin resistance
the disease. In this review, key biomarkers of each molecular mechanism will be discussed along with
Nucleotide excision repair
ERCC1
the current clinical research. Additionally, known polymorphisms for each biomarker will be discussed
Copper transporters in relation to their influence on cisplatin resistance.
ABC transporters Published by Elsevier Ltd.
Glutathione

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
2. Altered DNA repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2.1. ERCC1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2.2. Other NER genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
3. Altered accumulation of cisplatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
3.1. Decreased cellular uptake of cisplatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
3.1.1. Copper transporters CTR1 and CTR2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
3.1.2. Organic cation transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
3.2. Increased cellular export of cisplatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
3.2.1. P-type ATPase transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
3.2.2. ATP-binding cassette (ABC) transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4. Cytosolic inactivation of cisplatin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.1. Inactivation by glutathione conjugation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.1.1. Glutathione-S-transferase Pi 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.1.2. Glutathione-S-transferase Mu . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
4.2. Inactivation by metallothionein binding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
5. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33

Abbreviations: ABC, ATP-binding cassette; ASE-1, anti-sense ERCC1; ATP7A, ATPase copper-transporting alpha polypeptide; ATP7B, ATPase copper-transporting beta
polypeptide; CAST, T-cell receptor complex subunit CD3␧-associated signal transducer; CTR1, copper transporter 1; CTR2, copper transporter 2; ERCC1, excision repair
cross-complementation group 1 gene; GSH, glutathione; GST, glutathione-s-transferase; MRP, multidrug resistance associated protein; MT, metallothionein; NER, nucleotide
excision repair; NSCLC, non-small cell lung cancer; OCT, organic cation transporter; PCNA, proliferating cell nuclear antigen; Pol, polymerase; SLC, solute carrier; TFIIH,
transcription factor II H; UTR, untranslated region; XPA, xeroderma pigmentosum group A; XPB, xeroderma pigmentosum group B; XPD, xeroderma pigmentosum group D;
XPE, xeroderma pigmentosum group E; XPF, xeroderma pigmentosum group F; XPG, xeroderma pigmentosum group G.
∗ Corresponding author. Fax: +1 301 480 4490.
E-mail address: lauren.amable@nih.gov

http://dx.doi.org/10.1016/j.phrs.2016.01.001
1043-6618/Published by Elsevier Ltd.
28 L. Amable / Pharmacological Research 106 (2016) 27–36

1. Introduction The second issue associated with cisplatin treatment is resis-


tance to therapy. Initially the tumor responds to cisplatin but then
Rosenberg et al., first discovered in Escherichia coli that the the tumor comes back and is frequently refractory to further plat-
byproducts from platinum electrode activity resulted in the inhibi- inum therapy. There are two forms of resistance found in the clinic:
tion of cell division [1,2]. Within 15 years, cisplatin was approved innate and acquired. Innate resistance is resistance without out any
for the treatment of cancer by the FDA. Cisplatin is one of the most prior drug exposure. Acquired resistance is a result of drug expo-
widely used anticancer drugs in North America and Europe [3], sure. The differences between innate and acquired resistance are
treating a variety of cancers including: testicular, ovarian, non- not clear but it is generally thought that each operates through
small cell lung cancer (NSCLC), head and neck cancer, bladder, different signaling pathways. This review will only focus on resis-
gastric, and other malignancies [4]. The main issue with obtaining tance as a whole since discerning between the two requires further
the optimum cisplatin cancer treatment is the significant interpa- studies.
tient variability with outcome, efficacy, and toxicity. In the clinic, the definition of whether a patient is sensitive or
There are two problems associated with cisplatin usage in the resistant to cisplatin is generally as follows. If a patient is more than
clinic: toxicity and resistance. Cisplatin has a numerous toxicities two years from the last platinum dose, the patient is considered
including renal damage, deafness, and peripheral neuropathy, thus sensitive. There is a greater than 70% likelihood that the patient
the overall efficacy of the drug could not be reached due to the side will respond to treatment with platinum-based therapy [6]. The
effects. This has led to the development of cisplatin analogs that percentage of patients who will respond to cisplatin decreases with
would be clinically effective but without the toxicity. Carboplatin the shortening of the disease free period. Patients who have disease
and oxaliplatin (Fig. 1), are the most popular analogs and reached recurrence within the first months after the recent platinum dose
FDA approval for usage. Interestingly, there is a variation in the can- will have a low likelihood of treatment response with cisplatin and
cers treated by the cisplatin analogs. Carboplatin is not as effective are considered to have platinum resistant disease.
in treating germ cell malignancies compared to cisplatin. Oxali- When cisplatin is transported into the cell, cisplatin has several
platin is very effective for the treatment of colon cancer, a cancer fates (Fig. 2). First, cisplatin can be exported from the cell using a
where cisplatin is not effective. Understanding the molecular basis transmembrane transporter system. Second, cisplatin can be chem-
for the difference between these three compounds could provide ically neutralized by binding sulfhydryl groups in proteins such as
new insights and unlock novel mechanisms into how cancer cells glutathione or metallothioneins. Finally, cisplatin nonspecifically
counteract the effects of DNA-damaging drugs. While the analogs reacts with a variety of subcellular components: proteins, RNA, and
show hope for a better response with less toxicity the scope of this DNA. RNA is most sensitive to react with cisplatin, followed by DNA,
review will not cover cisplatin analog resistance. For a review on and then protein. The primary and widely accepted mechanism of
cisplatin analog resistance, the author directs the reader to a recent action for cisplatin is the binding to cellular DNA, resulting in DNA-
review from Perego and Robert [5]. platinum adducts. This prevents the cell from replicating its DNA

Fig. 1. Cisplatin and analogs.

Fig. 2. Cellular fate of cisplatin (Pt). Cisplatin crosses the cell membrane by passive diffusion or by transmembrane transporters. CTR1, CTR2, and OCT2 have been identified
as transporters that import cisplatin into the cell. Once inside the cell, cisplatin binds to DNA to cause DNA-platinum adducts. The damage is repaired by ERCC1 and members
of the NER pathway. Cisplatin is also inactivated by glutathione-s-transferase, which add a glutathione (GSH) to cisplatin. The conjugated cisplatin-GSH is then exported via
the MRP2 transporters. Cisplatin is also exported by ATP7A and ATP7B. Inactivation of cisplatin can also result from binding metallothionein proteins (MT).
L. Amable / Pharmacological Research 106 (2016) 27–36 29

until the damage is repaired. If the cell cannot repair the DNA or
the damage is too severe, then the cell dies.
Resistance to cisplatin occurs by the following molecular mech-
anisms: altered DNA repair, altered cellular accumulation of drug,
and cytosolic inactivation of drug. The processes of resistance were
studied in L1210 mouse leukemia cells and human ovarian cancer
cells [7–10]. The observations were similar in both cell models: all
three components contributed to cisplatin resistance. There were
differences of each mechanism regarding the relative contribu-
tions. At low levels of cisplatin resistance, about 10–15 fold above
baseline, the primary mechanism of resistance was DNA repair.
Intermediate levels of resistance, up to 40–50 fold over baseline,
was due to reduced cellular cisplatin accumulation. At very high
levels of resistance, cytosolic inactivation of cisplatin was the pri-
mary mechanism. However, in many cell lines it has been observed
that more than one mechanism can be occurring.
The goal of precision medicine is to generate better responses
in the clinic. Making an informed decision on predicting the tumor
response to cisplatin as well as the type of resistance that is occur-
ring allows for tailoring the chemotherapy program based on the
biology of the disease. Here in this review, we will comprehen-
sively discuss the mechanisms of cisplatin resistance-altered DNA
repair, altered cellular accumulation, and drug inactivation. For
each mechanism, the most promising biomarkers identified so far
will be discussed and are summarized in Table 1. Polymorphisms
of each biomarker that correlate with cisplatin resistance from cur-
rent clinical studies will also be presented, and are summarized in
Table 2.

2. Altered DNA repair

Once inside the cell, cisplatin binds to DNA and forms adducts.
The primary form of DNA damage are N7-d(GpG) and N7-d(ApG)
intrastrand DNA-platinum adducts. These bulky adducts result Fig. 3. Schematic of nucleotide excision repair (NER). DNA-platinum adducts are
in substantial kinking of the DNA (12), which is recognized and removed by the NER pathway. First the DNA-platinum adduct is detected. Then
repaired by the nucleotide excision repair (NER) pathway, shown the damage is verified and the pre-incision complex is set up containing RPA, XPA,
in Fig. 3. In this pathway, which requires more than 30 proteins, the and XPG. DNA is unwound by XPB and XPD. XPF-ERCC1 and XPG create incisions 5
and 3 from the damaged base. The oligonucleotide containing the damaged base is
DNA-platinum adduct is first recognized by XPE and XPC-DDB1/2.
removed. The gap is filled in by DNA repair synthesis complex: RPA, RFC, PCNA, and
The transcription factor II H (TFIIH) complex verifies the damage Pol ␦/␧. Finally, the DNA is ligated.
and assembles the pre-incision complex: RPA, XPA, and XPG. The
DNA is then unwound by the XPB and XPD helicases. ERCC1-XPF
another study comparing the six histologic types of ovarian cancer,
and XPG endonucleases create an excision several bases upstream
there is an upregulation of NER genes that correlates with cisplatin
and downstream from the DNA-platinum adduct. This releases the
resistance. Clear cell tumors are known to be the most chemore-
oligonucleotide containing the adduct. The gap is filled in by the
sistant to cisplatin, and they displayed the highest levels of ERCC1
DNA repair synthesis complex containing RPA, RFC, PCNA, and Pol
[14]. The evaluation of ERCC1’s potential role as a cisplatin resis-
␦/␧. In the final step, the DNA is ligated by DNA ligase I, thus com-
tance biomarker has been explored in other cancers. High ERCC1
pleting the DNA repair. The balance of DNA damage and DNA repair
levels that correlate with increased resistance to cisplatin have
dictates death versus survival after cisplatin therapy [11]. Changes
been observed in: ovarian [15,16], NSCLC [15,17–23], nasopha-
in the ability to repair the DNA adducts results in changes in cis-
ryngeal [24,25], esophageal [26], cervical [27,28], head and neck
platin sensitivity.
squamous carcinoma [29,30], liver [31], osteosarcoma [32], lung
adenocarcinoma [33], advanced biliary tract adenocarcinoma [34],
2.1. ERCC1 mesothelioma [35], pulmonary adenocarcinoma [36], and gastric
[11]. Thus, the expression of ERCC1 makes an attractive biomarker
ERCC1 is one of the most highly studied biomarkers for cisplatin for cisplatin resistance since the increased expression has been
resistance to date. The DNA damage excision step is catalyzed by observed in a variety of cancer types.
the ERCC1-XPF dimer is the rate limiting step in the NER path- There are two polymorphisms of ERCC1 that appear to have clin-
way. High ERCC1 levels are associated with increased removal of ical significance with sensitivity to cisplatin treatment. The first
DNA-platinum adducts and results in increased resistance to cis- polymorphism, rs11615, is located in codon 118, and codes for the
platin [12]. There is a linear correlation of ERCC1 expression and same amino acid-asparagine, was first described by Yu et al. [37,38].
cisplatin sensitivity, with resistant cells expressing more ERCC1 A point to note for this polymorphism: there is a discrepancy in
compared to sensitive. It was first demonstrated in ovarian can- the literature on whether the change is from C to T or T to C. The
cer that ERCC1 levels are increased in cancer tissue in comparison reader should take caution in evaluating studies, as these alleles are
to normal [13]. Even higher levels of ERCC1 mRNA are found in switched in some analyses. Here this review the rs11615 polymor-
patients with clinically resistant cancer. Lower levels are found phism is from C to T. The rs11615, or N118N, polymorphism was
in patients that are clinically sensitive to platinum therapy. In originally thought to result in reduced levels of ERCC1 mRNA and
30 L. Amable / Pharmacological Research 106 (2016) 27–36

Table 1
Proteins associated with cisplatin resistance.

Protein Relationship to resistance Cancer References

NER
ERCC1 Increased expression Ovarian, NSCLC, nasopharyngeal, esophageal, [13–16], [15], [17–23], [24,25], [26], [27,28],
cervical, head and neck squamous carcinoma, [29,30], [31], [32], [33], [34], [35], [36], [11]
liver, osteosarcoma, lung adenocarcinoma,
biliary tract adenocarcinoma, mesothelioma,
pulmonary adenocarcinoma, gastric
XPA Increased expression Ovarian cancer [53,54]
XPB Increased expression Ovarian cancer [53,54]
XPF Increased expression Ovarian and colon cancer cell lines; head and [55–57]
neck carcinoma
XPD Increased expression NSCLC and glioma cell lines [58,59]

Cellular uptake
CTR1 Decreased expression Ovarian cancer, NSCLC [73–75]
CTR2 Increased expression Ovarian cancer [80,81]
OCT2 No change Ovarian cancer [83]
Decreased expression Gastric cancer [84]

Cellular export
ATP7A Increased expression NSCLC, ovarian cancer [87,89,90]
ATP7B Increased expression Gastric, hepatocellular, esophageal, oral, [93–101]
breast, endometrial, lung, ovarian cancer
MRP2 Increased expression Colorectal, esophageal, hepatocellular cancer [111–113]

Drug inactivation
GSTP1 Increased expression Ovarian cancer, head and neck carcinoma, [122–124]
NSCLC
No change Ovarian, cervical cancer [130–132]
MT Increased expression Esophageal, ovarian cancer [151,123]

Table 2
Gene polymorphisms associated with cisplatin resistance.

Gene Polymorphism Response to cisplatin Cancer Reference

NER
ERCC1 rs11615, N118N Increased response Ovarian cancer, colorectal, pancreatic, osteosarcoma and NSCLC [38,40,42–45]
Decreased response NSCLC [46,47]
C8092A Increased response NSCLC, esophageal [49,50]
Decreased response Nasopharyngeal, mesothelioma [51,35]
No relationship NSCLC [52]
XPD Asp312Asn Increased response NSCLC, osteosarcoma, pancreatic cancer [54,60,62,63]

Cellular uptake
CTR1 rs7851395, rs12686377 Increased response NSCLC [76]
OCT2 rs195854, rs186941 Increased response NSCLC [85]

Cellular export
MRP2 C-24T Increased response NSCLC [116,117]

Drug inactivation
GSTM1 rs10431718 Increased response Lymphoblastoid cell lines [143]
Null allele Increased response NSCLC [144]

protein, as the codon is an infrequently used codon [37–39]. A clini- evaluated. Studies evaluating the C8092A polymorphism are few
cal molecular correlative study in ovarian cancer was confirmatory and are additionally associated with conflicting data. The clinical
[40] but in another recent study it was shown to not change the implication of the C8092A ERCC1 polymorphism has been studied
expression or function of ERCC1 but rather may be linked to other in lung [49] and esophageal cancer [50] and both studies demon-
causative variants [41]. There are conflicting data as to whether or strated the A polymorphism results in increased cisplatin response.
not this polymorphism determines sensitivity to cisplatin. In ovar- There is also conflicting data as to the A allele and clinical resistance
ian cancer, the T allele was associated with an increased response to platinum-based therapy. In a nasopharyngeal cancer study, the
to cisplatin therapy [40]. This was also observed in colorectal can- A allele of C8092A was associated with an increased risk of dis-
cer [42], pancreatic cancer [43], osteosarcoma [44] and NSCLC [45]. ease progression with patients on cisplatin-based chemotherapy
Yet in another study, the C allele was associated with a higher [51]. In a malignant pleural mesothelioma study, the A polymor-
response rate to cisplatin, progression free survival, and overall phism of C8092A was associated with a shorter progression free
survival [46,47]. Thus two opposite results have been observed. survival [35]. Yet, in a meta-analysis of 39 NSCLC studies, there
The second ERCC1 polymorphism relating to cisplatin sensitivity was no relationship of survival and sensitivity to treatment with
is C8092A. This polymorphism was first identified in gliomas and is platinum-based chemotherapy [52].
located in the 3 UTR of ERCC1 [48]. The A allele is thought to result
in decreased mRNA stability of ERCC1. This polymorphism results in 2.2. Other NER genes
an A substitution in two additional genes, nucleolar protein ASE-1
and t-cell receptor complex subunit CD3␧-associated signal trans- While the majority of the studies have focused on ERCC1, there
ducer (CAST) [48]. Thus the exact role of this polymorphism is not are several studies that suggest other NER genes are involved in
fully understood as these genes may have an effect that has not been cisplatin resistance. Dabholkar et al., showed that other NER genes
L. Amable / Pharmacological Research 106 (2016) 27–36 31

are additionally upregulated in patients who responded to cisplatin cisplatin [69]. CTR1 primarily transports copper, which is impor-
therapy [53]. XPA, which is part of the pre-incision complex, and tant in a variety of biological functions within a cell. Interestingly,
XPB, a helicase, displayed increased expression in cisplatin resis- resistance to cisplatin is accompanied by resistance to copper and
tant ovarian cancer tumors [53,54]. However, this has not been cisplatin resistant cells display reduced levels of copper [70]. Cis-
explored further in other clinical studies. Neither XPA nor XPB platin resistant cells show decreased levels of CTR1 mRNA and
polymorphisms have been discovered that correlate with cisplatin decreased cisplatin uptake [71,72]. In the clinic, CTR1 has been eval-
resistance. uated in two cancer types, ovarian and lung, both resulting in the
It is logical to think that XPF would be an additional cisplatin same observation. In ovarian cancer patients treated with cisplatin
resistance marker since it is dimerizes with ERCC1 to catalyze the chemotherapy, high levels of CTR1 mRNA expression was corre-
incision of the damaged DNA strand. However, XPF has been over- lated with increased disease free survival [73]. In NSCLC, the same
looked in many studies as to whether or not it is a valid biomarker pattern has emerged, high CTR1 protein levels were associated with
for cisplatin resistance. In ovarian and colon cancer cell lines, the a favorable cisplatin response [74,75].
increased protein expression of XPF was correlated with increased Only one study has examined the relationship of CTR1
cisplatin resistance [55]. There have only been two clinical stud- polymorphisms with cisplatin resistance. Xu et al. found two
ies that have examined XPF [56,57]. Both studies examined head polymorphisms in CTR1 that correlate with platinum resistance
and neck cancer and increased XPF expression was correlated with and survival: rs7851395 and rs12686377 [76]. These two poly-
cisplatin resistance. Vaezi et al. went on further to examine XPF morphisms are located in the intron region of CTR1 and are
polymorphisms, however the four polymorphisms they identified hypothesized to play a role in the epigenetic regulation of CTR1.
showed marginal association with treatment failure [57]. Further Currently, it is not known what effect these polymorphisms have
studies examining the role of XPF in other cancers are needed. on the function of CTR1, as this article was the first to describe them.
The helicase XPD was additionally identified to have a strong Cisplatin is also transported by another copper transporter,
correlation between increased expression and cisplatin resistance CTR2. While CTR2 is in the same family as CTR1, they only share
in NSCLC and glioma cell lines [58,59]. The expression of XPD has a 33% homology on the protein level [77]. CTR2 is found on the
not been evaluated in clinical samples. The majority of the clinical cellular membrane like CTR1, but it is also found on intracellular
studies have examined the effects of XPD polymorphisms on cis- organelle membranes and may have alternative cellular functions
platin resistance. Two polymorphisms in XPD have been identified, [78]. The opposite effect has been observed with CTR2 in terms of
Asp312Asn and Lys751Gln, both result in decreased DNA repair its correlation with cisplatin resistance. Unlike CTR1, knockdown
capacity. Both polymorphisms were found to be potential biomark- of CTR2 in cells results in increased uptake, increased cytotoxicity,
ers for clinical outcome in osteosarcomas and lung cancers treated and increased sensitivity to cisplatin [79]. In two clinical studies,
with cisplatin [44,60]. The Asp312Asn polymorphism was associ- lower levels of CTR2 are associated with a better outcome to cis-
ated with a better survival in osteosarcoma patients treated with platin therapy [80,81]. Interestingly, it has been suggested that the
cisplatin [61]. The Lys751Gln polymorphism was associated with CTR1/CTR2 ratio may be a useful biomarker for identifying tumors
longer progression free survival in pancreatic and NSCLC [62,63]. which may be more sensitive to cisplatin than on one of the trans-
porters alone [81]. No CTR2 polymorphisms have been identified
3. Altered accumulation of cisplatin to influence cisplatin sensitivity.

The second mechanism of cisplatin resistance is altered cellu- 3.1.2. Organic cation transporters
lar accumulation of cisplatin. It has long been noted that cisplatin The solute carrier (SLC) transporter family, specifically the SLC22
resistant cells tend to exhibit decreased levels of cisplatin [64]. Tis- family members, also transport cisplatin into cells. Members of
sue platinum concentrations are correlated with percent reduction this family, OCT1, OCT2, and OCT3, have been shown to uptake
of the tumor, meaning reduced tissue platinum concentrations are platinum-compounds into cells, but each transporter varies in the
associated with resistance [65]. Altered accumulation of cisplatin is expression and substrate [68]. OCT1 has been indicated to trans-
the result of two independent cellular pathways: decreased uptake port cisplatin, however the evidence is weak [68]. OCT1 transports
or increased export. the cisplatin analogs carboplatin and oxaliplatin. OCT3 is known to
primarily transport oxaliplatin.
3.1. Decreased cellular uptake of cisplatin Cisplatin is primarily transported by OCT2, or SLC22A2, and this
transporter is found in the kidney. OCT2 transfection into cells
Cisplatin has a simple chemistry, the core is a single platinum results in increased cellular levels of cisplatin [82]. There is not
metal bound to two amino groups and two chlorides (Fig. 1). At a lot of clinical studies examining OCT2’s role in cisplatin resis-
physiologic pH, the chlorides of cisplatin are replaced with OH tance, primarily due to the fact it is expressed in the kidney. In the
molecules, resulting in a neutral charge. This it makes it possi- NCI-60 panel of cancer cell lines, OCT2 was the most frequently
ble for diffusion across the cellular membrane, flowing from the expressed gene but its expression in clinical ovarian cancer spec-
high concentration of cisplatin outside to the lower concentration imens was low and did not correlate with the treatment outcome
inside the cell. Thus, cisplatin uptake was first thought to be via of a platinum-based regimen [83]. In one gastric cancer study,
passive diffusion as uptake was not saturated against time or drug higher levels of OCT2 were observed in responders to cisplatin-
concentrations, up to 3 mM [66,67]. However, it was discovered based neoadjuvant therapy in comparison to non-responders [84].
that cisplatin mostly enters the cell by membrane transporters. The majority of OCT2 polymorphism studies have primarily focused
This would explain why it has been observed that low levels of on the effect on cisplatin nephrotoxicity. There is one study in
transporters correlate with decreased levels of cellular cisplatin. lung cancer that evaluated the OCT2 polymorphisms. The poly-
Cisplatin uptake is performed by the copper transporters CTR1 and morphisms rs195854 and rs186941 were associated with increased
CTR2, as well as the organic cation transporter (OCT) family [68]. response to cisplatin [85].

3.1.1. Copper transporters CTR1 and CTR2 3.2. Increased cellular export of cisplatin
Copper transporter protein 1 (CTR1) was shown to be one of
the primary cisplatin transporters. It was first discovered in yeast, The export of cisplatin has been suggested to occur via passive
noting that knocking down CTR1 resulted in reduced uptake in efflux, however the issue with studies to examine this phenom-
32 L. Amable / Pharmacological Research 106 (2016) 27–36

ena are performed with high concentration of cisplatin. Thus it is not predict cisplatin response [100,114,115]. One polymorphism
thought that export of cisplatin from cells occurs via membrane in MRP2, C-24T, has been correlated with increased response to
transporters. There are two major pathways in which cisplatin is platinum-based chemotherapy in lung cancer [116,117]. The C-24T
removed from the cell: removal by P-type ATPase transporters or polymorphism is found in the promoter of MRP2 and its function
removal by ATP-binding cassette transporters. is not currently known.

3.2.1. P-type ATPase transporters 4. Cytosolic inactivation of cisplatin


Cisplatin is exported by ATP7A and ATP7B, which belong to the
transporter family of P-type ATPases which use ATP to export. These Finally, the last resistance mechanism is cytosolic inactivation
transporters are associated with removing excessive copper from of cisplatin. This inactivation results in the inability of cisplatin
cells. Under normal conditions within the cell, ATP7A and ATP7B to react with DNA. Less damage is produced and the cancer cell
are found in the trans-Golgi network and are trafficked to the cell survives the drug treatment. The primary form of inactivation is
membrane to remove copper. As mentioned earlier, copper levels conjugation of cisplatin with glutathione (GSH), resulting in cellu-
are lower in cisplatin resistant cells which is additionally regulated lar export by the MRP transporters, discussed in the prior section.
by ATP7A and ATP7B. Defects in these transporters are associated The secondary form of inactivation is by binding to metallothionein
with diseases with excessive copper accumulation: ATP7A is defec- proteins.
tive in Menkes disease while ATP7B is defective in Wilson’s disease
[86]. 4.1. Inactivation by glutathione conjugation
ATP7A is found is most tissues, aside from the liver. Increased
ATP7A expression is found in cancer cells but not in normal tis- Glutathione-S-transferases (GSTs) catalyze the conjuga-
sue [87]. Increased expression of ATP7A in cells renders the cells tion of glutathione (GSH) to cisplatin. The formation of
resistant to cisplatin but interestingly this was not due to altered platinum–glutathione conjugates inactivates the drug by increas-
cisplatin export [88,89]. There are only a few clinical studies eval- ing its solubility, leading to excretion. Inside the cell, glutathione
uating the role of ATP7A and cisplatin resistance. In NSCLC and acts as antioxidant. It maintains the redox environment by keeping
ovarian cancer, increased ATP7A expression is associated with a reduced sulfhydryl groups [118]. Depletion of cellular GSH in
poorer response to cisplatin [87,89]. ATP7A levels are addition- cisplatin resistant cells enhances the cytotoxicity of cisplatin [119].
ally higher in NSCLC tumors that are resistant to cisplatin [90]. However, the cisplatin sensitivity is not restored to levels of the
No ATP7A polymorphisms have been identified with sensitivity parental cell lines. In ovarian cancer cells, increased levels of GSH
to cisplatin, though studies are very limited in examining ATP7A were observed in platinum resistant cell lines [120]. There are two
polymorphisms in general [91]. families of GST enzymes involved in cisplatin detoxification-GSTP1
ATP7B is found mostly in the liver, kidney and brain. Similar and GSTM.
observations found with ATP7A in terms of cisplatin resistance
have also been observed with ATP7B. ATP7B was first proposed 4.1.1. Glutathione-S-transferase Pi 1
to be a biomarker of cisplatin resistance, as transfection of ATP7B GSTP1, also called GST Pi 1, is expressed in different epithelial
into cells resulted in an increase in cisplatin resistance accompa- tissues. The cellular and clinical studies of GSTP1 are inconclu-
nied by reduced cisplatin accumulation [92]. Increased expression sive as to whether or not it is an indicator of cisplatin resistance.
of ATP7B is associated with poorly differentiated tumors and are In colon, lung, and glioblastoma cancer cell lines, the levels of
poor responders to cisplatin therapy in a variety of cancers includ- GSTP1 are correlated with high GSTP1 expression and increased
ing: gastric, hepatocellular, esophageal, oral, breast, endometrial, cisplatin resistance [121]. In ovarian and head and neck carcinoma
lung, and ovarian [93–101]. Currently, there are no identified ATP7B patient samples, there is a correlation between high expression of
polymorphisms that are associated with cisplatin resistance. GSTP1 and cisplatin resistance [122–124]. Several studies in NSCLC
have demonstrated that low levels of GSTP1 are associated with
3.2.2. ATP-binding cassette (ABC) transporters increased sensitivity to cisplatin [125–129]. However, in other clin-
Multidrug resistance-associated proteins (MRPs), belong to the ical studies of ovarian and cervical cancer there was no association
ABCC subfamily of ABC (ATP-binding cassette) transporters and of GSTP1 levels and response to cisplatin chemotherapy [130–132].
been implicated in cisplatin resistance [102]. MRPs are membrane GSTP1 has two polymorphisms: rs1695 and rs1138272. Similar
transporters responsible for the efflux of glutathione-platinum to the expression data, GSTP1 polymorphism data is conflicting and
conjugates, in an ATP-dependent fashion. MRP1 was first explored inconclusive. The rs1695 polymorphism affects the ability of GSTP1
as a cisplatin transporter as it was found that cisplatin resis- to conjugate GSH to cisplatin [133]. Studies in NSLC have yielded
tant cells displaying increased levels of glutathione concurrently multiple results for the GSTP1 rs1695 polymorphism: associated
had increased levels of MRP1 [103]. Reports from other groups with a favorable response to cisplatin therapy [116,134], associated
suggested that MRP1 alone was not enough to confer cisplatin resis- with reduced survival to cisplatin therapy [135], and no association
tance and there is no relationship between MRP1 and cisplatin with survival [136]. The rs1138272 polymorphism additionally has
accumulation and cytotoxicity [104–106]. There are no polymor- different responses to cisplatin therapy: one study found it was
phisms of MRP1 associated with cisplatin response as well. Thus, associated with greater median survival [136] and another study
MRP1 is generally not thought to play a role in cisplatin resistance. correlated to the polymorphisms to a shorter event free survival
MRP2, also known as cMOAT (canalicular multispecific organic and shorter overall survival in osteosarcoma patients [61].
anion transporter), is the most favored MRP transporter contribut-
ing to cisplatin resistance. Overexpression of MRP2 is found in a 4.1.2. Glutathione-S-transferase Mu
variety of cisplatin resistant cells lines [107–109]. The expression GSTM, or GST Mu, is the other GST involved in the inactiva-
of MRP2 is induced by cisplatin as well [110]. In the clinic, there tion of cisplatin. GSTM is more known for the detoxification of
are different observations in correlating MRP2 expression with xenobiotics thus there is little research in evaluating GSTM in cis-
cisplatin sensitivity, which may reflect the tissue specific nature platin resistance. There are five GSTM genes: GSTM1–GST5. Earlier
of the transporter. Increased MRP2 expression is associated with studies showed that there was no difference and no contribution
cisplatin resistance in colorectal, esophageal, and hepatocellular by GSTMs to cisplatin resistance [137–139]. However, recent data
cancers [111–113]. However, in ovarian and lung cancer MRP2 did using a paired cisplatin sensitive/resistant breast cancer cell line
L. Amable / Pharmacological Research 106 (2016) 27–36 33

demonstrated decreased GSTM3 and GSTM4 levels were found larger sample sizes. Because resistance to cisplatin is multilayered
in cisplatin resistant cells compared to sensitive cells [140,141]. and multifactorial, different mechanisms are likely to be activated
This has not been investigated further as one would assume that depending on the cancer type and stage. It is highly likely that
decreased levels of GSTs would show decreased resistance to multiple resistance mechanisms will be activated within a patient.
cisplatin, like observed with GSTP1. However, pharmacologic inhi- While one biomarker may not be completely informative for all can-
bition of GSTM1 resulted in the increased sensitivity of cells to cers, a combination of biomarker expression and polymorphism
cisplatin [142]. There are no clinical studies examining the rela- screening may yield a comprehensive approach to elucidate the
tionship of GSTM levels with cisplatin sensitivity. resistance status of a patient.
The majority of GSTM polymorphism studies have focused on The method by which patients are screened is critical. The
the susceptibility to cancer and not so much on the relationship type of biospecimen used in the evaluation (blood, tissue, etc.),
with cisplatin resistance. GSTM2 and GSTM5 do not have any the expression type (DNA, mRNA, protein), and the method used
reported polymorphisms. Polymorphisms for GSTM3 and GSTM4 to examine expression (PCR-based, IHC, etc.) will all need to be
have not been evaluated for their relationship with cisplatin resis- standardized for analysis of resistance. The definition of what is
tance. There are few studies with polymorphisms of GSTM1. considered high versus low expression, as well as the cutoff points
Wheeler et al. showed that the rs10431718 GSTM1 polymorphism between the categories, will also require standardization. A recent
was associated with the cisplatin IC50 [143]. In a NSCLC meta- paper described the challenge of biomarker based screening. In
analysis, the GSTM1 null genotype was associated with improved a round robin analysis of three independent commercial labs, 18
response to platinum therapy [144]. tumor blocks were sent for testing of ERCC1 status, and the results
were inconsistent and unreliable [152]. Only 4 of 18 blocks tested
were fully concordant with ERCC1 status between all three labs.
4.2. Inactivation by metallothionein binding
Thus further evaluation and standardization are needed before
these assays become clinical standard-of-care.
Finally, cisplatin is also inactivated by binding to metalloth-
Precision medicine serves two purposes for cisplatin resistance:
ionein (MT) proteins. MT proteins are cysteine-rich, low molecular
to determine if resistance is occurring and to determine the nature
weight proteins that bind to metals such as copper, zinc, cadmium,
of the activated resistance mechanism(s). Screening the patient
and mercury. While there are multiple MTs, mostly MTI and MTII
prior to initiation of treatment, and during the course of treat-
have studied since they are ubiquitously expressed. However, it is
ment, allows for the improvement of cancer diagnosis by predicting
not always evident which MT is being examined. MTs function as
tumor response. Personalizing this therapy will increase the effi-
regulators of cellular metal homeostasis as well as detoxification of
cacy and decrease the toxicity of platinum-based chemotherapy.
heavy metal exposure in cells. In terms of cisplatin resistance, MTs
While there is a long road ahead, several of the biomarkers listed
serve as a heavy-metal detoxifier of cisplatin in tumors. Overex-
here may serve as a foundation for larger, prospective studies to
pression of metallothionein has been observed in several cell lines
determine which biomarker, or combination of biomarkers, would
that are resistant to cisplatin [145–147]. Additionally, the overex-
result in the best prediction of cisplatin sensitivity and resistance
pression of MTII confers resistance to cisplatin in cancer cells [148].
in patients.
Cisplatin treatment induces the expression of MT [149]. In germ
cell tumors, MT expression was higher in cell lines and tumors,
but there was no difference between patients who responded Conflict of interest
to cisplatin based therapy compared to non-responders [150]. In
esophageal cancer, expression of MT was associated with a shorter The author has no conflict of interest.
survival rate after cisplatin therapy [151]. This was additionally
observed in ovarian cancer patients receiving cisplatin based ther-
apy [123]. There are no MT polymorphisms that are associated with Acknowledgments
cisplatin resistance.
The research was supported by the Intramural Research Pro-
gram of the NIH, National Institute on Minority Health and Health
5. Summary Disparities. This manuscript is dedicated to my former mentor, Dr.
Eddie Reed.
Cisplatin is a clinical mainstay for the treatment of a vari-
ety of cancers. Unfortunately, many tumors develop resistance
and are refractory to treatment. Resistance stems from three References
overall mechanisms-increased DNA repair, altered drug cellular
[1] B. Rosenberg, L. Vancamp, T. Krigas, Inhibition of cell division in
accumulation, and increased drug cytosolic inactivation. In this
Escherichia coli by electrolysis products from a platinum electrode, Nature
review, potential biomarkers and their known polymorphisms for 205 (1965) 698–699.
each resistance mechanism were examined and are summarized [2] B. Rosenberg, et al., Platinum compounds: a new class of potent antitumour
agents, Nature 222 (5191) (1969) 385–386.
in Tables 1 and 2. While ERCC1 represents the most promising
[3] E. Reed, Cisplatin, carboplatin, and oxaliplatin, in: B.A. Chabner, D.L. Longo
biomarker for cisplatin resistance, as it has been extensively studied (Eds.), Cancer Chemotherapy and Biotherapy: Principles and Practice, 4th
in a variety of cancers, there are several opportunities and areas ripe edition, Lippincott, Williams, & Wilkins, Philadelphia, 2006, pp. 332–343.
for further study. Other components of NER, CTR1 and CTR2, OCT2, [4] E. Reed, B.A. Chabner, Platinum analogues, in: B.A. Chabner, D.L. Longo
(Eds.), Cancer Chemotherapy and Biotherapy: Principles and Practice, 5th
ATP7A and ATP7B, GSTs, and metallothioneins have the potential edition, Lippincott, Williams and Wilkins, Philadelphia, 2011, pp. 310–322.
to be valid cisplatin biomarkers as well, and would benefit from [5] P. Perego, J. Robert, Oxaliplatin in the era of personalized medicine: from
additional clinical studies. mechanistic studies to clinical efficacy, Cancer Chemother. Pharmacol. 77
(2016) 5–18.
While this list is comprehensive, there are several things to con- [6] M. Markman, et al., Second-line platinum therapy in patients with ovarian
sider. Not all biomarkers were examined in a multitude of cancer cancer previously treated with cisplatin, J. Clin. Oncol. 9 (3) (1991) 389–393.
types, so some may be tumor specific. Many biomarkers displayed [7] A. Eastman, N. Schulte, Enhanced DNA repair as a mechanism of resistance to
cis-diamminedichloroplatinum(II), Biochemistry 27 (13) (1988) 4730–4734.
conflicting evidence with their role in cisplatin resistance. Some [8] V.M. Richon, N. Schulte, A. Eastman, Multiple mechanisms of resistance to
of the conflicting data reflects the fact that many studies maybe cis-diamminedichloroplatinum(II) in murine leukemia L1210 cells, Cancer
underpowered for the analyses, and would benefit from having Res. 47 (8) (1987) 2056–2061.
34 L. Amable / Pharmacological Research 106 (2016) 27–36

[9] K.V. Ferry, T.C. Hamilton, S.W. Johnson, Increased nucleotide excision repair chemotherapy in malignant pleural mesothelioma, Clin. Lung Cancer 14 (5)
in cisplatin-resistant ovarian cancer cells: role of ERCC1-XPF, Biochem. (2013) 558–567, e3.
Pharmacol. 60 (9) (2000) 1305–1313. [36] S.H. Lee, et al., Thymidylate synthase and ERCC1 as predictive markers in
[10] S.W. Johnson, et al., Role of platinum-DNA adduct formation and removal in patients with pulmonary adenocarcinoma treated with pemetrexed and
cisplatin resistance in human ovarian cancer cell lines, Biochem. Pharmacol. cisplatin, Lung Cancer 81 (1) (2013) 102–108.
47 (4) (1994) 689–697. [37] J.J. Yu, et al., A nucleotide polymorphism in ERCC1 in human ovarian cancer
[11] R. Metzger, et al., ERCC1 mRNA levels complement thymidylate synthase cell lines and tumor tissues, Mutat. Res. 382 (1–2) (1997) 13–20.
mRNA levels in predicting response and survival for gastric cancer patients [38] J.J. Yu, et al., Comparison of two human ovarian carcinoma cell lines
receiving combination cisplatin and fluorouracil chemotherapy, J. Clin. (A2780/CP70 and MCAS) that are equally resistant to platinum: but differ at
Oncol. 16 (1) (1998) 309–316. codon 118 of the ERCC1 gene, Int. J. Oncol. 16 (3) (2000) 555–560.
[12] Q. Li, et al., Association between the level of ERCC-1 expression and the [39] E. Reed, ERCC1 and clinical resistance to platinum-based therapy, Clin.
repair of cisplatin-induced DNA damage in human ovarian cancer cells, Cancer Res. 11 (17) (2005) 6100–6102.
Anticancer Res. 20 (2A) (2000) 645–652. [40] S. Smith, et al., ERCC1 genotype and phenotype in epithelial ovarian cancer
[13] M. Dabholkar, et al., ERCC1 and ERCC2 expression in malignant tissues from identify patients likely to benefit from paclitaxel treatment in addition to
ovarian cancer patients, J. Natl. Cancer Inst. 84 (19) (1992) 1512–1517. platinum-based therapy, J. Clin. Oncol. 25 (33) (2007) 5172–5179.
[14] E. Reed, et al., Clear cell tumors have higher mRNA levels of ERCC1 and XPB [41] R. Gao, et al., The ERCC1 N118N polymorphism does not change cellular
than other histological types of epithelial ovarian cancer, Clin. Cancer Res. 9 ERCC1 protein expression or platinum sensitivity, Mutat. Res. 708 (1–2)
(14) (2003) 5299–5305. (2011) 21–27.
[15] M. Ulker, et al., ERCC1 and RRM1 as a predictive parameter for non-small [42] J. Viguier, et al., ERCC1 codon 118 polymorphism is a predictive factor for
cell lung: ovarian or pancreas cancer treated with cisplatin and/or the tumor response to oxaliplatin/5-fluorouracil combination
gemcitabine, Contemp. Oncol. (Pozn.) 19 (3) (2015) 207–213. chemotherapy in patients with advanced colorectal cancer, Clin. Cancer Res.
[16] J.A. Deloia, et al., Comparison of ERCC1/XPF genetic variation: mRNA and 11 (17) (2005) 6212–6217.
protein levels in women with advanced stage ovarian cancer treated with [43] H. Kamikozuru, et al., ERCC1 codon 118 polymorphism is a useful prognostic
intraperitoneal platinum, Gynecol. Oncol. 126 (3) (2012) 448–454. marker in patients with pancreatic cancer treated with platinum-based
[17] E. Cubukcu, et al., Immunohistochemical expression of excision repair chemotherapy, Int. J. Oncol. 32 (5) (2008) 1091–1096.
cross-complementing 1 (ERCC1) in non-small-cell lung cancer: implications [44] T. Hao, et al., Association of four ERCC1 and ERCC2 SNPs with survival of
for patient outcome, Clin. Transl. Oncol. 13 (11) (2011) 826–830. bone tumour patients, Asian Pac. J. Cancer Prev. 13 (8) (2012) 3821–3824.
[18] K.J. Xie, et al., Expression of ERCC1: MSH2 and PARP1 in non-small cell lung [45] Z.H. Shi, G.Y. Shi, L.G. Liu, Polymorphisms in ERCC1 and XPF gene and
cancer and prognostic value in patients treated with platinum-based response to chemotherapy and overall survival of non-small cell lung
chemotherapy, Asian Pac. J. Cancer Prev. 15 (6) (2014) 2591–2596. cancer, Int. J. Clin. Exp. Pathol. 8 (3) (2015) 3132–3137.
[19] Z. Li, et al., Predictive value of APE1: BRCA1, ERCC1 and TUBB3 expression in [46] J. Cheng, et al., A C118T polymorphism of ERCC1 and response to cisplatin
patients with advanced non-small cell lung cancer (NSCLC) receiving chemotherapy in patients with late-stage non-small cell lung cancer, J.
first-line platinum–paclitaxel chemotherapy, Cancer Chemother. Cancer Res. Clin. Oncol. 138 (2) (2012) 231–238.
Pharmacol. 74 (4) (2014) 777–786. [47] J.S. Ryu, et al., Association between polymorphisms of ERCC1 and XPD and
[20] F. Yamashita, et al., Prognostic value of EGFR mutation and ERCC1 in survival in non-small-cell lung cancer patients treated with cisplatin
patients with non-small cell lung cancer undergoing platinum-based combination chemotherapy, Lung Cancer 44 (3) (2004) 311–316.
chemotherapy, PLoS One 8 (8) (2013) e71356. [48] P. Chen, et al., Association of an ERCC1 polymorphism with adult-onset
[21] C. Li, et al., ERCC1 and the efficacy of cisplatin in patients with resected glioma, Cancer Epidemiol. Biomarkers Prev. 9 (8) (2000) 843–847.
non-small cell lung cancer, Tumour Biol. 35 (12) (2014) 12707–12712. [49] W. Zhou, et al., Excision repair cross-complementation group 1
[22] M. Tiseo, et al., ERCC1/BRCA1 expression and gene polymorphisms as polymorphism predicts overall survival in advanced non-small cell lung
prognostic and predictive factors in advanced NSCLC treated with or cancer patients treated with platinum-based chemotherapy, Clin. Cancer
without cisplatin, Br. J. Cancer 108 (8) (2013) 1695–1703. Res. 10 (15) (2004) 4939–4943.
[23] R.V. Lord, et al., Low ERCC1 expression correlates with prolonged survival [50] Y. Wang, et al., Genetic polymorphisms of ERCC1 and their effects on the
after cisplatin plus gemcitabine chemotherapy in non-small cell lung efficacy of cisplatin-based chemotherapy in advanced esophageal
cancer, Clin. Cancer Res. 8 (7) (2002) 2286–2291. carcinoma, Oncol. Rep. 25 (4) (2011) 1047–1052.
[24] P.Y. Huang, et al., Expression of ERCC1 predicts clinical outcome in [51] C. Chen, et al., Polymorphisms in ERCC1C8092A predict progression-free
locoregionally advanced nasopharyngeal carcinoma treated with survival in metastatic/recurrent nasopharyngeal carcinoma treated with
cisplatin-based induction chemotherapy, Oral Oncol. 48 (10) (2012) cisplatin-based chemotherapy, Cancer Chemother. Pharmacol. 72 (2) (2013)
964–968. 315–322.
[25] Z. Zhang, C. Jiang, L. Hu, Low expression of excision repair [52] T.P. Xu, et al., Association of ERCC1-C118T and -C8092A polymorphisms
cross-complementation group-1 protein predicts better outcome in patients with lung cancer risk and survival of advanced-stage non-small cell lung
with locally advanced nasopharyngeal cancer treated with concurrent cancer patients receiving platinum-based chemotherapy: a pooled analysis
chemoradiotherapy, Tumori 100 (3) (2014) 328–332. based on 39 reports, Gene 526 (2) (2013) 265–274.
[26] J. Huang, et al., A phase II study of biweekly paclitaxel and cisplatin [53] M. Dabholkar, et al., Increased mRNA levels of xeroderma pigmentosum
chemotherapy for recurrent or metastatic esophageal squamous cell complementation group B (XPB) and Cockayne’s syndrome
carcinoma: ERCC1 expression predicts response to chemotherapy, Med. complementation group B (CSB) without increased mRNA levels of
Oncol. 30 (1) (2013) 343. multidrug-resistance gene (MDR1) or metallothionein-II (MT-II) in
[27] Z.L. Bai, et al., ERCC1 mRNA levels can predict the response to platinum-resistant human ovarian cancer tissues, Biochem. Pharmacol. 60
cisplatin-based concurrent chemoradiotherapy of locally advanced cervical (11) (2000) 1611–1619.
squamous cell carcinoma, Radiat. Oncol. 7 (2012) 221. [54] J. Yu, et al., Platinum-sensitive and platinum-resistant ovarian cancer tissues
[28] A.O. Zwenger, et al., Expression of ERCC1 and TUBB3 in locally advanced show differences in the relationships between mRNA levels of p53: ERCC1
cervical squamous cell cancer and its correlation with different therapeutic and XPA, Int. J. Oncol. 8 (2) (1996) 313–317.
regimens, Int. J. Biol. Markers 30 (3) (2015) e301–14. [55] E.V. Stevens, et al., Predicting cisplatin and trabectedin drug sensitivity in
[29] M. Xuelei, et al., ERCC1 plays an important role in predicting survival ovarian and colon cancers, Mol. Cancer Ther. 7 (1) (2008) 10–18.
outcomes and treatment response for patients with HNSCC: a [56] B. Koberle, et al., Metastases of squamous cell carcinoma of the head and
meta-analysis, Oral Oncol. 51 (5) (2015) 483–492. neck show increased levels of nucleotide excision repair protein XPF in vivo
[30] M. Ciaparrone, et al., Predictive role of ERCC1 expression in head and neck that correlate with increased chemoresistance ex vivo, Int. J. Oncol. 36 (5)
squamous cell carcinoma patients treated with surgery and adjuvant (2010) 1277–1284.
cisplatin-based chemoradiation, Oncology 89 (4) (2015) 227–234. [57] A. Vaezi, et al., XPF expression correlates with clinical outcome in squamous
[31] S. Ueda, et al., Evaluation of ERCC1 expression for cisplatin sensitivity in cell carcinoma of the head and neck, Clin. Cancer Res. 17 (16) (2011)
human hepatocellular carcinoma, Ann. Surg. Oncol. 18 (4) (2011) 5513–5522.
1204–1211. [58] D.A.et al. Weaver, ABCC5, ERCC2, XPA and XRCC1 transcript abundance
[32] C.M. Hattinger, et al., Excision repair cross-complementation group 1 levels correlate with cisplatin chemoresistance in non-small cell lung cancer
protein expression predicts survival in patients with high-grade: cell lines, Mol. Cancer 4 (1) (2005) 18.
non-metastatic osteosarcoma treated with neoadjuvant chemotherapy, [59] R. Aloyz, et al., Regulation of cisplatin resistance and homologous
Histopathology 67 (3) (2015) 338–347. recombinational repair by the TFIIH subunit XPD, Cancer Res. 62 (19) (2002)
[33] T.C. Lai, et al., Expression of xeroderma pigmentosum complementation 5457–5462.
group C protein predicts cisplatin resistance in lung adenocarcinoma [60] M.R. Spitz, et al., Modulation of nucleotide excision repair capacity by XPD
patients, Oncol. Rep. 25 (5) (2011) 1243–1251. polymorphisms in lung cancer patients, Cancer Res. 61 (4) (2001)
[34] I.G. Hwang, et al., Different relation between ERCC1 overexpression and 1354–1357.
treatment outcomes of two platinum agents in advanced biliary tract [61] K. Goricar, et al., Genetic variability of DNA repair mechanisms and
adenocarcinoma patients, Cancer Chemother. Pharmacol. 68 (4) (2011) glutathione-S-transferase genes influences treatment outcome in
935–944. osteosarcoma, Cancer Epidemiol. 39 (2) (2015) 182–188.
[35] S. Ting, et al., ERCC1, MLH1, MSH2 MSH6, and betaIII-tubulin: resistance [62] G.J. Peters, et al., Predictive role of repair enzymes in the efficacy of cisplatin
proteins associated with response and outcome to platinum-based combinations in pancreatic and lung cancer, Anticancer Res. 34 (1) (2014)
435–442.
L. Amable / Pharmacological Research 106 (2016) 27–36 35

[63] V. Ludovini, et al., Association of cytidine deaminase and xeroderma [94] M. Ohbu, et al., Copper-transporting P-type adenosine triphosphatase
pigmentosum group D polymorphisms with response: toxicity, and survival (ATP7B) is expressed in human gastric carcinoma, Cancer Lett. 189 (1)
in cisplatin/gemcitabine-treated advanced non-small cell lung cancer (2003) 33–38.
patients, J. Thorac. Oncol. 6 (12) (2011) 2018–2026. [95] M. Higashimoto, et al., Expression of copper-transporting P-type adenosine
[64] D.P. Gately, S.B. Howell, Cellular accumulation of the anticancer agent triphosphatase in human esophageal carcinoma, Int. J. Mol. Med. 11 (3)
cisplatin: a review, Br. J. Cancer 67 (6) (1993) 1171–1176. (2003) 337–341.
[65] E.S. Kim, et al., Tissue platinum concentration and tumor response in [96] H. Miyashita, et al., Expression of copper-transporting P-type adenosine
non-small-cell lung cancer, J. Clin. Oncol. 30 (27) (2012) 3345–3352. triphosphatase (ATP7B) as a chemoresistance marker in human oral
[66] H. Eichholtz-Wirth, B. Hietel, The relationship between cisplatin sensitivity squamous cell carcinoma treated with cisplatin, Oral Oncol. 39 (2) (2003)
and drug uptake into mammalian cells in vitro, Br. J. Cancer 54 (2) (1986) 157–162.
239–243. [97] A. Kanzaki, et al., Copper-transporting P-type adenosine triphosphatase
[67] G.R. Gale, et al., Binding of an antitumor platinum compound to cells as (ATP7B) is expressed in human breast carcinoma, Jpn. J. Cancer Res. 93 (1)
influenced by physical factors and pharmacologically active agents, Cancer (2002) 70–77.
Res. 33 (4) (1973) 813–818. [98] T. Aida, et al., Expression of copper-transporting P-type adenosine
[68] M.D. Hall, et al., The role of cellular accumulation in determining sensitivity triphosphatase (ATP7B) as a prognostic factor in human endometrial
to platinum-based chemotherapy, Annu. Rev. Pharmacol. Toxicol. 48 (2008) carcinoma, Gynecol. Oncol. 97 (1) (2005) 41–45.
495–535. [99] K. Nakayama, et al., Prognostic value of the Cu-transporting ATPase in
[69] X. Lin, et al., The copper transporter CTR1 regulates cisplatin uptake in ovarian carcinoma patients receiving cisplatin-based chemotherapy, Clin.
Saccharomyces cerevisiae, Mol. Pharmacol. 62 (5) (2002) 1154–1159. Cancer Res. 10 (8) (2004) 2804–2811.
[70] K. Katano, et al., Acquisition of resistance to cisplatin is accompanied by [100] K. Nakayama, et al., Copper-transporting P-type adenosine triphosphatase
changes in the cellular pharmacology of copper, Cancer Res. 62 (22) (2002) (ATP7B) as a cisplatin based chemoresistance marker in ovarian carcinoma:
6559–6565. comparative analysis with expression of MDR1, MRP1, MRP2, LRP and BCRP,
[71] J. Zisowsky, et al., Relevance of drug uptake and efflux for cisplatin Int. J. Cancer 101 (5) (2002) 488–495.
sensitivity of tumor cells, Biochem. Pharmacol. 73 (2) (2007) 298–307. [101] Y. Inoue, et al., ATP7B expression is associated with in vitro sensitivity to
[72] Z.D. Liang, et al., Mechanistic basis for overcoming platinum resistance using cisplatin in non-small cell lung cancer, Oncol. Lett. 1 (2) (2010) 279–282.
copper chelating agents, Mol. Cancer Ther. 11 (11) (2012) 2483–2494. [102] H. Burger, et al., Drug transporters of platinum-based anticancer agents and
[73] S. Ishida, et al., Enhancing tumor-specific uptake of the anticancer drug their clinical significance, Drug Resist. Updat. 14 (1) (2011) 22–34.
cisplatin with a copper chelator, Cancer Cell 17 (6) (2010) 574–583. [103] T. Ishikawa, C.D. Wright, H. Ishizuka, GS-X pump is functionally
[74] H.H. Chen, et al., Predictive and prognostic value of human copper overexpressed in cis-diamminedichloroplatinum(II)-resistant human
transporter 1 (hCtr1) in patients with stage III non-small-cell lung cancer leukemia HL-60 cells and down-regulated by cell differentiation, J. Biol.
receiving first-line platinum-based doublet chemotherapy, Lung Cancer 75 Chem. 269 (46) (1994) 29085–29093.
(2) (2012) 228–234. [104] S.P. Cole, et al., Pharmacological characterization of multidrug resistant
[75] E.S. Kim, et al., Copper transporter CTR1 expression and tissue platinum MRP-transfected human tumor cells, Cancer Res. 54 (22) (1994) 5902–5910.
concentration in non-small cell lung cancer, Lung Cancer 85 (1) (2014) [105] K. Ikuta, et al., Expression of multidrug resistance proteins and
88–93. accumulation of cisplatin in human non-small cell lung cancer cells, Biol.
[76] X. Xu, et al., Genetic polymorphism of copper transporter protein 1 is Pharm. Bull. 28 (4) (2005) 707–712.
related to platinum resistance in Chinese non-small cell lung carcinoma [106] P. de Cremoux, et al., Role of chemotherapy resistance genes in outcome of
patients, Clin. Exp. Pharmacol. Physiol. 39 (9) (2012) 786–792. neuroblastoma, Pediatr. Blood Cancer 48 (3) (2007) 311–317.
[77] N.K. Wee, et al., The mammalian copper transporters CTR1 and CTR2 and [107] K. Taniguchi, et al., A human canalicular multispecific organic anion
their roles in development and disease, Int. J. Biochem. Cell Biol. 45 (5) transporter (cMOAT) gene is overexpressed in cisplatin-resistant human
(2013) 960–963. cancer cell lines with decreased drug accumulation, Cancer Res. 56 (18)
[78] P.V. van den Berghe, et al., Human copper transporter 2 is localized in late (1996) 4124–4129.
endosomes and lysosomes and facilitates cellular copper uptake, Biochem. J. [108] M. Kool, et al., Analysis of expression of cMOAT (MRP2): MRP3, MRP4, and
407 (1) (2007) 49–59. MRP5, homologues of the multidrug resistance-associated protein gene
[79] B.G. Blair, et al., Copper transporter 2 regulates the cellular accumulation (MRP1), in human cancer cell lines, Cancer Res. 57 (16) (1997) 3537–3547.
and cytotoxicity of cisplatin and carboplatin, Clin. Cancer Res. 15 (13) (2009) [109] P. Borst, M. Kool, R. Evers, Do cMOAT (MRP2), other MRP homologues, and
4312–4321. LRP play a role in MDR? Semin. Cancer Biol. 8 (3) (1997) 205–213.
[80] Y.Y. Lee, et al., Prognostic value of the copper transporters: CTR1 and CTR2, [110] W.K. Kaufmann, et al., TGF-alpha sustains clonal expansion by
in patients with ovarian carcinoma receiving platinum-based promoter-dependent: chemically initiated rat hepatocytes, Carcinogenesis
chemotherapy, Gynecol. Oncol. 122 (2) (2011) 361–365. 18 (7) (1997) 1381–1387.
[81] H. Yoshida, et al., Association of copper transporter expression with [111] E. Hinoshita, et al., Increased expression of an ATP-binding cassette
platinum resistance in epithelial ovarian cancer, Anticancer Res. 33 (4) superfamily transporter: multidrug resistance protein 2, in human
(2013) 1409–1414. colorectal carcinomas, Clin. Cancer Res. 6 (6) (2000) 2401–2407.
[82] K.K. Filipski, et al., Interaction of cisplatin with the human organic cation [112] M. Yamasaki, et al., Role of multidrug resistance protein 2 (MRP2) in
transporter 2, Clin. Cancer Res. 14 (12) (2008) 3875–3880. chemoresistance and clinical outcome in oesophageal squamous cell
[83] H. Burger, et al., Differential transport of platinum compounds by the carcinoma, Br. J. Cancer 104 (4) (2011) 707–713.
human organic cation transporter hOCT2 (hSLC22A2), Br. J. Pharmacol. 159 [113] P.V. Korita, et al., Multidrug resistance-associated protein 2 determines the
(4) (2010) 898–908. efficacy of cisplatin in patients with hepatocellular carcinoma, Oncol. Rep.
[84] A. Naka, et al., Organic cation transporter 2 for predicting cisplatin-based 23 (4) (2010) 965–972.
neoadjuvant chemotherapy response in gastric cancer, Am. J. Cancer Res. 5 [114] Y. Ohishi, et al., ATP-binding cassette superfamily transporter gene
(7) (2015) 2285–2293. expression in human primary ovarian carcinoma, Clin. Cancer Res. 8 (12)
[85] Y. Wang, et al., The association of transporter genes polymorphisms and (2002) 3767–3775.
lung cancer chemotherapy response, PLoS One 9 (3) (2014) e91967. [115] M. Filipits, et al., Multidrug resistance proteins do not predict benefit of
[86] E. Luk, L.T. Jensen, V.C. Culotta, The many highways for intracellular adjuvant chemotherapy in patients with completely resected non-small cell
trafficking of metals, J. Biol. Inorg. Chem. 8 (8) (2003) 803–809. lung cancer: International Adjuvant Lung Cancer Trial Biologic Program,
[87] Z.H. Li, et al., Copper-transporting P-type adenosine triphosphatase (ATP7A) Clin. Cancer Res. 13 (13) (2007) 3892–3898.
is associated with platinum-resistance in non-small cell lung cancer [116] N. Sun, et al., MRP2 and GSTP1 polymorphisms and chemotherapy response
(NSCLC), J. Transl. Med. 10 (2012) 21. in advanced non-small cell lung cancer, Cancer Chemother. Pharmacol. 65
[88] G. Samimi, et al., Increased expression of the copper efflux transporter (3) (2010) 437–446.
ATP7A mediates resistance to cisplatin: carboplatin, and oxaliplatin in [117] B. Han, et al., Association of ABCC2 polymorphisms with platinum-based
ovarian cancer cells, Clin. Cancer Res. 10 (14) (2004) 4661–4669. chemotherapy response and severe toxicity in non-small cell lung cancer
[89] G. Samimi, et al., Increase in expression of the copper transporter ATP7A patients, Lung Cancer 72 (2) (2011) 238–243.
during platinum drug-based treatment is associated with poor survival in [118] C.A. Rabik, M.E. Dolan, Molecular mechanisms of resistance and toxicity
ovarian cancer patients, Clin. Cancer Res. 9 (16 Pt 1) (2003) 5853–5859. associated with platinating agents, Cancer Treat. Rev. 33 (1) (2007) 9–23.
[90] Y. Inoue, et al., Association of ATP7A expression and in vitro sensitivity to [119] S.S. Byun, et al., Augmentation of cisplatin sensitivity in cisplatin-resistant
cisplatin in non-small cell lung cancer, Oncol. Lett. 1 (5) (2010) 837–840. human bladder cancer cells by modulating glutathione concentrations and
[91] J.H. Deng, et al., Clinical outcome of cisplatin-based chemotherapy is glutathione-related enzyme activities, BJU Int. 95 (7) (2005) 1086–1090.
associated with the polymorphisms of GSTP1 and XRCC1 in advanced [120] A.K. Godwin, et al., High resistance to cisplatin in human ovarian cancer cell
non-small cell lung cancer patients, Clin. Transl. Oncol. 17 (9) (2015) lines is associated with marked increase of glutathione synthesis, Proc. Natl.
720–726. Acad. Sci. U. S. A. 89 (7) (1992) 3070–3074.
[92] M. Komatsu, et al., Copper-transporting P-type adenosine triphosphatase [121] S. Goto, et al., Significance of nuclear glutathione S-transferase pi in
(ATP7B) is associated with cisplatin resistance, Cancer Res. 60 (5) (2000) resistance to anti-cancer drugs, Jpn. J. Cancer Res. 93 (9) (2002) 1047–1056.
1312–1316. [122] T. Satoh, et al., Expression of glutathione S-transferase pi (GST-pi) in human
[93] H. Sugeno, et al., Expression of copper-transporting P-type adenosine malignant ovarian tumors, Eur. J. Obstet. Gynecol. Reprod. Biol. 96 (2) (2001)
triphosphatase (ATP7B) in human hepatocellular carcinoma, Anticancer Res. 202–208.
24 (2C) (2004) 1045–1048.
36 L. Amable / Pharmacological Research 106 (2016) 27–36

[123] P. Surowiak, et al., Augmented expression of metallothionein and [138] A.J. Townsend, C.P. Tu, K.H. Cowan, Expression of human mu or alpha class
glutathione S-transferase pi as unfavourable prognostic factors in glutathione S-transferases in stably transfected human MCF-7 breast cancer
cisplatin-treated ovarian cancer patients, Virchows Arch. 447 (3) (2005) cells: effect on cellular sensitivity to cytotoxic agents, Mol. Pharmacol. 41
626–633. (2) (1992) 230–236.
[124] T. Nishimura, et al., Immunohistochemical staining for glutathione [139] D.J. Waxman, et al., Gene-specific oligonucleotide probes for alpha, mu, pi,
S-transferase predicts response to platinum-based chemotherapy in head and microsomal rat glutathione S-transferases: analysis of liver transferase
and neck cancer, Clin. Cancer Res. 2 (11) (1996) 1859–1865. expression and its modulation by hepatic enzyme inducers and platinum
[125] K. Nakagawa, et al., Levels of glutathione S transferase pi mRNA in human anticancer drugs, Cancer Res. 52 (20) (1992) 5797–5802.
lung cancer cell lines correlate with the resistance to cisplatin and [140] M.B. Watson, et al., Expression microarray analysis reveals genes associated
carboplatin, Jpn. J. Cancer Res. 79 (3) (1988) 301–304. with in vitro resistance to cisplatin in a cell line model, Acta Oncol. 46 (5)
[126] K.H. Kim, et al., Clinicopathologic significance of ERCC1: thymidylate (2007) 651–658.
synthase and glutathione S-transferase P1 expression for advanced gastric [141] L. Smith, et al., The proteomic analysis of cisplatin resistance in breast
cancer patients receiving adjuvant 5-FU and cisplatin chemotherapy, cancer cells, Oncol. Res. 16 (11) (2007) 497–506.
Biomarkers 16 (1) (2011) 74–82. [142] C.H. Wang, et al., Inhibition of glutathione S-transferase M1 by new
[127] T. Arai, et al., Correlation between the immunohistochemical and mRNA gabosine analogues is essential for overcoming cisplatin resistance in lung
expression of glutathione S-transferase-pi and cisplatin plus etoposide cancer cells, J. Med. Chem. 54 (24) (2011) 8574–8581.
chemotherapy response in patients with untreated primary non-small cell [143] H.E. Wheeler, et al., Genome-wide meta-analysis identifies variants
lung cancer, Int. J. Oncol. 11 (1) (1997) 127–131. associated with platinating agent susceptibility across populations,
[128] T. Arai, et al., Immunohistochemical expression of glutathione transferase-pi Pharmacogenomics J. 13 (1) (2013) 35–43.
in untreated primary non-small-cell lung cancer, Cancer Detect. Prev. 24 (3) [144] Y. Yang, L. Xian, The association between the GSTP1 A313 G and GSTM1
(2000) 252–257. null/present polymorphisms and the treatment response of the
[129] T. Hirano, et al., Identification of postoperative adjuvant chemotherapy platinum-based chemotherapy in non-small cell lung cancer (NSCLC)
responders in non-small cell lung cancer by novel biomarker, Int. J. Cancer patients: a meta-analysis, Tumour Biol. 35 (7) (2014) 6791–6799.
117 (3) (2005) 460–468. [145] Y.Y. Yang, et al., Human metallothionein isoform gene expression in
[130] K. Ikeda, et al., Multivariate analysis for prognostic significance of histologic cisplatin-sensitive and resistant cells, Mol. Pharmacol. 45 (3) (1994)
subtype: GST-pi, MDR-1, and p53 in stages II-IV ovarian cancer, Int. J. 453–460.
Gynecol. Cancer 13 (6) (2003) 776–784. [146] K. Kasahara, et al., Metallothionein content correlates with the sensitivity of
[131] A.G. van der Zee, et al., Value of P-glycoprotein: glutathione S-transferase pi, human small cell lung cancer cell lines to cisplatin, Cancer Res. 51 (12)
c-erbB-2, and p53 as prognostic factors in ovarian carcinomas, J. Clin. Oncol. (1991) 3237–3242.
13 (1) (1995) 70–78. [147] M.J. Siegsmund, et al., Cisplatin-resistant bladder carcinoma cells: enhanced
[132] I. Konishi, et al., Tumor response to neoadjuvant chemotherapy correlates expression of metallothioneins, Urol. Res. 27 (3) (1999) 157–163.
with the expression of P-glycoprotein and PCNA but not GST-pi in the tumor [148] S.L. Kelley, et al., Overexpression of metallothionein confers resistance to
cells of cervical carcinoma, Gynecol. Oncol. 70 (3) (1998) 365–371. anticancer drugs, Science 241 (4874) (1988) 1813–1815.
[133] M.A. Watson, et al., Human glutathione S-transferase P1 polymorphisms: [149] S.E. Theocharis, A.P. Margeli, A. Koutselinis, Metallothionein: a
relationship to lung tissue enzyme activity and population frequency multifunctional protein from toxicity to cancer, Int. J. Biol. Markers 18 (3)
distribution, Carcinogenesis 19 (2) (1998) 275–280. (2003) 162–169.
[134] H. Lv, et al., Genetic polymorphism of GSTP1 and ERCC1 correlated with [150] C. Meijer, et al., Role of metallothionein in cisplatin sensitivity of germ-cell
response to platinum-based chemotherapy in non-small cell lung cancer, tumours, Int. J. Cancer 85 (6) (2000) 777–781.
Med. Oncol. 31 (8) (2014) 86. [151] Y. Hishikawa, et al., Overexpression of metallothionein correlates with
[135] X. Wu, et al., Germline genetic variations in drug action pathways predict chemoresistance to cisplatin and prognosis in esophageal cancer, Oncology
clinical outcomes in advanced lung cancer treated with platinum-based 54 (4) (1997) 342–347.
chemotherapy, Pharmacogenet. Genomics 18 (11) (2008) 955–965. [152] J.G. Schneider, et al., Commercial laboratory testing of excision repair
[136] C. Lu, et al., Association between glutathione S-transferase pi cross-complementation group 1 expression in non-small cell lung cancer,
polymorphisms and survival in patients with advanced nonsmall cell lung Oncologist 19 (5) (2014) 459–465.
carcinoma, Cancer 106 (2) (2006) 441–447.
[137] Y. Saburi, et al., Increased expression of glutathione S-transferase gene in
cis-diamminedichloroplatinum(II)-resistant variants of a Chinese hamster
ovary cell line, Cancer Res. 49 (24 Pt 1) (1989) 7020–7025.

You might also like