| Home | E-Submission | Sitemap | Contact Us |  
top_img
Cancer Research and Treatment > Volume 48(3); 2016 > Article
Kim, Han, Lee, Cha, Lee, Kim, Oh, Kim, Im, Bang, and Kim: Splenomegaly and Its Associations with Genetic Polymorphisms and Treatment Outcome in Colorectal Cancer Patients Treated with Adjuvant FOLFOX

Abstract

Purpose

Splenomegaly is a clinical surrogate of oxaliplatin-induced sinusoidal obstruction syndrome (SOS). We investigated development of splenomegaly and its association with treatment outcome and genetic polymorphisms following adjuvant 5-fluorouracil, leucovorin, and oxaliplatin (FOLFOX) in colorectal cancer (CRC) patients.

Materials and Methods

Splenomegaly was determined by spleen volumetry using computed tomography images obtained before initiation of chemotherapy and after completion of adjuvant FOLFOX in CRC patients. Ten genetic polymorphisms in 4 SOS-related genes (VEGFA, MMP9, NOS3, and GSTP1) were analyzed using DNA from peripheral blood mononuclear cells.

Results

Of 124 patients included, increase in spleen size was observed in 109 (87.9%). Median change was 31% (range, –42% to 168%). Patients with splenomegaly had more severe thrombocytopenia compared to patients without splenomegaly during the chemotherapy period (p < 0.0001). The cumulative dose of oxaliplatin and the lowest platelet count during the chemotherapy period were clinical factors associated with splenomegaly. However, no significant associations were found between genetic polymorphisms and development of splenomegaly. Disease-free survival was similar regardless of the development of splenomegaly.

Conclusion

Splenomegaly was frequently observed in patients receiving adjuvant FOLFOX and resulted in more severe thrombocytopenia but did not influence treatment outcome. Examined genetic polymorphisms did not predict development of splenomegaly.

Introduction

Colorectal cancer (CRC) is a common malignancy and a leading cause of cancer death worldwide [1], and the incidence of CRC is increasing rapidly in Eastern countries including Korea as a result of recent changes in diet and lifestyle [2]. The primary treatment of CRC is a complete resection in stage I-III patients followed by adjuvant chemotherapy in stage III and high-risk stage II patients. In the considerable number of patients with distant metastases (i.e., stage IV disease) at the time of diagnosis, chemotherapy is the mainstay of treatment and selected patients with resectable metastases undergo surgery. Complete resection of colorectal liver metastases can provide a chance for long-term survival in approximately 20% of these patients [3].
Oxaliplatin plus fluoropyrimidine combination chemotherapy is widely used in both the adjuvant and palliative setting to improve survival of CRC patients [4,5]. With increasing incorporation of liver metastasectomy in the treatment strategy, many patients treated with oxaliplatin-containing chemotherapy undergo hepatic resection. Sinusoidal obstruction syndrome (SOS) in the liver is a long-term toxicity of oxaliplatin reported in patients who underwent hepatic resection after oxaliplatin-based chemotherapy [6-10].
SOS, previously known as hepatic veno-occlusive disease, was most commonly reported to occur after bone marrow transplantation for hematologic malignancies [11]. SOS has been reported in 19%-79% of CRC patients who underwent hepatic resection after oxaliplatin-based chemotherapy [6,7,12]. Toxic effect of oxaliplatin on sinusoidal endothelial cells (SEC) causes disruption of the sinusoidal wall, subsequently causing congestive obstruction with impairment of sinusoidal blood flow [13]. As a result, diffuse sinusoidal injury leads to portal hypertension, hepatomegaly, and hyperbilirubinemia with severe complications such as ascites and variceal bleeding in rare cases [14]. In addition, development of SOS may be associated with increased morbidity and mortality following hepatic resection in patients treated with preoperative oxaliplatin-based regimens [7].
Prediction of SOS development is important in selection of proper candidates for hepatic resection and preoperative management of patients at risk of SOS. Increase in spleen size, elevated aspartate aminotransferase to platelet ratio index, and hyaluronic acid levels have been reported as reliable indicators of SOS [9,15,16]. However, there is no reliable biomarker for prediction of SOS before oxaliplatin treatment that could help in the decision of oxaliplatin use. In addition, association of the susceptibility to SOS with antitumor efficacy in the adjuvant setting has not been studied.
Many recent efforts have been made to clarify the pathogenesis of SOS at the molecular level and to use this as a molecular marker of SOS. A rat model based on monocrotaline gavage by Deleve et al. [17] suggested that SEC injury is a major initiating event of SOS. This model also suggested that additional mechanisms such as an increase in expression of matrix metalloproteinase-9 (MMP-9) (and to a lesser extent MMP-2), reduced synthesis in nitric oxide, and oxidative stress contribute to SEC injury. Vascular endothelial growth factor (VEGF) is known to regulate MMP-9 activation by inducing its expression, and the degree of the increase in VEGF serum level parallels the clinical severity of SOS [18]. Therefore, VEGF blockade may attenuate sinusoidal injury by down-regulating MMP-9 production [19]. In the liver, nitric oxide is produced by the nitric oxide synthase 3 (NOS3) expressed in the SEC. Decreased activity of glutathione S-transferase (GST) leads to a decrease of adduct formation between glutathione and platinum, consequently attenuating a defense mechanism against oxaliplatin. In this study, we have chosen several genes related to the pathogenesis of SOS (VEGFA, MMP9, NOS3, and GSTP1), as shown in the previous model, and studied the association between their genetic polymorphisms and SOS following oxaliplatin treatment using spleen size as a surrogate of SOS. CRC patients receiving adjuvant 5-fluorouracil, leucovorin, and oxaliplatin (FOLFOX) were selected in order to eliminate the potential bias caused by the extent of liver metastasis. We also analyzed the association of SOS with disease-free survival (DFS) following adjuvant FOLFOX.

Materials and Methods

1. Study population and treatment

This retrospective analysis included stage III or high-risk stage II CRC patients who received adjuvant FOLFOX chemotherapy after complete resection of CRC from September 2005 to December 2009 at Seoul National University Hospital (SNUH) and participated in pharmacogenomics study of chemotherapy among cancer patients. Other eligibility criteria were age over 18 years, adenocarcinoma histology, complete resection of primary tumor with negative margin, adequate organ function, completion of at least six cycles of the planned 12 cycles of chemotherapy, and computed tomography (CT) images obtained before and after chemotherapy adequate for measurement of spleen size. For adequate measurement, contrast-enhanced CT images were obtained with at least 5 mm slice thickness or less. Patients with underlying severe liver disease such as active viral hepatitis, severe steatohepatitis, or liver cirrhosis before chemotherapy were excluded.
Patients received a maximum of 12 cycles of FOLFOX-4 or modified FOLFOX-6. Each cycle of FOLFOX-4 consisted of oxaliplatin (85 mg/m2) on day 1 and leucovorin (200 mg/m2) and a bolus of 5-fluorouracil (5-FU; 400 mg/m2) followed by a 22-hour infusion of 5-FU (600 mg/m2) on days 1 and 2, which was repeated every 2 weeks. Modified FOLFOX-6 consisted of oxaliplatin (85 mg/m2), leucovorin (400 mg/m2), and a bolus of 5-FU (400 mg/m2) followed by a 46-hour infusion of 5-FU (2,400 mg/m2) repeated every 2 weeks. All patients gave informed consent to the pharmacogenomics study prior to peripheral blood collection. The study protocol was reviewed and approved by the institutional review board of SNUH and was conducted in accordance with the Declaration of Helsinki.

2. Genotyping

Genomic DNA was extracted from peripheral blood mononuclear cells using a QIAamp DNA kit (Qiagen, Valencia, CA), and each polymorphism was determined using a pyrosequencing method (PyroMark Q96 ID, Qiagen). Primer sequence and biological effect of each polymorphism are shown in Supplementary Table 1. Ten polymorphisms in 4 potentially SOS-related genes were analyzed: –2578C/A, –1154G/A, –634G/C, and 936C/T in VEGFA; –1562C/T, 836A/G, and 2003G/A in MMP9; –786T/C and 894G/T in NOS3, and Ile105Val in GSTP1.

3. Spleen size measurement

Spleen size was measured by loading the CT images into a 3D Workstation (Rapidia ver. 2.8, Infinitt Healthcare, Seoul, Korea) and using volume viewer software. The outline of the spleen on each axial image of CT scans was traced using an electronic free-curve in the software, cross-sectional areas were calculated, and then the sum of the areas was multiplied by slice thickness for calculation of spleen volume. Spleen sizes measured from CT images obtained before chemotherapy and after completion of chemotherapy were compared.

4. Statistical analysis

Association between change in spleen size and each genetic polymorphism was analyzed using Student’s t test. The relationship between splenomegaly and each polymorphism was also analyzed using Pearson’s chi-square or Fisher’s exact test. Splenomegaly was functionally defined as a ≥ 50% increase in spleen size after oxaliplatin-based chemotherapy.
Multivariate logistic regression analysis was performed to evaluate the effect of other clinicopathologic factors on splenomegaly. In this analysis, the backward stepwise regression model including only variables with a p-value < 0.10 in univariate analysis was used. Survival functions for DFS were estimated using the Kaplan-Meier method, and differences between groups were tested using a log-rank test. Two-sided p-values of < 0.05 were considered significant. Analyses were performed using SPSS ver. 17.0 (SPSS Inc., Chicago, IL).

Results

1. Patient characteristics and changes in spleen size

Of a total of 124 patients included in this study, 73 patients (58.9%) were male (Table 1). The median age was 60 years (range, 30 to 76 years). Patients received median 12 cycles (range, 6 to 12 cycles) of adjuvant FOLFOX; 108 patients (87.1%) completed all 12 cycles of adjuvant FOLFOX. The median cumulative dose of oxaliplatin was 935 mg/m2 (range, 459 to 1,020 mg/m2).
Increase in spleen size was observed in 109 patients (87.9%) after completion of chemotherapy compared with baseline size before chemotherapy. The median change in spleen size was 31% (range, –42% to 168%). Splenomegaly (≥ 50% increase) was observed in 47 patients (37.9%). The mean increase in spleen size was higher in patients receiving a full dose of oxaliplatin during 12 cycles (n=54) compared with patients receiving a reduced dose of oxaliplatin (n=70) (50.7±40.4% vs. 30.2±34.4%, p=0.003).

2. Factors associated with splenomegaly

Ten genetic polymorphisms within four genes were analyzed (VEGFA, MMP9, NOS3, and GSTP1). When the relationship between these genetic polymorphisms and change in spleen size or splenomegaly was analyzed in univariate analysis, no genetic polymorphism was associated with change in spleen size or splenomegaly (p-value, not significant) (Table 2).
In univariate analysis for development of splenomegaly, only the cumulative dose of oxaliplatin (full vs. reduced dose) was significantly associated with development of splenomegaly. No significant associations were found between splenomegaly and other clinical factors (age [> 65 years vs. ≤ 65 years], lowest platelet count during chemotherapy [< 75,000/mm3 vs. ≥ 75,000/mm3 ] and the presence of chronic liver disease). In multivariate logistic regression analysis performed using variables with p-values < 0.10 in univariate analysis, the cumulative dose of oxaliplatin and the lowest platelet count during the chemotherapy period were associated with splenomegaly (Table 3).

3. Splenomegaly and thrombocytopenia

During adjuvant FOLFOX chemotherapy and the follow-up period after completion of chemotherapy, patients with splenomegaly had lower values of mean platelet count compared to patients without splenomegaly. This difference in mean platelet count was the most prominent from 3 months to 6 months after initiation of FOLFOX (p < 0.05, by Student’s t test), and the difference was gradually reduced after completion of chemotherapy (Fig. 1). In addition, patients with splenomegaly had more severe thrombocytopenia compared to those without splenomegaly during the chemotherapy period (mean lowest platelet count, 85,000±28,000/mm3 vs. 115, 000±42,000/mm3; p < 0.001).

4. Splenomegaly and DFS

We also analyzed the association of splenomegaly, a surrogate marker of SOS, with treatment outcome of adjuvant FOLFOX chemotherapy. DFS was similar according to development of splenomegaly. The 3-year DFS rate was 89.1% (95% confidence interval [CI], 79.9% to 98.3%) in patients who developed splenomegaly and 85.7% (95% CI, 77.7% to 93.7%) in patients without splenomegaly (p=0.42 by log-rank test) (Fig. 2).

Discussion

In the current study, splenomegaly, a surrogate of SOS, was frequently observed after adjuvant FOLFOX chemotherapy in CRC patients. An increase in spleen size compared with baseline size before starting oxaliplatin-based chemotherapy was observed in 109 patients (87.9%), with a median increase in spleen size of 31%. Although the direct comparison was difficult due to the difference in the cumulative dose of oxaliplatin and treatment duration every study, this change in spleen size is comparable to that reported in another study [9].
The cumulative dose of oxaliplatin and the lowest platelet count during chemotherapy were clinical factors associated with splenomegaly. Patients with splenomegaly showed more severe thrombocytopenia than patients without splenomegaly during or after oxaliplatin-based chemotherapy. This is in line with a previous study reporting on the relationship between splenomegaly and thrombocytopenia, which suggested splenic sequestration induced by portal hypertension as a possible mechanism of thrombocytopenia in patients with SOS [9]. Thrombocytopenia related to SOS is common, but usually not severe [20]. In this study, thrombocytopenia less than 50,000 mm3 was only observed in five cases and there were no significant bleeding events. Thrombocytopenia due to oxaliplatin-induced SOS can be prolonged until 2-3 years after completion of oxaliplatin treatment [21]. This slow recovery in platelet count was also observed in our study (Fig. 1). Besides these clinical factors, oxaliplatin-induced SOS is infrequently presented with ascites, jaundice, and hepatomegaly suggestive of portal hypertension [14]. In our study, none of the patients showed clinically apparent symptoms and signs of portal hypertension except splenomegaly.
A number of studies have been conducted for discovery of molecular predictive biomarkers of SOS based on its pathogenesis [10,22,23]. Rubbia-Brandt et al. [22], who examined gene expression profiles in livers with oxaliplatin-induced SOS and matched normal controls, found 913 differentially expressed genes. Results of pathway analysis showed significant upregulation of expression in six pathways: acute phase response, coagulation system, hepatic fibrosis/hepatic stellate cell activation, and oxidative stress. In addition, angiogenic and hypoxic factors including VEGFC and hypoxia-inducible factor 1-alpha (HIF1A) were upregulated [22]. A similar study conducted by a French group also confirmed the upregulation of genes involved in angiogenesis and coagulation in oxaliplatin-induced sinusoidal injuries [23]. Glutathione forms an adduct with platinum by GST, which leads to detoxification of oxaliplatin. In another study evaluating the role of GST polymorphism as a risk factor for SOS, GSTM1-null genotype was significantly related to the presence of moderate-severe SOS [10].
We have analyzed the association between splenomegaly and genetic polymorphisms in four SOS-related genes. We hypothesized that these genes might be relevant to the pathogenesis of SOS, as suggested in an animal model [13]. In this model, upregulation of MMP-9 and subsequent decrease of nitric oxide contributed to SEC injury, as well as glutathione depletion followed by production of reactive oxygen species. However, we found no association between splenomegaly and the genetic polymorphisms. Further studies are needed to determine the role of other genes or polymorphisms as a risk factor of SOS.
Some studies have suggested a negative impact of oxaliplatin-related SOS on long term outcomes with early recurrence and decreased overall survival in patients with colorectal liver metastases [24]. In addition, more severe grade of SOS was correlated with lower histopathological tumor regression [25]. In our study, splenomegaly, used as a biomarker of SOS, showed no association with survival outcome. A possible explanation is that, unlike previous studies, our study was conducted in patients in the adjuvant setting without distant metastasis. In addition, the frequency of hepatic recurrence was not significantly different according to the development of splenomegaly (2.1% [1/47] in patients with splenomegaly vs. 5.2% [4/77] in patients without splenomegaly, p=0.65).
The current study has some limitations including the retrospective nature of the study and no histopathological confirmation of SOS. However, the homogeneity of the study population including only patients in the adjuvant setting is the strength of the study.

Conclusion

In summary, we show that splenomegaly occurred in 87.9% of CRC patients receiving adjuvant FOLFOX treatment and it was also associated with more severe thrombocytopenia. Importantly, the development of splenomegaly did not affect DFS. As we found no association between the genetic polymorphisms analyzed herein and development of splenomegaly, future studies investigating other biomarker candidates are warranted for prediction of SOS.

Electronic Supplementary Material

Supplementary materials are available at Cancer Research and Treatment website (http://www.e-crt.org).

Conflicts of Interest

Conflict of interest relevant to this article was not reported.

Acknowledgments

This research was supported by grants from the Korea Health Technology R&D Project (HI14C1277 and HI13C2163) through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health & Welfare and the Converging Research Center Program (2014M3C1A8048802) funded by the Ministry of Science, ICT and Future Planning, Republic of Korea.

Fig. 1.
Changes in platelet counts during or after chemotherapy in patients with or without splenomegaly. FOLFOX, 5-fluorouracil, leucovorin, and oxaliplatin. *p < 0.05, **p < 0.01.
crt-2015-296f1.gif
Fig. 2.
Kaplan-Meier survival curve of disease-free survival according to the development of splenomegaly
crt-2015-296f2.gif
Table 1.
Patient characteristics
Characteristic No. (%) (n=124)
Age
 Median (range, yr) 60 (30-76)
Sex
 Male 73 (58.9)
 Female 51 (41.1)
Primary site
 Proximal 36 (29.0)
 Distal 88 (71.0)
Stage
 II 16 (12.9)
 III 108 (87.1)
Underlying chronic liver diseasea) 21 (16.9)
Cycle of chemotherapy
 Median (range) 12 (6-12)
Cumulative dose of oxaliplatin
 Full (1,020 mg/m2) 54 (43.5)
 Reduced (< 1,020 mg/m2) 70 (56.5)

a) Chronic liver disease included chronic inactive viral hepatitis such as hepatitis B and C, nonalcoholic steatohepatitis and fatty liver diseases.

Table 2.
Associations between genetic polymorphisms and change in spleen size
Polymorphism No. (%) Change in spleen size
Splenomegalya)
Mean % p-valueb) No. (%) p-valuec)
VEGFA C2578A
 CC 69 (55.6) 38.6±39.3 0.86 28 (40.6) 0.49
 CA or AA 55 (44.4) 39.8±37.5 19 (34.5)
VEGFA C634G
 CC 22 (17.7) 45.8±32.3 0.37 9 (40.9) 0.75
 CG or GG 102 (82.3) 37.7±39.5 38 (37.3)
VEGFA C936T
 CC 75 (60.5) 38.6±37.9 0.85 27 (36.0) 0.59
 CT or TT 49 (39.5) 39.9±39.5 20 (40.8)
VEGFA G1154A
 GG 85 (68.5) 37.2±39.2 0.42 31 (36.5) 0.63
 GA or AA 39 (31.5) 43.2±36.8 16 (41.0)
MMP9 C1562T
 CC 87 (70.2) 37.8±36.8 0.56 30 (34.5) 0.23
 CT or TT 37 (29.8) 42.2±42.2 17 (45.9)
MMP9 A836G
 AA 19 (15.3) 42.8±36.6 0.65 7 (36.8) 0.92
 AG or GG 105 (84.7) 38.4±38.8 40 (38.1)
MMP9 G2003A
 GG 87 (70.2) 37.8±36.8 0.56 30 (34.5) 0.23
 GA or AA 37 (29.8) 42.2±42.2 17 (45.9)
NOS3 G894T
 GG 104 (83.9) 38.6±39.0 0.74 39 (37.5) 0.83
 GT or TT 20 (16.1) 41.8±35.5 8 (40.0)
NOS3 T786C
 TT 101 (81.5) 37.8±37.8 0.43 36 (35.6) 0.28
 TC 23 (18.5) 44.8±41.3 11 (47.8)
GSTP1 Ile105Val
 Ile/Ile 71 (57.3) 37.8±40.8 0.65 25 (35.2) 0.48
 Ile/Val or Val/Val 53 (42.7) 40.9±35.2 22 (41.5)

VEGFA, vascular endothelial growth factor A; MMP, matrix metalloproteinase; NOS, nitric oxide synthase; GST, glutathione S-transferase.

a) Splenomegaly was defined as a ≥ 50% increase in spleen size after oxaliplatin-based chemotherapy,

b) Student’s t test was used,

c) Pearson’s chi square or Fisher’s exact tests were used.

Table 3.
Univariate and multivariate analyses of development of splenomegaly
Characteristic Univariate analysis
Multivariate analysis
No. (%) p-value OR (95% CI) p-value
Age
 ≤ 65 yr 38 (41.8) 0.142 - -
 > 65 yr 9 (27.3) -
Oxaliplatin cumulative dose
 Reduced dose 19 (27.1) 0.005 1 (reference) 0.003
 Full dose 28 (51.9) 3.29 (1.50-7.18)
The lowest PLT count during chemotherapy
 ≥ 75,000/mm3 30 (33.3) 0.088 1 (reference) 0.040
 < 75,000/mm3 17 (50.0) 2.45 (1.04-5.76)
Presence of chronic liver diseasea)
 No 42 (40.8) 0.144 - -
 Yes 5 (23.8) -

OR, odds ratio; CI, confidence interval; PLT, platelet.

a) Chronic liver disease included chronic inactive viral hepatitis such as hepatitis B and C, nonalcoholic steatohepatitis and fatty liver diseases.

References

1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62:10–29.
crossref pmid
2. Jung KW, Won YJ, Kong HJ, Oh CM, Lee DH, Lee JS. Cancer statistics in Korea: incidence, mortality, survival, and prevalence in 2011. Cancer Res Treat. 2014;46:109–23.
crossref pmid pmc pdf
3. Choti MA, Sitzmann JV, Tiburi MF, Sumetchotimetha W, Rangsin R, Schulick RD, et al. Trends in long-term survival following liver resection for hepatic colorectal metastases. Ann Surg. 2002;235:759–66.
crossref pmid pmc
4. de Gramont A, Figer A, Seymour M, Homerin M, Hmissi A, Cassidy J, et al. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol. 2000;18:2938–47.
crossref pmid
5. Andre T, Boni C, Mounedji-Boudiaf L, Navarro M, Tabernero J, Hickish T, et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med. 2004;350:2343–51.
crossref pmid
6. Rubbia-Brandt L, Audard V, Sartoretti P, Roth AD, Brezault C, Le Charpentier M, et al. Severe hepatic sinusoidal obstruction associated with oxaliplatin-based chemotherapy in patients with metastatic colorectal cancer. Ann Oncol. 2004;15:460–6.
crossref pmid
7. Nakano H, Oussoultzoglou E, Rosso E, Casnedi S, Chenard-Neu MP, Dufour P, et al. Sinusoidal injury increases morbidity after major hepatectomy in patients with colorectal liver metastases receiving preoperative chemotherapy. Ann Surg. 2008;247:118–24.
crossref pmid
8. Angitapalli R, Litwin AM, Kumar PR, Nasser E, Lombardo J, Mashtare T, et al. Adjuvant FOLFOX chemotherapy and splenomegaly in patients with stages II-III colorectal cancer. Oncology. 2009;76:363–8.
crossref pmid
9. Overman MJ, Maru DM, Charnsangavej C, Loyer EM, Wang H, Pathak P, et al. Oxaliplatin-mediated increase in spleen size as a biomarker for the development of hepatic sinusoidal injury. J Clin Oncol. 2010;28:2549–55.
crossref pmid
10. Vreuls CP, Olde Damink SW, Koek GH, Winstanley A, Wisse E, Cloots RH, et al. Glutathione S-transferase M1-null genotype as risk factor for SOS in oxaliplatin-treated patients with metastatic colorectal cancer. Br J Cancer. 2013;108:676–80.
crossref pmid pmc
11. Levitsky J, Sorrell MF. Hepatic complications of hematopoietic cell transplantation. Curr Gastroenterol Rep. 2007;9:60–5.
crossref pmid
12. Vauthey JN, Pawlik TM, Ribero D, Wu TT, Zorzi D, Hoff PM, et al. Chemotherapy regimen predicts steatohepatitis and an increase in 90-day mortality after surgery for hepatic colorectal metastases. J Clin Oncol. 2006;24:2065–72.
crossref pmid
13. Rubbia-Brandt L. Sinusoidal obstruction syndrome. Clin Liver Dis. 2010;14:651–68.
crossref pmid
14. Rubbia-Brandt L, Lauwers GY, Wang H, Majno PE, Tanabe K, Zhu AX, et al. Sinusoidal obstruction syndrome and nodular regenerative hyperplasia are frequent oxaliplatin-associated liver lesions and partially prevented by bevacizumab in patients with hepatic colorectal metastasis. Histopathology. 2010;56:430–9.
crossref pmid
15. Soubrane O, Brouquet A, Zalinski S, Terris B, Brezault C, Mallet V, et al. Predicting high grade lesions of sinusoidal obstruction syndrome related to oxaliplatin-based chemotherapy for colorectal liver metastases: correlation with post-hepatectomy outcome. Ann Surg. 2010;251:454–60.
crossref pmid
16. van den Broek MA, Vreuls CP, Winstanley A, Jansen RL, van Bijnen AA, Dello SA, et al. Hyaluronic acid as a marker of hepatic sinusoidal obstruction syndrome secondary to oxaliplatin-based chemotherapy in patients with colorectal liver metastases. Ann Surg Oncol. 2013;20:1462–9.
crossref pmid
17. DeLeve LD, McCuskey RS, Wang X, Hu L, McCuskey MK, Epstein RB, et al. Characterization of a reproducible rat model of hepatic veno-occlusive disease. Hepatology. 1999;29:1779–91.
crossref pmid
18. Iguchi A, Kobayashi R, Yoshida M, Kobayashi K, Matsuo K, Kitajima I, et al. Vascular endothelial growth factor (VEGF) is one of the cytokines causative and predictive of hepatic venoocclusive disease (VOD) in stem cell transplantation. Bone Marrow Transplant. 2001;27:1173–80.
crossref pmid
19. Ribero D, Wang H, Donadon M, Zorzi D, Thomas MB, Eng C, et al. Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer. 2007;110:2761–7.
crossref pmid
20. Jung EJ, Ryu CG, Kim G, Kim SR, Park HS, Kim YJ, et al. Splenomegaly during oxaliplatin-based chemotherapy for colorectal carcinoma. Anticancer Res. 2012;32:3357–62.
pmid
21. Jardim DL, Rodrigues CA, Novis YA, Rocha VG, Hoff PM. Oxaliplatin-related thrombocytopenia. Ann Oncol. 2012;23:1937–42.
crossref pmid
22. Rubbia-Brandt L, Tauzin S, Brezault C, Delucinge-Vivier C, Descombes P, Dousset B, et al. Gene expression profiling provides insights into pathways of oxaliplatin-related sinusoidal obstruction syndrome in humans. Mol Cancer Ther. 2011;10:687–96.
crossref pmid
23. Agostini J, Benoist S, Seman M, Julie C, Imbeaud S, Letourneur F, et al. Identification of molecular pathways involved in oxaliplatin-associated sinusoidal dilatation. J Hepatol. 2012;56:869–76.
crossref pmid
24. Tamandl D, Klinger M, Eipeldauer S, Herberger B, Kaczirek K, Gruenberger B, et al. Sinusoidal obstruction syndrome impairs long-term outcome of colorectal liver metastases treated with resection after neoadjuvant chemotherapy. Ann Surg Oncol. 2011;18:421–30.
crossref pmid
25. Vreuls CP, Van Den Broek MA, Winstanley A, Koek GH, Wisse E, Dejong CH, et al. Hepatic sinusoidal obstruction syndrome (SOS) reduces the effect of oxaliplatin in colorectal liver metastases. Histopathology. 2012;61:314–8.
crossref pmid
Editorial Office
Korean Cancer Association
Room 1824, Gwanghwamun Officia
92 Saemunan-ro, Jongno-gu, Seoul 03186, Korea
TEL: +82-2-3276-2410   FAX: +82-2-792-1410   E-mail: journal@cancer.or.kr
About |  Browse Articles |  Current Issue |  For Authors and Reviewers
Copyright © Korean Cancer Association.                 Developed in M2PI