MOLECULAR MARKERS FOR CAPECITABINE THERAPY: A REVIEW
HTML Full TextMOLECULAR MARKERS FOR CAPECITABINE THERAPY: A REVIEW
Ramalakshmi *1, S. Kavimani 2, Satish Srineevas 3, V. Vetriselvi 4 and LVKS Bhaskar 5
K. College of Pharmacy1, Gerugambakkam, Chennai, Tamilnadu, India
Department of Pharmacology 2, Mother Theresa Post Graduate & Research Institute of Health sciences, Puducherry, India
Department of Radiation Oncology 3, Department of Human Genetics 4, Sri Ramachandra University, Porur, Chennai, Tamilnadu - 600116, India
Sickle Cell Institute Chhattisgarh Genetic Lab 5, Department of Biochemistry, Pt. JNM Medical College, Raipur, Chhattisgarh, India
ABSTRACT: Capecitabine an oral prodrug of 5- fluorouracil is widely used for the treatment of variety of solid tumors, particularly colorectal cancer. However it is not devoid of toxicities and may limit therapy. A little is known about predictors of toxicity, response and survival in patients treated with capecitabine. The pharmacogenetic testing methods can identify such variants and thus indicate those patients who are at risk for adverse effects with capecitabine. Various studies have been carried out to assess the various genetic predictive and prognostic markers with treatment. The purpose of this review is to describe the comprehensive reports and draw conclusion with the available information on capecitabine pharmacogenetics and future directions on ongoing research. An extensive literature search was carried out on the genes encoding the enzymes involved in the metabolism of capecitabine. Overall, evidence indicates multiple genes associated with the response/toxicity with capecitabine therapy, however majority of reports indicate DPD deficiency as a source of life-threatening toxic effects. Hence, prospective studies correlating the enzymes and the concentration of the drug and its metabolites in the body are needed before validated SNP tests can enter routine clinical practice.
Keywords: Capecitabine, Pharmacogenetic, Polymorphism, Toxicity, Molecular Marker
INTRODUCTION: Capecitabine (CAP), carbomate of fluropyrimidine is an orally administered anti neoplastic agent. It was synthesized in 1990s by Japanese researchers as an oral formulation designed to overcome the toxicity of 5'-deoxy-5-fluorouridine (5′d5-FUrd) 1.
This 5′d5-FUrd is related to GI toxicity, attributed to the liberation of 5FU in the small intestine under the action of thymidine phosphorylase (TP) 2. Thus CAP was designed as a prodrug of 5′d5-FUrd that crosses the GI barrier intact and is rapidly and almost completely absorbed 3.
The anti tumor activity is related to fluorouracil which is a fluorinated pyrimidine antimetabolite that inhibits thymidylate synthetase, blocking the methylation of deoxyuridylic acid to thymidylic acid, interfering with DNA, and to a lesser degree, RNA synthesis 4. Fluorouracil appears to be phase specific for the G1 and S phases of the cell cycle.
Capecitabine is FDA-approved for: Adjuvant in colorectal cancer Stage III Dukes' C & used as first-line monotherapy and in metastatic colorectal cancer as well. If appropriate, it is used in combination with docetaxel, after failure of anthracycline-based treatment in metastatic breast cancer. It is also used as monotherapy, if the patient has failed paclitaxel-based and anthracycline-based treatment has failed or cannot be continued for other reasons (i.e., the patient has already received the maximum lifetime dose of an anthracycline).
In the UK, capecitabine is approved by the National Institute for Health and Clinical Excellence (NICE) for colon and colorectal cancer, and locally advanced or metastatic breast cancer.5 On March 29, 2007, the European Commission approved Capecitabine, in combination with platinum-based therapy (with or without epirubicin), for the first-line treatment of advanced stomach cancer. Unlabeled it is used in the treatment of CNS lesions for metastatic breast cancer, esophageal cancer, gastric cancer, hepatobiliary cancers (advanced), neuroendocrine (pancreatic/islet cell) tumors (metastatic or unresectable), ovarian cancer (platinum-refractory), pancreatic cancer (metastatic), unknown primary cancer.
Dosing:
The first line treatment of metastatic colorectal cancer patients on whom single agent capecitabine therapy is preferred, the approved FDA dose is 1,250 mg/m2 which is given orally twice daily, over 12 hours interval for 14 days followed by a period of rest for 1 week. The FDA has also approved the same dose as colorectal cancer for metastatic breast cancer patients who have developed resistance to both anthracycline and taxane based regimens or in whom any further anthracycline therapy is contraindicated, as single agent therapy. Additionally, the approved combination regimen with docetaxel 75 mg/m2 as a 1-hour IV infusion on the first day for metastatic breast cancer patients, in whom prior anthracycline therapy has failed and 1,250 mg/m2 capecitabine administered orally twice a day for the first 2 weeks of every 3-week cycle. In prostate, renal cell, ovarian and pancreatic cancers, the majority of evidence is from smaller, Phase II trials, and efficacy has been mostly in the form of partial response and stable disease. Combination therapy in these patients appears to be more beneficial than single-agent capecitabine. 6 Further, based on tolerability dosage modifications are recommended. 7 The ideal dosing of capecitabine varies as regional differences have been seen in the tolerance of oral fluoropyrimidines.8 The product data recommends the tablets should be administered within 30 minutes of a meal; this may decrease gastrointestinal discomfort.
Toxicities with capecitabine:
Capecitabine is not devoid of toxicities. It poses a risk of adverse effects, some of which can be serious and dose limiting. The predominant adverse effects observed with capecitabine include hand and foot syndrome, Stomatitis, nausea, vomiting and diarrhoea. The Toxicity Profile of Capecitabine monotherapy is tabulated in the Table 1.
TABLE 1: TOXICITY PROFILE CAPECITABINE MONOTHERAPY IN COLON AND RECTAL CANCERS
Study | No of patients, Cancer type | Place of study | setting | Toxicities |
Twelves et al.9
|
1004 ,CRC
Phase III |
164 centers worldwide | Adjuvant Colon Cancer | Diarrhoea (11%), nausea vomiting (3%), Stomatitis (2%), hand and foot syndrome (17%), neutropenia (2%), hyperbilirubinemia (20%) |
Hoff et al.10
|
302,CRC
Phase III |
US, Canada, brazil and Mexico | Metastatic Colorectal Cancer | Diarrhoea(15.4%),vomiting(3.6%), Stomatitis(3%), hand and foot syndrome(18.1%),neutropenia(2.6%), hyperbilirubinemia(17.3%) |
Van Custem et al.11 | 297,CRC
Phase III |
59 centers in Europe, Australia, New Zealand, Taiwan, and Israel | Metastatic Colorectal Cancer | Diarrhoea10.7%, Stomatitis1.3%, hand and foot syndrome16.2%, neutropenia 2%, hyperbilirubinemia 28.3% |
Chi-Ching Law et al12 | 58,single arm | China | Adjuvant colon cancer | Diarrhoea (0), nausea vomiting (0), Stomatitis(1.7%), hand and foot syndrome( 41.4%), neutropenia (1.7%), hyperbilirubinemia (1.7%) |
Janneke Baan et al13 | 9CRC,28breast cancer, single centre | Netherland | Metastatic and adjuvant setting | Nausea/Vomiting(16.2%), Mucositis (e.g. stomatistis, gastritis) (27.0%), Diarrhoea (24.3%), Hand and foot
Syndrome (10.8%), Bone marrow suppression (8.1%), Thromboembolic event (2.7%) |
In its standard dose schedule of capecitabine is 2000 mg/ m2 per day (days 1–14) and oxaliplatin 130 mg /m2 (day 1) every 3 weeks (CAPOX 2000), the reported rate of grades 3 and 4 diarrhoea rate was about 20% 14-17. This severe diarrhoea rate further increased when CAPOX 2000 was combined with cetuximab 18. Differential reports of toxicities by patients from different racial groups were observed. Regional differences in tolerability of capecitabine as adjuvant therapy was noted with toxicities being worse in the US population compared with rest of the world 19, 20 and whereas in Chinese population a much higher incidence of serious hand-foot syndrome and a lower rate of severe diarrhoea were observed12.
Pharmacogenetics:
Although in recent years, there has been a vast improvement in the outcome of patients, present day protocols are still limited by the unpredictable response to the drug and to its severe toxic effects. The different responses to a particular drug are not only due to the specific clinic pathological features of the patients but also ethnic origins and particular genotype of a single individual. Here, we provide an overview of the known polymorphism present in the genes which codify for key metabolic factors involved in the mechanism of action of the drugs responsible for significant toxicity with capecitabine
FIG. 1: CAPECITABINE IN ITS METABOLIC PATHWAY
5'-DFUR ,5'-deoxy-5-fluorouridine ; 5'-DFCR, 5'-Deoxy-5-fluorocytidine; 5-dUrd, 5-fluorodeoxyuridine ; 5-10 CH=FH4, 5-10 methenyltetrahydrofolate; 5-10 CH2FH4, 5-10 methylene-tetrahydrofolate; 5-CH3FH4, 5-methyltetrahydrofolate; 5-CHOFH4 (FA), 5-formyltetrahydrofolate; DHFR, dihydrofolate reductase; FdUMP, 5-fluorodeoxyuridine 5′-monophosphate; FdUrd, 5-fluorodeoxyuridine; FH2, dihydrofolate; FH4, tetrahydrofolate; MS, methionine synthase; TK, thymidine kinase; TP, thymidine phosphorylase.
Carboxylesterases:
Carboxylesterases (CES) are members of the α/β hydrolase fold family and are a group of enzymes that function in the metabolism of ester and amide prodrugs to their free acid forms 21. They are ubiquitously expressed, but levels are highest in the small intestine, liver, and lungs. There are five genes of carboxylesterases reported in humans, named CES1-CES5. CES1 substrates generally contain a large acyl and a small alcohol group, while substrates for CES2 contain a small acyl and a large alcohol moiety. Carboxylesterases hydrolyze capecitabine's carbamate side chain to form 5′-deoxy-5-fluorocytidine (5′-DFCR). Both CES1A1 and CES2 are responsible for the activation of capecitabine, whereas CES3 plays little role in 5′-DFCR formation. For the first time N Ribelles et al reported an association between a polymorphism in the CES2 gene(CDD 943insC and CES 2 Exon 3 6046 G/A) and the efficacy of capecitabine with a non-statistically significant higher incidence of grade 3 hand-foot syndrome (HFS) (p=0.07) and grade 3-4 diarrhoea (p=0.09), respectively.
It was also observed that the patients heterozygous or homozygous for the polymorphism CES 2 5UTR 823 C/G exhibited a significantly greater response rate to capecitabine, and time to progression of disease (59%, 8.7 months) than patients with the wild type gene sequence (32%; 5.3 months) 22.
Thymidine phosphorylase:
Thymidine phosphorylase (TP), also known as platelet-derived endothelial cell growth factor (PD-ECGF), is an enzyme expressed in many normal tissues and cells with a key role in pyrimidine salvage pathway that recovers pyrimidine nucleosides formed during RNA or DNA degradation 23. TP catalyzes the conversion of thymidine, deoxyuridine and their analogs to their respective bases and 2-deoxyribose-1- phosphates. Furthermore, TP is often overexpressed in tumor sites. The high TP activity in the tumor can selectively activate the 5FU prodrug 5'-deoxy-5-fluorouridine to 5FU. 24 Thymidinephosphorylase is located at chromosome 22 in the region of q13.33. cDNA is approximately 1.8 kb long, consisting of 10 exons in a 4.3 kb region .25, 26 TP is highly expressed in liver tissues.
Thymidine phosphorylase is upregulated in a wide variety of solid tumors such as breast, bladder, gastric, colorectal, pancreatic, lung and esophageal cancer 27-31. Several literatures shows a correlation between the TP expression level with tumor angiogenesis, growth and progression illustrating that TP promotes tumor growth and metastasis by preventing apoptosis and inducing angiogenesis.32, 33
As TP is the rate-limiting enzyme for the activation of capecitabine, it might be a useful predictor of tumor response to capecitabine-based chemotherapy. Reports of a study by bokos et al confer that Median ratio TP/DPD of patients with proven pathological "response" (downstaging of the disease) was higher than the "no response" group, 4.40 and 1.42, respectively (P = 0.0001) indicating that Thymidine phosphorylase to dihydropyrimidine dehydrogenase ratio as a predictive factor of response to preoperative chemoradiation with capecitabine in patients with advanced rectal cancer 34.
Similarly, Meropol et al. assessed in the tissues from primary and metastatic sites for TP protein expression in patients with previously untreated metastatic CRC on irinotecan plus capecitabine chemotherapy and concluded that the objective tumor response to chemotherapy was associated with high TP expression in primary tumors (OR: 4.77; 95% CI: 1.25-18.18), with a similar trend in metastases (OR: 8.67; 95% CI: 0.95-79.1). These reports seem to indicate that TP expression might be a predictive marker for response. In addition, overall survival (OS) was also based on TP expression. The median survival in TP-expressing primary tumors was 28.2 months (95% CI: 15.5--39.8 months) compared to 14.9 months (95% CI: 9.0--20.5) in patients with TP-negative primary tumors. 35
Another study by Petrioli R et a indicated a significant associated with high TP expression in metastatic tissue with response to treatment (P=0.019), and also with a trend towards a better median progression-free survival and overall survival compared with patients expressing low TP (P=0.056; P=0.073) suggesting that patients with high levels of intratumoral TP expression were the ideal candidates for capecitabine-based chemotherapy patients with metastatic colorectal cancer treated with continuous oral capecitabine and biweekly oxaliplatin 36. Lindskog EB et al reports that high TP gene expression in non-microdissected tumour tissues of patients with advanced colorectal cancer correlates with longer time to progression.37
Several studies report a correlation between elevated TP (as measured by either IHC or ELISA) and benefit with high response rate in gastric cancer patients and breast cancer on capecitabine based therapy as well.38-43 These findings support the potentially predictive role of TP and a promising marker that can give helpful information to benefit from capecitabine-based chemotherapy.
Palmar-plantar erythordysesthesia (PPE) is the most common toxicity of capecitabine. Saif M W et al showed no significant association of PPE with higher tumor thymidine phosphorylase in patients receiving capecitabine. 44 With an extensive literature survey, it was observed that TP plays a dual role in cancer development and therapy. On the one hand, TP activity is necessary for the activation and in contrast, high levels of TP in tumor tissues are correlated with a poor prognosis because of higher tumor aggressiveness. This apparently opposite effect of TP illustrates the complexity of this marker in tumor progression and in the clinical response to capecitabine therapy and strongly suggesting the involvement of other markers.
Cytidine Deaminase:
The human CDA spans approximately 30 kB and consists of 4 exons. No splice variant was reported. The CDA gene encodes an enzyme involved in the pyrimidine salvage pathway and catalyzes irreversibly the hydrolytic deamination of cytidine and deoxycytidine into uridine and deoxyuridine, respectively 45. In addition, CDA plays an essential role in the metabolism of a number of antitumor cytosine nucleoside analogues, leading to their pharmacologic activation to 5-FU.Few case reports indicate the association of CDA with toxicity. A report by Mercier at al suggests severe toxicity with capecitabine linked to CDA without the deficiency Of DPD. 46 A study by Caronia et al. investigated the association between grade 3 HFS and the genes involved in capecitabine metabolism and found that the deleted allele of rs3215400 showed an increased allele-specific expression and was significantly associated with an increased risk of capecitabine-induced HFS. 47
Thymidylate Synthase:
The prime target for 5FU is thymidylate synthase (TS). TS in association with a methyl cofactor, catalyses the conversion deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP). by methylation.
In a trial by Uchida K et al involving predominantly pre-treated colorectal cancer patients, higher levels of TS mRNA expression appeared to have a predictive value in identifying those subjects with a greater probability of early disease progression during treatment with XELOX 48and a study by lee et al reports that the median PFS was significantly lower in patients with high TS (6.6 vs. 3.0 months; P = 0.017) 42. Another study by Lee S et al showed that high expression of TS and TP was associated with a higher RR than was low expression of TS and TP (54.1 vs 40.5%, P=0.022) in advanced oesophageal squamous cell carcinoma with capecitabine and cisplatin therapy.49 Study reports of Agustin A. Garcia, et al indicated severe nausea/vomiting associated with TS expression (P = 0.039), but not with other severe toxicities50. The results of a recent study presented by Martinez-Balibrea et al and Spindler K L et al reported a less pronounced impact of the TS and ERCC1 genotypes on XELOX efficacy compared with the outcome of an 5-FU-oxaliplatin infusional regimen (FUOX) 51, 52. Among the three common polymorphisms associated with altered TS gene expression the variable number of tandem repeats (VNTR) polymorphism in 5’untranslated region mainly leads to double or triple tandem repeats (2R or 3R) of a 28-bp sequence. These repeats are found in Caucasians, Asians and Africans. The Four, five or nine tandem repeats have also been described for this polymorphism. A low TS levels in vitro and in tumour tissue samples were observed with homozygous 2R/2R genotype.53, 54 A deletion of 6 bp in the TS gene has been associated with decreased TS expression 55. Several studies indicate association between treatment outcome and TS polymorphism which is depicted in Table 2 .57-59
TABLE 2: SUMMARY OF PUBLISHED LITERATURE ON TS POLYMORPHISM WITH CAPECITABINE
Publication | No of patients | Type of cancer | Genotype | Response | Toxicity | survival |
David J. Park et al,200256 | 24 | Colorectal cancer patients | 5' untranslated region | 75% (3/4) of individuals with the S/S variant responded to capecitabine, compared to 8% (1/12) and 25% (2/8) of those with the S/L and L/L variants, respectively | 100% S/S variant reported toxicity level of grade 3 and 56% of those with S/L and 37.5% with L/L | |
Rémy Largillier et al,200657 | 105 | Breast cancer | TS 5’ genotype
28 bp tandem repeats |
(3RG3RG genotype)
were prone to rapid disease progression |
a higher global toxicity grade 3 and 4 was observed in patients
homozygous for the TS 3RG allele compared with patients heterozygous for the 3RG allele or patients not carrying the 3RG allele (50% versus 19% versus 13% respectively, P = 0.064). was not related to toxicity |
|
J. Gao et al,201258 | 123 | Advanced gastric cancer patients | TS 3′-UTR | TS ins6/ins6 genotype was the independent poor OS predictor (P = 0.001, HR = 3.182) |
In the second repeat of the 3R allele, a SNP resulting in a G>C change has been observed. Increased gene expression and protein levels have been associated with the G allele of this SNP. Another common polymorphism has been reported at position 1494 in the 3’UTR of TS 59. 3’UTRs modulate gene regulation at a posttranscriptional level through control of mRNA stability. A 6-bp insertion/deletion (indel) polymorphism in TS 3’R is associated with decreased TS mRNA stability in vitro and reduced expression of TS protein in colorectal tumor tissue 60. According to a study by A Loganayagam et al the presence of a homozygous del/del genotype approximately doubled the risk of grade 3–4 toxicity 61. However, other studies have failed to show an association between a homozygous del/del genotype and severe toxicity 62-64.
Methylenetetrahydrofolate Reductase:
An important enzyme in the folate pathway is Methylenetetrahydrofolate reductase (MTHFR) which converts 5,10-Methylenetetrahydrofolate (5,10-MTHF) to 5-methyltetrahydrofolate.An increased levels of 5,10-MTHF available for inhibiting TS has been linked to reduction in enzyme activity and therefore leading to increased efficacy of 5-FU.65 The two common polymorphism of MTHFR gene are 677c>T and 1298A>C are known to reduce the enzyme activity leading to a decreased pool of methyl THF and associated with hyperhomocysteine particularly in folate deficiency 66. The 677c>T transition at exon 4 causes an amino acid substitution from alanine to valine at codon 222 within the catalytic region of the enzyme.the677c>T variants MTHFR TT genotype show-30% of the enzyme activity found among those with wild type(CC). While individuals who are heterozygous for mutation CT have -65% of wild type enzyme activity. Individuals with the TT genotype, particularly if combined with a diet low in folate, have elevated plasma homocysteine levels.
The A1298C polymorphism, resulting in an amino acid change of glutamine to alanine, also leads to reduced enzyme activity of the minor allele; however, this is to a lesser extent than for the 677T allele.65 Studies related to the association of these genotypes with reponse/toxities with capecitabine is represented in the Table 3.
TABLE 3: SUMMARY OF PUBLISHED LITERATURE ON MTHFR POLYMORPHISM WITH CAPECITABINE
Study | No of patients | Type of cancer | Genotype | Response | Toxicity | survival |
van Huis-Tanja LH et al
201367 |
127 | CRC | MTHFR 1298A>C
677C>T |
No significant association | MTHFR 1298CC homozygotes showed a borderline significantly higher incidence of grade 3-4 diarrhoea compared with MTHFR 1298AC or AA individuals | - |
J. Dimovskiet al,
2010 et al 68 |
136 | Colon cancer | 677C>T | The MTHFR 677 TT genotype was predictive of absence of side effects (OR 0.094, 95% CI 0.01-0.87, p = 0.014). | MTHFR 677TT genotype are predictive for mid-term relapse-free survival | |
Rohini Sharma et al,
200862 |
CRC | MTHFR c.677C>T and c.1298A>C genotypes and diplotypes predicted for grade 2/3
Toxicities |
MTHFR c.677 genotype tended to
predict overall survival (P = 0.08) |
|||
Remy Largillier et al,200657 | 105 | Breast cancer | MTHFR genotypes
677C>T and 1298A>C were not related to toxicity |
|||
Lei-zhen Zheng et al69 | 93 | Gastric cancer | MTHFR 1298C/A had marginally significant correlation with the survival of patients (χ2=3.447,P=0.062), |
Dihydropyrimidine Dehydrogenase:
Dihydropyrimidine dehydrogenase (DPD) acts in the degradation of 5-FU and a rate limiting enzyme accounting for more than 80% in its catabolism. The clearance of 5FU depends on DPD activity. Hence, reduced DPD activity leads to both increased toxicity and efficacy of the drug 70. Till date at atleast 30 polymorphism in DPD gene have been described 71. In two studies Common genetic polymorphisms in the DPD gene (DPD-C1896T, DPD-T85C, DPD-A496G, DPD-A557G, DPD-A1627G, DPD-T3351C, DPD-G3649A, DPD-A3844G and DPD-T3856C) leading to altered enzyme activity were examined. These studies reveal no significant associations between DPD genotype and toxicity or response. 72, 73. The most common polymorphism is DPYD*2A,G>A splice site transition that causes skipping of exon 14 has been found in upto 40-50% of people with partial or complete DPD deficiency. The point mutation G to A in the invariant slice donor site leads to skipping of exon 14 immediately upstream of the mutated splice donor site in the process of DPD prem RNA splicing segment coding the aminoacid 581-63574. Patients heterozygous for this polymorphism have low DPD activity and developing toxicity is represented in Table 4 and several case reports also indicate toxicity with DPD deficiency 81-85.
TABLE 4: SUMMARY OF DPD POLYMORPHISM AND ASSOCIATED TOXICITIES WITH CAPECITABINE
Publication | No of patients | Type of cancer | Genotype | Toxicity |
Maarten J. et al 75 | 568 | Advanced Colorectal Cancer | IVS14+1G>A, 1236G>A, 2846A>T, 2194G>A, or 496A>G | IVS14+1G>A and 1236G>A were strongly associated (P < 0.05; FDR < 0.3) with grade 3 to 4 diarrhea. |
M. Wasif et al 76
|
23 | GI malignancies | (IVS14+1 G>A, DPYD | severe leucopenia demonstrated |
M. Del Re et al,77 | 450 patients
Italy
|
with gastrointestinal, breast and pancreas cancers | IVS14+1G>A and 2846T>C DPD variants | Toxicities in all subjects were G3/4 diarrhea (100%), G3/4 mucositis (48%), febrile neutropenia (45%), G3/4 thrombocytopenia (38%), G3/4 anemia (24%), G2/3 hand-foot syndrome (14%), G3 dermatitis (7%) and G2/4 alopecia (7%). |
Timur Ceric et al 78
|
50 subjects | GI malignanacy | IVS14 +1 G>A, | Diarrhea, neutropenia, mucositis |
Muhammad Wasif Saif et al,2013 79 |
colon (n=871, 62%)breast (n=184, 13%)227 patientsUSA |
Colon cancer, breast cancer | IVS14 +1 G>A, D949V | hand-foot syndrome 9%,
myelosupression-9% |
Loganayagam A et al 80 | 47 patients | GI malignanacy | 1905+1G>A, 2846A>T,1679T>G | 1905+1G>A developed severe diarrhoea |
The simultaneous presence of variants DPYD*2A and 2846A>T was shown to be lethal in several cases shortly after initiation of treatment with fluoropyrimidine. 86, 87 Deenen et al. found that 100% of patients carrying the IVS14+1 polymorphism developed severe toxicity.88 Measuring DPYD activity has been suggested to be a better biomarker for fluoropyrimidine-induced toxicity than DPYD genotyping, although recent findings confirm that genotyping and haplotyping could be acceptable options for stratifying patients according to risk of toxicity 89 or establishing dose recommendations for capecitabine and as shown by the Pharmacogenetics Working Group of the Royal Dutch Pharmacists Association.90
Recently Recommended dosing of fluoropyrimidines by DPD phenotype has been laid 91, Hélène Blasco et al indicated that, once this deficiency has been identified on the basis of both DPD genotype and phenotype with capecitabine, it is possible to tailor 5-FU dose in DPD-deficient patients, using TDM 92. However a study by Garcia AA et al expression of DPD was not associated with any of the severe toxicities.93 Saif MW et al reports discoloration of his palms consistent with HFS, contrary to the pattern and degree of HFS reported in the current guidelines as proposed in the drug insert. This case suggests that the pattern of cutaneous manifestations varying in patients with different ethnic backgrounds, especially whites Versus non-whites with the normal limits of DPD.94
A study by Vallböhmer D et al reports a Higher gene expression levels of DPD were associated with resistance to capecitabine (P=0.032). Patients with a lower mRNA amount of DPD (<or=0.46) had a longer progression-free survival compared with patients that had a higher mRNA amount (8.0 vs. 3.3 months; adjusted P=0.048; log-rank test).95 Nishimura G et al reports patients with high TP but low DPD had the best disease-free survival, whereas the low TP but high DPD group had the worst survival in colorectal cancer patients on capecitabine.96
CONCLUSION: Predicting response and limiting drug induced toxicity are two important challenges faced by clinicians in the treatment of colorectal cancer. The introduction of genetic testing individualize treatment regimens will hope fully allow better response prediction and limit drug induced toxicity leading to improved patient outcomes. Determination of polymorphisms in metabolizing enzymes before the administration of chemotherapy could offer new strategies for optimizing the treatment of individual patients. However, phase wise studies on large population simultaneously accounting for other co variables should be carried out for predicting and confirming the effects of multiple genetic factors.
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST:
No potential conflicts of interest were disclosed.
REFERENCES:
- Arazaki M, Ishitsuka H, Kuruma I et al. N-oxycarbonyl substituted 5′-deoxy-5-fluorocytidines. Eur Patent Appl 1992; no. 92121538.0.
- Haraguchi M, Miyadera K, Uemura K et al. Angiogenic activity of enzymes. Nature 1994; 368:198.
- Judson IR, Beale PJ, Trigo JM et al. A human capecitabine excretion balance and pharmacokinetic study after administration of a single oral dose of 14C-labelled drug. Invest New Drugs 1999; 17:49–56.
- Pinedo, H. M. and G. F. Peters (1988).Fluorouracil: biochemistry and pharmacology. J Clin Oncol 6(10): 1653-64.
- Lacy, Charles F; Armstrong, Lora L; Goldman, Morton P; Lance, Leonard L (2004). Lexi-Comp's Drug Information Handbook (12th Edition). Lexi-Comp Inc. ISBN 1-59195-083-X.
- Georgios V. Koukourakis,Vassilios Kouloulias, Michael J. Koukourakis, Georgios A. Zacharias, Haralabos Zabatis and John Kouvaris. Efficacy of the Oral Fluorouracil Pro-drug Capecitabine in Cancer Treatment: a Review. Molecules 2008; 13(8): 1897-1922.
- Nutley, N.J. Xeloda (capecitabine) [product information]; Roche Pharmaceuticals, 2003.
- Daniel G. Haller,Jim Cassidy, Stephen J. Clarke, David Cunningham, Eric Van Cutsem, Paulo M. Hoff MaceL. Rothenberg et al. Potential Regional Differences for the Tolerability Profiles of Fluoropyrimidines 2008; 26( 13 ):2118-2123.
- Twelves C, Wong A, Nowacki MP, et al. Capecitabine as adjuvant treatment for stage III colon cancer. N Engl J Med 2005; 352:2696–704.
- Hoff PM, Ansari R, Batist G, et al. Comparison of oral capecitabine versus intravenous fluorouracil plus leucovorin as first-line treatment in 605 patients with metastatic colorectal cancer: results of a randomized phase III study. J Clin Oncol 2001; 19: 2282–92.
- Van Cutsem E, Twelves C, Cassidy J, et al. Oral capecitabine compared with intravenous fluorouracil plus leucovorin in patient with metastatic colorectal cancer: results of a large phase III study. J Clin Oncol 2001; 19:4097–106.
- Chi-Ching Law, Yiu-Tung F, Kwok-Kwan Chau, Tim-Shing Choy, Ping-Fai So, Kam-Hung Wong. Toxicity Profile and Efficacy of Oral Capecitabine as Adjuvant Chemotherapy for Chinese Patients with Stage III Colon Cancer Dis Colon Rectum 2007; 50: 2180–2187
- Janneke Baan, Monique MEM Bos, Savita U Gonesh-Kisoensingh, Iwan A Meynaar, Jelmer Alsma, Erik Meijer and Arnold GVulto. Capecitabine-induced Toxicity: An Outcome Study into Drug Safety. J Integr Oncol 2014, 3:1.
- Schmoll HJ, Cartwright T, Tabernero J, Nowacki MP, Figer A, Maroun J, Price T, Lim R, Van Cutsem E, Park YS, McKendrick J, Topham C, Soler-Gonzalez G, de Braud F, Hill M, Sirzen F, Haller DG. Phase III trial of capecitabine plus oxaliplatin as adjuvant therapy for stage III colon cancer: a planned safety analysis in 1,864 patients. J Clin Oncol. 2007; 25:102–109.
- Cassidy J, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F, Couture F, Sirzén F, Saltz L.Randomized phase III study of capecitabine plus oxaliplatin compared with fluorouracil/folinic acid plus oxaliplatin as first-line therapy for metastatic colorectal cancer. J Clin Oncol. 2008 Apr 20; 26(12):2006-12.
- Rothenberg ML, Cox JV, Butts C, Navarro M, Bang YJ, Goel R, Gollins S, Siu LL, Laguerre S, Cunningham D Capecitabine plus oxaliplatin (XELOX) versus 5-fluorouracil/folinic acid plus oxaliplatin (FOLFOX-4) as second-line therapy in metastatic colorectal cancer: a randomized phase III non inferiority study. Ann Oncol. 2008 Oct; 19(10):1720-6.
- Haller DG, Cassidy J, Tabernero J, Maroun JA, De Braud FG, Price TJ, Van Cutsem E, Hill M, Gilberg F, Schmoll H. Efficacy findings from a randomized phase III trial of capecitabine plus oxaliplatin versus bolus 5-FU/LV for stage III colon cancer (NO16968): impact of age on disease-free survival (DFS). J Clin Oncol (Meeting Abstracts) 2010; 28:3521.
- Adams RA, Meade AM, Madi A, Fisher D, Kay E, Kenny S, Kaplan RS, Maughan TS. Toxicity associated with combination oxaliplatin plus fluoropyrimidine with or without cetuximab in the MRC COIN trial experience.Br J Cancer. 2009 Jan 27; 100(2):251-8.
- Haller DG, Cassidy J, Clarke SJ, Cunningham D, Van Cutsem E, Hoff PM et al. Potential regional differences for the tolerability profiles of fluoropyrimidines.J Clin Oncol. 2008 May 1; 26(13):2118-23.
- Hochster HS, Hart LL, Ramanathan RK, Childs BH, Hainsworth JD, Cohn AL et al. Safety and efficacy of oxaliplatin and fluoropyrimidine regimens with or without bevacizumab as first-line treatment of metastatic colorectal cancer: results of the TREE Study.J Clin Oncol. 2008 Jul 20; 26(21):3523-9.
- Imai T, Taketani M, Shii M, Hosokawa M, Chiba K. Substrate specificity of carboxylesterase isozymes and their contribution to hydrolase activity in human liver and small intestine. Drug Metab Dispos (2006); 34:1734–1741.
- Ribelles N, Lopez-Siles J, Sanchez A, Gonzalez E, Sanchez MJ, Carabantes F et al. A carboxylesterase 2 gene polymorphism as predictor of capecitabine on response and time to progression. Curr Drug Metab. 2008; 9 (4):336–343.
- Hagiwara, Hidaka, H. Cyclic nucleotides phosphodiesterases inhibitors. Nihon Rinsho. 49(9): 1993-8.
- Temmink, O. H.de Bruin, M. Laan, A. C. Turksma, A. W. Cricca, S. Masterson, A. J. Noordhuis, P. Peters G. J. The role of thymidine phosphorylase and uridine phosphorylase in (fluoro) pyrimidine metabolism in peripheral blood mononuclear cells. Int J Biochem Cell Biol. 2006: 38(10): 1759-65.
- Shimada H, Takeda A, Shiratori T, Nabeya Y, Okazumi S,Matsubara H, Funami Y, Hayashi H, Gunji Y, Kobayashi S,Suzuki T and Ochiai T: Prognostic significance of serumthymidine phosphorylase concentration in esophageal squamouscell carcinoma. Cancer 2002; 94: 1947-1954.
- Stenman, G. Sahlin, P. Dumanski, J. P. Hagiwara, K. Ishikawa, F. Miyazono, K. Collins, V. P. Heldin, C. H. Regional localization of the human platelet-derived endothelial cell growth factor (ECGF1) gene to chromosome 22q13. Cytogenet Cell Genet. 1992; 59(1): 22-3.
- Furukawa T, Yoshimura A, Sumizawa T, Haraguchi M, Akiyama S, Fukui K, Ishizawa M and Yamada Y: Angiogenic factor. Nature 1992;356: 668.
- Shimaoka S, Matsushita S, Nitanda T, Matsuda A, Nioh T, Suenaga T, Nishimata Y, Akiba S, Akiyama S and Nishimata H:The role of thymidine phosphorylase expression in the invasiveness of gastric carcinoma. Cancer 2000; 88: 2220-2227.
- Sawase K, Nomata K, Kanetake H and Saito Y: The expressionof platelet-derived endothelial cell growth factor in humanbladder cancer. Cancer Lett1998; 14: 35-41.
- Droller MJ: Prognostic significance of platelet-derivedendothelial cell growth factor/thymidine phosphorylase expression in stage pT1 G3 bladder cancer. J Urol 2002; 168: 854.
- Jones A, Fujiyama C, Turner K, Cranston D, Williams K, Stratford I, Bicknell R and Harris AL: Role of thymidinephosphorylase in an in vitro model of human bladder cancerinvasion. J Urol 2002; 167: 1482-1486.
- Bronckaers A, Gago F, Balzarini J, Liekens S. The dual role of thymidine phosphorylase in cancer development and chemotherapy. Med Res Rev. 2009;29:903-53
- Yu EJ, Lee Y, Rha SY, Kim TS, Chung HC, Oh BK, Yang WI, Noh SH, Jeung HC. Angiogenic factorthymidine phosphorylase increases cancer cell invasion activity in patients with gastricadenocarcinoma. Mol Cancer Res. 2008; 6:1554-66.
- Boskos, C. S. Liacos, C. Korkolis, D. Aygerinos, K. Lamproglou, I. Terpos, E. Stoupa et al. Thymidine phosphorylase to dihydropyrimidine dehydrogenase ratio as a predictive factor of response to preoperative chemoradiation with capecitabine in patients with advanced rectal cancer. J Surg Oncol. 2010 Oct 1; 102(5):408-12.
- Meropol, N. J. Gold, P. J. Diasio, R. B. Andria, M .Dhami, M. Godfrey, T. Kovatich, A. J. Lund, K. A. Mitchell, E. Schwarting, R. Thymidine phosphorylase expression is associated with response to capecitabine plus irinotecan in patients with metastatic colorectal cancer. J Clin Oncol.2006 Sep 1; 24(25):4069-77.
- Petrioli R, Bargagli G, Lazzi S, Pascucci A, Francini E, Bellan C, Conca R, Martellucci I, Fiaschi AI, Lorenzi B, Francini G; Multidisciplinary Oncology Group in Gastrointestinal Tumors. Thymidine phosphorylase expression in metastatic sites is predictive for response in patients with colorectal cancer treated with continuous oral capecitabine and biweekly oxaliplatin. Anticancer Drugs. 2010 Mar; 21(3):313-9.
- Lindskog EB, Derwinger K, Gustavsson B, Falk P, Wettergren Y. Thymidine phosphorylase expression is associated with time to progression in patients with metastatic colorectal cancer. BMC Clin Pathol. 2014 Jun 10; 14:25.
- Lu, M.Gao, J. Wang, X. C. Shen, L. Expressions of Thymidylate Synthase, Thymidine Phosphorylase, Class III beta-tubulin, and Excision Repair Cross-complementing Group 1predict Response in Advanced Gastric Cancer Patients Receiving Capecitabine Plus Paclitaxel or Cisplatin. Chin J Cancer Res. 2011 Dec; 23(4): 288–294.
- Gao, J. Lu, M.Yu, J. W. Li, Y. Y. Shen, L. Thymidine Phosphorylase/beta-tubulin III expressions predict the response in Chinese advanced gastric cancer patients receiving first-line capecitabine plus paclitaxel. BMC Cancer 2011;11:177
- Koizumi, W. Okayasu, I. Hyodo, I. Sakamoto, J. Kojima, H. Prediction of the effect of capecitabine in gastric cancer by immunohistochemical staining of thymidine phosphorylase and dihydropyrimidine dehydrogenase. Anticancer Drugs.2008 Sep;19(8):819-24
- Bonotto, M. Bozza, C. Di Loreto, C.Osa, E. O.Poletto, E.Puglisi, F. Making capecitabine targeted therapy for breast cancer: which is the role of thymidine phosphorylase. Clin Breast Cancer.2013 Jun;13(3):167-72
- Lee, S. J.Choi, Y. L.Park, Y. H.Kim, S. T.Cho, E. Y.Ahn, J. S.Im, Y. H. Thymidylate synthase and thymidine phosphorylase as predictive markers of capecitabine monotherapy in patients with anthracycline- and taxane-pretreated metastatic breast cancer. Cancer Chemother Pharmacol.2011 Sep;68(3):743-51
- Andreetta, C.Puppin, C.Minisini, A.Valent, F.Pegolo, E.Damante, G.Di Loreto, C.Pizzolitto, S.Pandolfi, M.Fasola, G.Piga, A.Puglisi, F.Thymidine phosphorylase expression and benefit from capecitabine in patients with advanced breast cancer. Ann Oncol.2009 Feb;20(2):265-71
- Saif MW, Juneja V, Black G, Thronton J, Johnson MR, Diasio RB.Support Cancer Ther. Palmar-plantar erythrodysesthesia in patients receiving capecitabine and intratumor thymidine phosphorylase and dihydropyrimidine dehydrogenase: is there a pharmacologic explanation? 2007 Sep 1;4(4):211-8.
- FitzgeraldSM, GoyalRK, OsborneWR, RoyJD, WilsonJW, FerrellRE. Identification of functional single nucleotide polymorphism haplotypes in the cytidine deaminase promoter.Hum Genet 2006; 119: 276–83.
- Mercier C, Dupuis C, Blesius A, Fanciullino R, Yang CG, Padovani L, Giacometti S, Frances N, Iliadis A, Duffaud F, Ciccolini J. Early severe toxicities after capecitabine intake: possible implication of a cytidine deaminase extensive metabolizer profile. Cancer Chemother Pharmacol.2009 May; 63(6):1177-80.
- Caronia D, Martin M, Sastre J, de la Torre J, García-Sáenz JA, Alonso MR, Moreno LT, Pita G, Díaz-Rubio E, Benítez J, González-Neira A.A polymorphism in the cytidine deaminase promoter predicts severe capecitabine-induced hand-foot syndrome. Clin Cancer Res.2011 Apr 1; 17(7):2006-13.
- Uchida K, Danenberg PV, Danenberg KD, Grem JL. Thymidylate synthase, dihydropyrimidine dehydrogenase, ERCC1, and thymidine phosphorylase gene expression in primary and metastatic gastrointestinal adenocarcinoma tissue in patients treated on a phase I trial of oxaliplatin and capecitabine.BMC Cancer. 2008 Dec 23; 8:386.
- S Lee, Y H Park, K H Kim, E Y Cho, Y C Ahn, K Kim,Y-M Shim, J S Ahn, K Park,and Y-H Im. Thymidine synthase, thymidine phosphorylase, and excision repair cross-complementation group 1 expression as predictive markers of capecitabine plus cisplatin chemotherapy as first-line treatment for patients with advanced oesophageal squamous cell carcinoma. Br J Cancer. Sep 7, 2010; 103(6): 845–851.
- Garcia, A. A.Blessing, J. A.Lenz, H. J.Darcy, K. M. Mannel, R. S.Miller, D. S.Husseinzadeh, N. Phase II clinical trial of capecitabine in ovarian carcinoma recurrent 6-12 months after completion of primary chemotherapy, with exploratory TS, DPD, and TP correlates: a Gynecologic Oncology Group study. Gynecol Oncol 2005:96(3): 810-7.
- Martinez-Balibrea E, Abad A, Aranda E et al. Pharmacogenetic approach forcapecitabine or 5-fluorouracil selection to be combined with oxaliplatin as firstlinechemotherapy in advanced colorectal cancer. Eur J Cancer 2008; 44(9):1229–1237.
- Spindler KL, Andersen RF, Jensen LH, Ploen J, Jakobsen A. EGF61A>G polymorphism as predictive marker of clinical outcome to first-line capecitabine and oxaliplatin in metastatic colorectal cancer. Ann Oncol 2010; 21 (3): 535-539.
- Horie N, Aiba H, Oguro K, Hojo H, Takeishi K: Functional analysis and DNA polymorphism of the tandemly repeated sequences in the 5'-terminal regulatory region of the human gene for thymidylate synthase. Cell Struct. Funct. 1995;20(3), 191–197
- Kawakami K, Omura K, Kanehira E, Watanabe Y: Polymorphic tandem repeats in the thymidylate synthase gene is associated with its protein expression in human gastrointestinal cancers. Anticancer Res 1999; 19(B4): 3249–3252
- Ruzzo A, Graziano F, Loupakis F et al.: Pharmacogenetic profiling in patients with advanced colorectal cancer treated with first-line FOLFOX-4 chemotherapy. J. Clin. Oncol. (2007); 25(10), 1247–1254.
- David J. Park, Jan Stoehlmacher, Wu Zhang, Denice Tsao-Wei, Susan Groshen, Heinz-Josef Lenz. Thymidylate synthase gene polymorphism predicts response to capecitabine in advanced colorectal cancer. Disease. January 2002; 17(1) : 46-49.
- Largillier R, Etienne-Grimaldi MC, Formento JL, Ciccolini J, Nebbia JF, Ginot A, Francoual M, Renée N, Ferrero JM, Foa C, Namer M, Lacarelle B, Milano G. Pharmacogenetics of capecitabine in advanced breast cancer patients. Clin Cancer Res. 2006 Sep 15; 12(18):5496-502.
- Gao, J.He, Q.Hua, D.Mao, Y.Li, Y.Shen, L. Polymorphism of TS 3'-UTR predicts survival of Chinese advanced gastric cancer patients receiving first-line capecitabine plus paclitaxel. Clin Transl Oncol. 2013:15(8): 619-25.
- Ulrich CM, Bigler J, Velicer CM, Greene EA, Farin FM, Potter JD. Searching expressed sequence tag databases: discovery and confirmation of a common polymorphismin the thymidylate synthase gene. Cancer Epidemiol Biomarkers Prev 2000; 9:1381- 5.
- Mandola MV, Stoehlmacher J, Zhang W, et al. A 6 bp polymorphism in the thymidylate synthase gene causes message instability and is associated with decreased intratumoral TS mRNA levels. Pharmacogenetics 2004; 14:319-27.
- A Loganayagam, M Arenas Hernandez, A Corrigan, L Fairbanks, C M Lewis, P Harper, N Maisey, P Ross, J D Sanderson and A M Marinaki. Pharmacogenetic variants in the DPYD, TYMS, CDA and MTHFR genes are clinically significant predictors of fluoropyrimidine toxicity. British Journal of Cancer 2013;108: 2505–2515
- Sharma R, Hoskins JM, Rivory LP, Zucknick M, London R, Liddle C, Clarke SJ Thymidylate synthase and methylenetetrahydrofolate reductase gene polymorphisms and toxicity to capecitabine in advanced colorectal cancer patients. Clin Cancer Res 2008; 14(3): 817–825.
- Braun MS, Richman SD, Thompson L, Daly CL, Meade AM, Adlard JW, Allan JM, Parmar MK, Quirke P, Seymour MT .Association of molecular markers with toxicity outcomes in a randomized trial of chemotherapy for advanced colorectal cancer: the FOCUS trial. J Clin Oncol 2009; 27(33): 5519–5528.
- Lurje G, Manegold PC, Ning Y, Pohl A, Zhang W, Lenz HJ. Thymidylate synthase gene variations: predictive and prognostic markers. Mol Cancer Ther 2009; 8(5): 1000–1007.
- Jakobsen A, Nielsen JN, Gyldenkerne N, Lindeberg J.Thymidylate synthase and methylenetetrahydrofolate reductase gene polymorphism in normal tissue as predictors of fluorouracil sensitivity. J. Clin. Oncol. 2005; 23(7), 1365–1369.
- Cohen V, Panet-Raymond V, Sabbaghian N, Morin I, Batist G, Rozen R: Methylenetetrahydrofolate reductase polymorphism in advanced colorectal cancer: a novel genomic predictor of clinical response to fluoropyrimidine-based chemotherapy. Clin. Cancer Res. 2003; 9(5), 1611–1615.
- van Huis-Tanja, L. H.Gelderblom, H.Punt, C. J.Guchelaar, H. J. MTHFR polymorphisms and capecitabine-induced toxicity in patients with metastatic colorectal cancer. Pharmacogenet Genomics 2013:23(4): 208-18.
- J. Dimovski, N. Angelovska-Petrusevska, B. Angelovska-Grozdanovska, A. Kapedanovska Nestorovska, N. Matevska, A. Eftimov and T. Josifovski. Predictive value of MSI, 18qLOH, TS promoter, and MTHFR C677T variants on efficacy/toxicity of capecitabine adjuvant monotherapy: A mid-term report of a prospective observational study on 136 stage II/III colon cancer patients. Journal of Clinical Oncology, 2010; 28(15). (suppl; abstr e14070) .
- Lei-Zhen Zheng, Li Zhang, Si-Yu Chen, Ji-Fang Gong, Jian-Chun Gu, Xiao-Ping Li, Jing Sun, Xiao-Dong Jiao, Jie-Jun Wang. Relationship between polymorphisms of related genes in the folic acid metabolic pathway and the survival of gastric cancer patients treated with capecitabine combined with paclitaxel. Tumor 2011;31(5):442-447.
- Joseph Ciccolini, Eva Gross, Laetitia Daha, Bruno Lacarelle, Cédric Mercier.Routine Dihydropyrimidine Dehydrogenase Testing for Anticipating 5-Fluorouracil–Related Severe Toxicities: Hype or Hope? Clinical Colorectal Cancer. October 2010; 9(4):224–228.
- Mc Leod H.L., Collie-Duguid E.S., Vreken P., et al. Nomenclature for human DPYD alleles. Pharmacogenetics. 1998; 8:455-459.
- Zhu AX, Puchalski TA, Stanton VP Jr et al. Dihydropyrimidine dehydrogenase and thymidylate synthase polymorphisms and their association with 5-fluorouracil/leucovorin chemotherapy in colorectal cancer. Clin. Colorectal Cancer (2004);3(4); 225–234.
- Salgado J, Zabalegui N, Gil C, Monreal I, Rodriguez J, Garcia-Foncillas J: Polymorphisms in the thymidylate synthase and dihydropyrimidine dehydrogenase genes predict response and toxicity to capecitabine-raltitrexed in colorectal cancer. Oncol. Rep. 2007; 17(2), 325–328.
- Wei X, McLeod HL, McMurrough J, Gonzalez FJ, Fernandez-Salguero P. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J Clin Invest.1996 Aug 1; 98(3):610-5.
- Maarten J. Deenen, Jolien Tol Artur M Burylo, Valerie D. Doodeman, Anthonius de Boer, Andrew Vincent, Henk-Jan Guchelaar, Paul H.M. Smits, Jos H. Beijnen , Cornelis J.A. Punt, Jan H.M. Schellens and Annemieke Cats. Relationship between Single Nucleotide Polymorphisms and Haplotypes in DPYD and Toxicity and Efficacy of Capecitabine in Advanced Colorectal Cancer. Clin Cancer Res.2011 May 15; 17(10):3455-68.
- Wasif Saif, Kostas Syrigos, Ranee Mehra, Lori K. Mattison, Robert B Diasio. Dihydropyrimidine Dehydrogenase Deficiency (Dpd) In GI Malignancies: Experience of 4-Years. Pak J Med Sci Q. 2007; 23(6): 832–839.
- Marzia Del Re, Fotios Loupakis, Cecilia Barbara, Tiziana Latiano, Enrico Vasile, Elena Zafarana. Impact of IVS14+1G>A and 2846A>T DPYD polymorphisms on toxicity outcome of patients treated with fluoropyrimidine-containing regimens. J Clin Oncol 2013; 31: (suppl; abstr 11058)
- Timur Ceric, Nermina Obralic, Lejla Kapur-Pojskic,Draženka Macic, Semir Bešlija, Anes Pašic, Šejla Ceric. Investigation of IVS14+1G>A polymorphism of DPYD gene in a group of Bosnian patients treated with 5-fluorouraciland capecitabine. Bosnian journal of basic medical sciences 2010; 10 (2): 133-139.
- Muhammad Wasif SaiF. Dihydropyrimidine Dehydrogenase Gene (DPYD) Polymorphism among Caucasian and non-Caucasian Patients with 5-FU- and Capecitabine-related Toxicity Using Full Sequencing of DPYD. Cancer Genomics and Proteomics 2013;10(2) 89-92.
- Loganayagam A, Arenas-Hernandez M, Fairbanks L, Ross P, Sanderson JD, Marinaki AM.The contribution of deleterious DPYD gene sequence variants to fluoropyrimidine toxicity in British cancer patients. Cancer Chemother Pharmacol. 2010 Jan; 65(2):403-6.
- González-Haba E. Severe capecitabine-associated toxicity in a patient carrying a mutation in the dihydropyrimidine dehydrogenase gen. Farm Hosp. 2012 Nov-Dec; 36(6):554-5.
- Hooiveld EA, van Kuilenburg AB, Haanen JB, Westermann AM. Severe toxicity after treatment with capecitabine and fluorouracil due to partial dihydropyrimidine dehydrogenase deficiency. Ned Tijdschr Geneeskd. 2004 Mar 27; 148(13):626-8.
- Toshima T, Kodera M, Yamashita Y, Oishi M, Seshimo K, Yamamura M, Kato H, Ikeda H, Mizuno K.A case in which dihydropyrimidine dehydrogenase deficiency was strongly suspected during adjuvant chemotherapy with capecitabine for colon cancer. See comment in PubMed Commons belowGan to Kagaku Ryoho. 2013 Nov; 40(11):1549-52.
- Saif MW, Elfiky A, Diasio R. Hand-foot syndrome variant in a dihydropyrimidine dehydrogenase-deficient patient treated with capecitabine. Clin Colorectal Cancer. 2006 Sep; 6(3):219-23.
- Coursier, S.Martelet, S.Guillermet, A.Emptoz, J.Villier, C. Severe toxicity following capecitabine administration because of dihydropyrimidine deshydrogenase (DPD) deficiency. Gastroenterol Clin Biol, 2010; 34:3: 218-23.
- Boisdron-Celle M, Remaud G, Traore S et al. 5-Fluorouracil-related severe toxicity: a comparison of different methods for the pretherapeutic detection of dihydropyrimidine dehydrogenase deficiency. Cancer Lett. (2007). 249 (2), 271–282.
- Ezzeldin H, Johnson MR, Okamoto Y, Diasio R. Denaturing high performance liquid chromatography analysis of the DPYD gene in patients with lethal 5-fluorouracil toxicity. Clin. Cancer Res. 2003; 9 (8), 3021–3028.
- Deenen MJ, Tol J, Burylo AM et al. Relationship between single nucleotide polymorphisms and haplotypes in DPYD and toxicity and efficacy of capecitabine in advanced colorectal cancer. Clin. Cancer Res. 2011; 17(10), 3455–3468.
- Kleibl Z, Fidlerova J, Kleiblova P et al. Influence of dihydropyrimidine dehydrogenase gene (DPYD) coding sequence variants on the development of fluoropyrimidine-related toxicity in patients with high-grade toxicity and patients with excellent tolerance of fluoropyrimidine-based chemotherapy. Neoplasma 2009; 56 (4), 303–316.
- Swen JJ, Nijenhuis M, de Boer A et al. Pharmacogenetics: from bench to byte – an update of guidelines. Clin. Pharmacol. Ther. 2011; 89 (5), 662–673.
- K E Caudle, C F Thorn, T E Klein, J J Swen, H L McLeod, R B Diasio, M Schwab. Clinical Pharmacogenetics Implementation Consortium Guidelines for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing. Clin Pharmacol Ther. Dec 2013; 94(6): 640–645.
- Blasco, H.Boisdron-Celle, M.Bougnoux, P.Calais, G.Tournamille, J. F.Ciccolini, J. Autret-Leca, E.Le Guellec, C. A well-tolerated 5-FU-based treatment subsequent to severe capecitabine-induced toxicity in a DPD-deficient patient. Br J Clin Pharmacol 2008: 65(6): 966-70.
- Garcia, A. A.Blessing, J. A.Lenz, H. J.Darcy, K. M. Mannel, R. S.Miller, D. S.Husseinzadeh, N. Phase II clinical trial of capecitabine in ovarian carcinoma recurrent 6-12 months after completion of primary chemotherapy, with exploratory TS, DPD, and TP correlates: a Gynecologic Oncology Group study. Gynecol Oncol 2005:96(3): 810-7.
- Saif, M. W, Sandoval, A Atypical hand-and-foot syndrome in an African American patient treated with capecitabine with normal DPD activity: is there an ethnic disparity? Cutan Ocul Toxicol.2008; 27(4):311-5.
- Vallbohmer, D. Yang, D. Y.Kuramochi, H.Shimizu, D. Danenberg, K. D.Lindebjerg, J.Nielsen, J. N.Jakobsen, A. Danenberg, P. V. DPD is a molecular determinant of capecitabine efficacy in colorectal cancer. Int J Oncol 2007: 31(2): 413-8.
- Nishimura G, Terada I, Kobayashi T, Ninomiya I, Kitagawa H, Fushida S, Fujimura T, Kayahara M, Shimizu K, Ohta T, Miwa K. Thymidine phosphorylase and dihydropyrimidine dehydrogenase levels in primary colorectal cancer show a relationship to clinical effects of 5'-deoxy-5-fluorouridine as adjuvant chemotherapy. Oncol Rep. 2002 May-Jun; 9(3):479-82.
How to cite this article:
Ramalakshmi S, Kavimani S, Srineevas S, Vetriselvi V and Bhaskar LVKS: Molecular Markers for Capecitabine Therapy: A Review. Int J Pharm Sci Res 2016; 7(11): 4315-26.doi: 10.13040/IJPSR.0975-8232.7(11).4315-26.
All © 2013 are reserved by International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
3
4315-26
580
1829
English
IJPSR
S. Ramalakshmi *, S. Kavimani, Satish Srineevas, V. Vetriselvi and LVKS Bhaskar
K. K. College of Pharmacy, Gerugambakkam, Chennai, Tamilnadu, India
sramalakshmi@rediffmail.com
20 May, 2016
28 July, 2016
02 August, 2016
10.13040/IJPSR.0975-8232.7(11).4315-26
01 November, 2016