Skip to main content
Disease Markers logoLink to Disease Markers
. 2016 Jan 5;2016:8345286. doi: 10.1155/2016/8345286

Systemic Inflammatory Response Based on Neutrophil-to-Lymphocyte Ratio as a Prognostic Marker in Bladder Cancer

Hyung Suk Kim 1, Ja Hyeon Ku 1,*
PMCID: PMC4736380  PMID: 26880857

Abstract

A growing body of evidence suggests that systemic inflammatory response (SIR) in the tumor microenvironment is closely related to poor oncologic outcomes in cancer patients. Over the past decade, several SIR-related hematological factors have been extensively investigated in an effort to risk-stratify cancer patients to improve treatment selection and to predict posttreatment survival outcomes in various types of cancers. In particular, one readily available marker of SIR is neutrophil-to-lymphocyte ratio (NLR), which can easily be measured on the basis of absolute neutrophils and absolute lymphocytes in a differential white blood cell count performed in the clinical setting. Many investigators have vigorously assessed NLR as a potential prognostic biomarker predicting pathological and survival outcomes in patients with urothelial carcinoma (UC) of the bladder. In this paper, we aim to present the prognostic role of NLR in patients with UC of the bladder through a thorough review of the literature.

1. Introduction

Cancer is a leading cause of morbidity and mortality presenting multifactorial features affected by a variety of factors, including tumor-related and host (patient)-related factors. Until recently, predicting outcomes in cancer patients have mainly depended upon tumor characteristics, such as pathologic tumor stage and tumor grade. However, various host-related factors, including weight loss (cachexia), performance status, and systemic inflammatory response (SIR), have been suggested as potential prognostic indicators in cancer patients.

Since Virchow first described a possible connection between inflammation and cancer in 1876 after observing the presence of leukocytes within neoplastic tissues [1], clear evidence now supports the crucial role played by SIR in the development, progression, metastasis, and survival of malignant cells in most cancers [2]. Most solid malignancies trigger an intrinsic inflammatory response that builds up a protumorigenic microenvironment. Inflammation in the tumor microenvironment may promote angiogenesis, invasion, and metastasis via the signaling of tumor-promoting chemokines and cytokines (i.e., IL-1, IL-6, tumor necrosis factor- [TNF-] a, and IL-23), which are produced by innate immune cells (macrophages, neutrophils, mast cells, myeloid-derived suppressor cells, dendritic cells, and natural killer cells) and adaptive ones (T and B lymphocytes) [1, 2]. Based on this background, in recent years many clinical studies have supported SIR as a meaningful predictor of survival outcomes in various types of cancers, including cancers of the lung [35], colorectum [611], gastrointestinal tract [12, 13], liver [14, 15], esophagus [1618], breast [1921], ovaries [2224], cervix [25, 26], and pancreas [27]. In addition, the prognostic value of SIR has been vigorously assessed in urologic cancers, including prostate cancer [2831], kidney cancer [3234], and urothelial carcinoma (UC) (cancers of the bladder [3548] and upper urinary tract (UUT) [4960]).

UC is the second-most frequently diagnosed urologic malignancy. Clinical outcomes vary. A majority of UCs (90~95%) originate in the bladder, and UC of the UUT only accounts for 5~10% of all UCs. Radical cystectomy (RC) and radical nephroureterectomy (RNU), respectively, are applied as the gold standard local treatment for muscle-invasive or high-risk, non-muscle-invasive UC of the bladder and UUT. However, in spite of these aggressive local approaches, long-term prognosis remains poor due to disease recurrence accompanied by local and/or distant metastasis [6163]. These poor outcomes suggest a need for ongoing risk stratification and proper selection of multimodal treatment approaches, such as chemotherapy in the neoadjuvant or adjuvant setting. To address these issues, a number of studies have explored SIR-related biomarkers as potential predictors of oncologic outcomes in UC. Among these, NLR, defined as the ratio of absolute neutrophils to absolute lymphocytes, has recently gained considerable attention as a biomarker in urothelial carcinoma (UC) arising from the bladder or upper urinary tract (UUT).

In this paper, we reviewed the clinical studies dealing with SIR-related biomarkers in association with oncologic outcomes in UC, with a special focus on NLR.

2. SIR-Based Prognostic Scoring System

Potential hematological biomarkers representing SIR in cancer patients include C-reactive protein (CRP), albumin, Glasgow Prognostic Score (GPS), modified GPS (mGPS), and neutrophil-to-lymphocyte ratio (NLR). The association of these SIR-related biomarkers with oncological outcomes has been extensively studied by many investigators in many types of nonurologic cancers (Table 1). Because hematological tests are routinely performed in most cancer patients, these biomarkers may be used as easily measurable, objective, reproducible, robust, and inexpensive parameters able to express the severity of SIR in cancer patients.

Table 1.

Clinical studies on the prognostic value of SIR-related hematological biomarkers in various types of cancers other than UC.

Study Marker Type of cancer Threshold Assessment period Results
Parker et al. [22] Albumin Ovarian cancer 3.5 & 4.1 g/dL Before operation Low-albumin level (continuous value) was associated with worse OS

Lis et al. [19] Albumin Breast cancer 3.5 g/dL Before operation Low-albumin level (<3.5 g/dL) was related to higher death rate

Lai et al. [7] Albumin Colon cancer 3.5 g/dL Before operation Hypoalbuminemia (<3.5 g/dL) was associated with increased morbidity and mortality

Seebacher et al. [67] Albumin Endometrial cancer 4.21 g/dL or continuous Before operation Increased albumin level (continuous) was related to better DFS and PFS

Hashimoto et al. [14] CRP HCC 1.0 mg/dL Before operation Elevated CRP (>1) was significant predictor of worse OS and RFS

Lehrer et al. [28] CRP Prostate cancer NA (continuous) Before radiation There was a significant correlation of CRP level with PSA

Crumley et al. [16] CRP Gastroesophageal cancer 1.0 mg/dL Before operation Elevated CRP (>1) was independent predictor of CSS

Jones et al. [4] CRP Lung cancer 0.4 mg/dL Before operation Elevated CRP (>0.4) was related to larger tumor size, advanced tumor stage, and incomplete resection

Karakiewicz et al. [32] CRP RCC 0.4 & 2.3 mg/dL Before nephrectomy Elevated CRP (>2.3) was an informative predictor of worse CSS

Beer et al. [29] CRP Metastatic prostate cancer 0.8 mg/dL Before docetaxel based chemotherapy Elevated CRP (>0.8) was a strong predictor of poor OS and lower PSA response to chemotherapy

Hefler et al. [23] CRP Ovarian cancer 1.0 mg/dL Before surgery Elevated CRP (>1.0 & continuous) was associated with postoperative residual tumor and worse OS

Shiu et al. [8] CRP Colorectal cancer 0.5 mg/dL Before surgery Elevated CRP (>0.5) was correlated with larger tumor size, higher stage, and poorer CSS

Crumley et al. [17] GPS Inoperable gastroesophageal cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before nonsurgical treatment High GPS was significant predictor of worse CSS

Al Murri et al. [20] GPS Metastatic breast cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before non-surgical treatment High GPS was significant predictor of worse CSS

Ramsey et al. [33] GPS Metastatic RCC 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before treatment High GPS was significant predictor of worse CSS

Polterauer et al. [25] GPS Cervical cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before surgery High GPS was significant predictor of worse OS and DFS

Vashist et al. [18] GPS Esophageal cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before surgery High GPS was a strong prognosticator of perioperative morbidity and worse DFS and OS

Kinoshita et al. [15] GPS HCC 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before treatment High GPS was independently associated with worse CSS

Leitch et al. [9] mGPS Colorectal cancer (operable or unresectable) 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before treatment High mGPS was independently associated with worse CSS in patients with either operable or unresectable colorectal cancer

Jiang et al. [13] mGPS Gastric cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before surgery High mGPS was independently associated with worse OS irrespective of cancer stage

Ishizuka et al. [10] mGPS Unresectable colorectal cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before chemotherapy High mGPS (1/2) was an independent risk factor of poor CSS

La Torre et al. [27] mGPS Pancreatic cancer 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before surgery High mGPS was independently associated with worse OS irrespective of cancer stage

Lamb et al. [34] mGPS RCC 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
Before surgery High mGPS was significantly independent predictors of worse OS and CSS

Cho et al. [24] NLR Ovarian cancer 2.6 Before surgery Positive NLR (>2.6) showed worse OS and DFS than negative NLR (<2.6)

Chua et al. [11] NLR Metastatic colorectal cancer 5 Before chemotherapy Elevated NLR (>5) was independently associated with less clinical response to chemotherapy and worse OS and PFS

Azab et al. [21] NLR Breast cancer Multiple cut-offs
(1.8, 2, 45, 3, 33)
Before chemotherapy High NLR (>3.3) was an independent significant predictor of all-cause mortality

Keizman et al. [70] NLR Metastatic CRPC 3 Before ketoconazole Low NLR (≤3.0) was significantly associated with better PFS

Keizman et al. [71] NLR Metastatic RCC 3 Before sunitinib Low NLR (≤3.0) was independent predictor of better response to sunitinib and favorable PFS and OS

Lee et al. [26] NLR Cervical cancer 1.9 Before treatment High NLR (≥1.9) was related to more advanced stage and increased NLR (continuous) was an independent predictor of worse PFS and OS

Yao et al. [5] NLR Advanced lung cancer 2.63 Before chemotherapy Low NLR (≤2.63) was independently associated with better clinical response to chemotherapy and favorable OS and PFS

RCC: renal cell carcinoma, HCC: hepatocellular carcinoma, CRP: C-reactive protein, GPS: Glasgow Prognostic Score, mGPS: modified Glasgow Prognostic Score, NLR: neutrophil-to-lymphocyte ratio, OS: overall survival, DFS: disease-free survival, PFS: progression-free survival, and CSS: cancer specific survival.

CRP is a nonspecific but sensitive marker of the acute phase response and is expressed in selected tumor cells [64]. The biological basis for the correlation between expression of this marker, cancer risk, outcome, and survival is not completely understood. Several proinflammatory cytokines, such as interleukin-1 (IL-1), IL-6, and TNF-a, expressed by the tumor environment induce CRP synthesis from the liver and other tissues [1, 2]. Based on many recent studies, it is now widely accepted that an elevated CRP value is a reliable indicator of poor prognosis in a variety of types of cancers [4, 8, 14, 16, 23, 28, 29, 31, 32, 65, 66].

Serum albumin, another marker of acute phase response to an inflammatory state, is generally used to assess nutritional status, severity of disease, disease progression, and prognosis [64]. Malnutrition and inflammation suppress albumin synthesis. In an adult, the normal range of serum albumin level is 3.5–5.0 g/dL. When levels drop below 3.5 g/dL, the condition is called hypoalbuminemia. The lower serum albumin concentration may be due to the production of cytokines such as IL-6, which modulate the production of albumin by hepatocytes [64]. Alternatively, TNF-a may increase the permeability of the microvasculature, thus allowing an increased transcapillary passage of albumin. Presence of micrometastatic tumor cells in the liver may induce the Kupffer cells to produce a variety of cytokines (IL-1, IL-6, and TNF-a), which may modulate albumin synthesis by hepatocytes [1, 2]. Thus, hypoalbuminemia is uncommon in early-stage cancer but as the disease progresses, albumin levels drop significantly and serve as good prognostic indicators in patients with various cancers [7, 19, 22, 67].

GPS and mGPS are inflammation-based prognostic scores developed by combining CRP and albumin to predict the clinical outcomes in cancer patients [68, 69]. GPS and mGPS, as routinely available, easily measured, and well standardized worldwide hematologic biomarkers, have subsequently been the subject of prognostic studies in wide variety of operable [13, 15, 18, 25, 27, 34] and inoperable [9, 10, 17, 20, 33] cancers. Indeed, these scoring systems have been extensively validated in various clinical scenarios and are now recognized to have prognostic value independent of tumor-based factors, such as pathologic tumor stage, tumor grade, lymphovascular invasion, and lymph node involvement.

It is also well recognized that SIR is related to changes in circulating white blood cells, especially an abnormal increase in neutrophils (neutrophilia) along with an abnormal decrease in lymphocytes (lymphocytopenia) [2, 64]. In light of this phenomenon under inflammatory conditions, NLR, being the ratio of neutrophils to lymphocytes, has gained considerable interest over the past decade not only as a potential prognostic factor associated with outcomes in a variety of cancers but also as a means of refining risk stratification of patients to treatment and predicting survival rates. Currently, NLR has been demonstrated to have significant prognostic value in urologic cancers, such as prostate [70] and renal cancer [71, 72], and also in cancers outside the urinary system [5, 11, 21, 24, 26].

3. SIR in Bladder Cancer

Prognosis in bladder cancer utilizes the same factors utilized for other types of cancers, including tumor-related factors, such as tumor stage, grade, lymphovascular invasion (LVI), and lymph node involvement (LNI) [61, 63]. However, all of these factors feature postoperative parameters. Given that SIR-related hematological biomarkers are easily obtained through pretreatment routine blood examination and have provided reliable prognostic information in other types of cancers, these biomarkers have been investigated in risk stratification for recurrence and mortality of patients with bladder cancer in both pre- and posttreatment settings. Several clinical studies have found an association between SIR-related hematological biomarkers, including CRP, albumin, and GPS, and oncologic outcomes of UC of the bladder (Table 3). In each different treatment setting, elevated CRP, defined as different cut-off (1.0 or 0.5 mg/dL),was significantly related to worse cancer-specific-survival (CSS) [35, 36]. One study demonstrated that in muscle-invasive bladder cancer (MIBC) patients with elevated CRP levels showed significantly more adverse pathologic features, such as extravesical disease (≥pT3), larger tumor size, lymph node involvement, and positive surgical margin prior to undergoing RC compared to patients with normal CRP levels. In addition, one-unit elevation in pre-RC CRP levels was significantly associated with a 20% increased risk of cancer-related death after RC [37]. In inoperable advanced bladder cancer, hypoalbuminemia and GPS 2 measured prior to chemotherapy were independently associated with shortened progression-free survival (PFS) and overall survival (OS), respectively [39]. Recently, Ku et al. developed a nomogram incorporating albumin, lymphocyte count, and platelet count to predict the probability of 5-year OS and disease-specific survival (DSS) after RC that demonstrated higher predictive accuracy than the existing staging system [46].

Table 3.

Clinical studies on the prognostic value of SIR-related hematological biomarkers in upper urinary tract urothelial carcinoma.

Study Marker Publication year Number of patients Threshold Assessment period Main findings
Saito et al. [49] CRP 2007 130 0.5 mg/dL Before surgery Patients with elevated (>0.5) CRP showed higher hemoglobin, advanced tumor stage (≥pT3), positive lymph node, high grade, and LVI; moreover, elevated (>0.5) CRP was significant prognostic factor for DSS and RFS

Obata et al. [52] CRP 2013 183 0.5 mg/dL Before surgery Patients with elevated (>0.5) CRP showed advanced tumor stage (≥pT3), LVI, and higher number of metastases; moreover, elevated (>0.5) CRP was significant prognostic factor for worse RFS and CSS

Tanaka et al. [58] CRP 2014 564 Multiple cut-offs (0.5, 2.0 mg/dL) Before surgery Elevated CRP (0.5–2.0 or >2.0) level was an independent predictor of worse RFS and CSS relative to normal CRP (≤0.5); in elevated pre-CRP (>0.5) group, postoperative normalization of CRP (≤0.5) was an independent predictor of better CSS

Ku et al. [55] Albumin 2014 181 3.5 g/dL Before surgery Hypoalbuminemia (<3.5) was a significant predictor of worse DSS and OS; also, scoring model incorporated albumin discriminated patients well according to risk of DSS and OS

Hashimoto et al. [51] Neutrophil count 2013 84 4000/uL Before surgery Elevated neutrophil count (≥4000/uL) was an independent prognostic factor for worse RFS

Azuma et al. [50] NLR 2013 137 2.5 Before surgery Elevated (≥2.5) NLR was significantly associated with worse RFS and CSS; also, scoring model incorporated NLR discriminated patients well according to risk of RFS and CSS

Dalpiaz et al. [53] NLR 2014 202 2.7 Before surgery Elevated (≥2.7) NLR was significantly associated with worse OS and CSS

Luo et al. [56] NLR 2014 234 3 Before surgery Elevated (≥3) NLR was significantly associated with worse MFS and CSS; also, the use of a NLR of >3 further identified a poor prognostic group, especially in patients with pT3 for MFS and CSS

Tanaka et al. [57] NLR 2014 665 3 Before surgery Patients with elevated (>3) NLR significantly showed high tumor grade (Gr 3), advanced tumor stage, positive lymph node, and LVI; elevated (≥3) NLR was an independent risk factor for worse RFS and CSS; furthermore, addition of pre-NLR slightly improved the accuracies of the base model for predicting both RFS and CSS

Sung et al. [60] NLR 2015 410 2.5 Before surgery Elevated NLR (≥2.5) was independent predictor of worse PFS, OS, and CSS, along with elevated ESR

CRP: C-reactive protein, NLR: neutrophil-to-lymphocyte ratio, LVI: lymphovascular invasion, OS: overall survival, DSS: disease-specific survival, RFS: recurrence-free survival, MFS: metastasis-free survival, and CSS: cancer specific survival.

4. NLR in Non-Muscle-Invasive Bladder Cancer (NMIBC)

To date, few studies have assessed the association between NLR and the prognosis of NMIBC initially treated with transurethral resection of the bladder tumor (TURBT). Indeed, the evaluation of the prognostic role of NLR has been conducted with focus on MIBC patients undergoing RC or a mixed cohort of muscle-invasive and non-muscle-invasive tumors (Table 2). One recent study assessed the predictive value of preoperative NLR in 107 patients initially diagnosed with NMIBC following TURBT [47]. When applying each different cut-off point for NLR using the standardized cut-off finder algorithm, NLR > 2.41 and NLR > 2.43 were significantly associated with unfavorable disease progression and recurrence. Owing to the limited sample size of this study, further studies will be required to validate the role of NLR as a predictor for recurrence and progression in NMIBC.

Table 2.

Clinical studies on the prognostic value of SIR-related hematological biomarkers in UC of the bladder.

Study Marker Publication year Number of patients (NMIBC/MIBC) Threshold Assessment period Main findings
Hilmy et al. [35] CRP 2005 105 (76/29) 1.0 mg/dL Before surgery (TURBT) Elevated preoperative CRP (>1) was independently associated with worse CSS

Yoshida et al. [36] CRP 2008 88 (0/88) 0.5 mg/dL Before radiochemotherapy Elevated preoperative CRP (≥0.5) was independent predictor of worse CSS

Gakis et al. [37] CRP 2011 246 (0/246) 0.5 mg/dL or continuous 1–3 days before RC Patients with elevated CRP (>0.5) showed advanced age, more extravesical disease, larger tumor size, node positive disease, and positive surgical margin and increased CRP (continuous) was independent predictor of worse CSS

Hwang et al. [39] GPS, Albumin 2012 67 (0/67) 1.0 mg/dL (CRP)
3.5 g/dL (Albumin)
1 day before first chemotherapy cycle Hypoalbuminemia (<3.5) and GPS 2 was independently associated with reduced PFS and OS, respectively

Ku et al. [46] Albumin
Neutrophil count
Platelet count
2015 419 (173/246) 3.5 g/dL (Albumin)
7500/uL (Neutrophil)
400 × 105/ul (Platelet)
Before RC Low albumin, high lymphocyte count, and high platelet count were significantly
associated with worse OS and CSS

Gondo et al. [38] NLR 2012 189 (62/127) 2.5 Before RC Elevated NLR (≥2.5) was an independent predictor of worse DSS

Demirtaş et al. [40] NLR 2013 201 (35/166) 2.5 Before RC Elevated NLR (>2.5) was not associated with overall survival

Hermanns et al. [41] NLR 2014 424 3 Before RC Patients with elevated NLR (≥3) significantly showed more advanced pathologic tumor stage
Elevated NLR (≥3) was significantly associated with RFS, OS, and CSS

Kaynar et al. [42] NLR 2014 291 (192/99) NA (continuous) 1 day before surgery (TURBT or RC) Patients with MIBC showed significantly higher NLR value than those with NMIBC
Also, higher NLR significantly correlated with advanced age, larger tumor size, and aggressive tumor invasiveness

Potretzke et al. [43] NLR 2014 102 (31/71) NA (continuous) Before RC NLR was significant predictor of pathological upstaging after RC; also, patients with pathological upstaging to ≥pT3 had a significantly greater NLR compared to patents who remained at ≤pT2

Viers et al. [44] NLR 2014 899 (392/507) 2.7 Within 90 days before RC Elevated NLR (≥2.7) was significantly associated with adverse pathologic finding (higher pathologic tumor stage, node positive, and larger tumor size); increased NLR (continuous) was independently associated with worse RFS, OS, and CSS

Mano et al. [47] NLR 2015 107 (107/0) 2.41 (for progression)
2.43 (for recurrence)
Before TURBT Elevated NLR (>2.41) showed more pT1 tumors and was significantly associated with disease progression;
elevated NLR (>2.43) was independent predictor of disease recurrence

Seah et al. [48] NLR 2015 26 (0/26) NA Before NACH, during NACH, and after RC Significant NLR decrease from before NACH to before RC was observed in patients with pathological response after NACH and RC

Kang et al. [45] NLR 2015 385 2.0 (postoperative)
2.1 (preoperative)
Within 1 month before RC and within 3 months after RC Patients with elevated postoperative NLR (≥2.0) had higher rates of ≥pT3, LVI, and positive lymph node and elevated postoperative NLR (≥2.0) was an independent predictor of OS and CSS; also, patients with perioperative continuous elevated NLR (2.1–>2.0) showed worse OS and CSS compared with other change groups

CRP: C-reactive protein, GPS: Glasgow Prognostic Score, mGPS: modified Glasgow Prognostic Score, NLR: neutrophil-to-lymphocyte ratio, TURBT: transurethral resection of bladder tumor, RC: radical cystectomy, NACH: neoadjuvant chemotherapy, NMIBC: nonmuscle invasive bladder cancer, OS: overall survival, DSS: disease-specific survival, RFS: recurrence-free survival, and CSS: cancer specific survival.

5. NLR in Muscle-Invasive Bladder Cancer (MIBC)

In the past five years, the prognostic role of NLR in MIBC has been actively investigated in association with various oncological outcomes, including pathologic outcome, post-RC recurrence, and survival (Table 2). Several studies evaluated the association between NLR and post-RC survival outcomes [38, 40, 41, 44]. The cut-off point chosen to define an elevated NLR differed across studies, ranging from 2.5 to 3. Although one study reported no significant association between elevated NLR and OS [40], elevated NLR has been regarded as an independent predictor of RFS (recurrence-free survival), OS, and CSS in most studies [38, 41, 44]. One study reported that higher NLR values were observed in MIBC patients compared with NMIBC patients [42]. In addition, several studies demonstrated a significant correlation between a higher NLR and adverse pathologic outcomes, such as larger tumor size, pathological upstaging to locally advanced disease (pT3), and LNI after RC [4144]. In locally advanced MIBC treated with neoadjuvant chemotherapy (NACH) prior to RC, continuous NLR decrease from before NACH to before RC was observed only in patients showing a pathological response after RC; therefore, sustained NLR decrease during NACH was suggested as a potential surrogate marker reflecting the effect of NACH [48]. The aforementioned studies mainly dealt with the prognostic value of NLR in the pretreatment setting. Interestingly, one recent study elucidated the influence of posttreatment NLR measured in the early post-RC period on oncologic outcomes [45]. The cut-off point of pre- and post-RC NLR (2.1 and 2.0, resp.) was differently determined according to each receiver operating characteristics (ROC) curve analysis. Similar to the aforementioned study results, elevated NLR after RC was also significantly associated with adverse pathologic outcomes, such as pT3/T4 disease, LVI, and LNI, and was an independent predictor of OS and CSS. Moreover, patients with perioperative continuous elevated NLR (2.1 ->2.0) showed worse OS and CSS compared with other change groups. Therefore, pre- and posttreatment NLR might have prognostic value in predicting postoperative survival outcome in patients with MIBC.

6. NLR in Upper Urinary Tract Urothelial Carcinoma (UTUC)

Similar to bladder cancer, the prognostic significance of other SIR-related hematological biomarkers, including CRP, albumin, and neutrophil count, has been proven to be reliable in terms of predicting adverse pathologic and survival outcomes following definitive surgery in UTUC [49, 51, 52, 55, 58]. In recent years, the prognostic role of NLR has also been vigorously assessed in UTUC [50, 53, 56, 57, 60] (Table 3). Although all of the studies involved cohorts of patients with operable UTUC, the threshold to determine elevated NLR levels was not uniform, ranging from 2.5 to 3. However, irrespective of the choice of NLR threshold, elevated NLR over the threshold was consistently correlated with adverse postoperative pathologic findings (high tumor grade, advanced tumor stage, LVI, and LNI) and worse survival outcomes following RNU.

7. Clinical Implications of SIR in Bladder Cancer

NMIBC can primarily be treated with TURBT. However, frequent recurrence (50~70%) and progression (10~20%) rates after TURBT are a major concern [61, 73]. Management of NMIBC might involve lifelong surveillance and place a considerable economic burden on patients. Currently, cystoscopy is the standard of care during the surveillance period. It is, however, invasive, and repeated cystoscopic examinations can cause substantial discomfort and pain to patients. Although investigators have developed various models to predict recurrence and progression after TURBT for NMIBC including nomogram, scoring systems, and risk tables [7477], these models mainly incorporated tumor-related factors, such as tumors number, tumor diameter, T category, World Health Organization (WHO) tumor grade, and carcinoma in situ (CIS). Considering the significant correlation of elevated NLR with disease recurrence and progression in NMIBC [47], the addition of NLR to the existing prediction model may contribute to more accurate stratification of patients with NMIBC according to risk of recurrence and progression. Also, according to risk stratification based on pretreatment NLR values, selective cystoscopic examination and additional treatment, including intravesical Bacillus Calmette-Guérin (BCG) immunotherapy or chemotherapy, will be possible in patients with high-risk NMIBC, thereby reducing their economic burden and the potential discomfort caused by repeated cystoscopy.

In terms of MIBC, one significant challenge has been the limited, pretreatment, risk-stratification data that exists for patients undergoing RC. The well-established risk factors for recurrence and survival in MIBC included tumor-related factors, including pathologic tumor stage, pathologic tumor grade, CIS, LVI, and LNI [7880]. Moreover, most predictive models (nomogram) predicting recurrence and survival in bladder cancer have been heavily based on postoperative pathologic factors, such as pathologic tumor stage, pathological grade, LVI, and LNI [8183], with minimal consideration for associated host-related factors. Meanwhile, the accuracy of clinical staging in bladder cancer remains poor, reporting upstaging rate of 50% at RC specimen [84]. Thus, not enough data exists to facilitate appropriate patient counseling and guide clinical trial enrollment. As such, it is required to identify biomarkers that can assist with preoperative patient risk stratification and counseling. To achieve these goals, SIR-related hematological biomarkers can be a potential and promising factor. Assessment of SIR-related biomarkers in bladder cancer may be particularly relevant, because the inflammatory process seems to play an important role in the genesis and progression of, as well as mortality from, bladder cancer [1, 2]. Based on the previous study result [43], demonstrating a significant association between pretreatment elevated NLR and pathologic upstaging after RC, the performance of early cystectomy or NACH prior to RC might be considered in patients with pretreatment high NLR to attain tumor downstaging and improve postoperative survival. In addition, the pattern of change in NLR during NACH will be a valuable surrogate marker for monitoring and predicting pathological response to NACH [48]. Several studies reported the incorporation of SIR-related hematological biomarkers, such as CRP, NLR, albumin, and lymphocyte and platelet count, with a predictive model for survival outcomes in MIBC [37, 38, 46] or UTUC [50, 55, 57], improved predictive accuracy of the model, and consequently discriminated patients well according to risk stratification. It follows that pretreatment evaluation of NLR will be helpful in counseling patients about their prognosis.

A recent study revealed that the NLR value measured during the early postoperative period (from 1 to 3 months) after RC had a significant correlation with adverse oncological and survival outcomes [45]. Thus, postoperative NLR and the pattern of NLR change in the perioperative period may also provide valuable information in determining which patients should be referred for additional multimodal treatment, such as radiation and adjuvant chemotherapy.

The limitations of current NLR-associated studies in cancer are as follows. First, as mentioned earlier, there was no uniform cut-off point for NLR; each threshold was adopted according to a variety of statistical methodologies. Unlike tumor-related prognostic factors, including pathologic tumor stage and grade, NLR as a host-related factor can be affected by a variety of physiologic conditions, such as patients' comorbidities (hypertension and diabetes mellitus) and type of cancer, which can trigger immune response to cancer so that the establishment of definite NLR threshold may be difficult in consideration of these changeable physiologic conditions among cancer patients. Second, nearly all of the studies were both clinical and retrospective. Further large-scale prospective clinical or experimental animal (preclinical) research using a unified and robust statistical methodology will be required to determine the definite cut-off value of NLR and to discover the biological mechanisms supporting the correlation between NLR and oncologic outcomes in cancer patients.

8. Conclusion

Elevated NLR has shown a significant association with adverse oncologic and survival outcomes in patients with UC. Thus, NLR as a potential marker of SIR may become a promising tool in the management of patients with UC of the bladder and UUT, in terms of improved risk assessment for prognosis and guidance for treatment. Moreover, the ease and convenience of routine blood examinations in the clinical setting mean that NLR can be an objective, inexpensive, reproducible, and cost-effective measurement for the prediction of prognosis in UC. However, current NLR-related studies have not applied uniform NLR thresholds and thus require cautious interpretation because of many statistical methodological limitations. For the introduction of NLR into the clinical practice, rigorous attempts should be made in proper prospective study design.

Conflict of Interests

The authors declare that there is no conflict of interests regarding the publication of this paper.

References

  • 1.Balkwill F., Mantovani A. Inflammation and cancer: back to virchow? The Lancet. 2001;357(9255):539–545. doi: 10.1016/s0140-6736(00)04046-0. [DOI] [PubMed] [Google Scholar]
  • 2.Grivennikov S. I., Greten F. R., Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Espinosa E., Feliu J., Zamora P., et al. Serum albumin and other prognostic factors related to response and survival in patients with advanced non-small cell lung cancer. Lung Cancer. 1995;12(1-2):67–76. doi: 10.1016/0169-5002(95)00407-R. [DOI] [PubMed] [Google Scholar]
  • 4.Jones J. M., McGonigle N. C., McAnespie M., Cran G. W., Graham A. N. Plasma fibrinogen and serum C-reactive protein are associated with non-small cell lung cancer. Lung Cancer. 2006;53(1):97–101. doi: 10.1016/j.lungcan.2006.03.012. [DOI] [PubMed] [Google Scholar]
  • 5.Yao Y., Yuan D., Liu H., Gu X., Song Y. Pretreatment neutrophil to lymphocyte ratio is associated with response to therapy and prognosis of advanced non-small cell lung cancer patients treated with first-line platinum-based chemotherapy. Cancer Immunology, Immunotherapy. 2013;62(3):471–479. doi: 10.1007/s00262-012-1347-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Heys S. D., Walker L. G., Deehan D. J., Eremin O. E. Serum albumin: a prognostic indicator in patients with colorectal cancer. Journal of the Royal College of Surgeons of Edinburgh. 1998;43(3):163–168. [PubMed] [Google Scholar]
  • 7.Lai C.-C., You J.-F., Yeh C.-Y., et al. Low preoperative serum albumin in colon cancer: a risk factor for poor outcome. International Journal of Colorectal Disease. 2011;26(4):473–481. doi: 10.1007/s00384-010-1113-4. [DOI] [PubMed] [Google Scholar]
  • 8.Shiu Y.-C., Lin J.-K., Huang C.-J., et al. Is C-reactive protein a prognostic factor of colorectal cancer? Diseases of the Colon and Rectum. 2008;51(4):443–449. doi: 10.1007/s10350-007-9133-z. [DOI] [PubMed] [Google Scholar]
  • 9.Leitch E. F., Chakrabarti M., Crozier J. E. M., et al. Comparison of the prognostic value of selected markers of the systemic inflammatory response in patients with colorectal cancer. British Journal of Cancer. 2007;97(9):1266–1270. doi: 10.1038/sj.bjc.6604027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ishizuka M., Nagata H., Takagi K., Kubota K. Influence of inflammation-based prognostic score on mortality of patients undergoing chemotherapy for far advanced or recurrent unresectable colorectal cancer. Annals of Surgery. 2009;250(2):268–272. doi: 10.1097/SLA.0b013e3181b16e24. [DOI] [PubMed] [Google Scholar]
  • 11.Chua W., Charles K. A., Baracos V. E., Clarke S. J. Neutrophil/lymphocyte ratio predicts chemotherapy outcomes in patients with advanced colorectal cancer. British Journal of Cancer. 2011;104(8):1288–1295. doi: 10.1038/bjc.2011.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dutta S., Crumley A. B. C., Fullarton G. M., Horgan P. G., McMillan D. C. Comparison of the prognostic value of tumour and patient related factors in patients undergoing potentially curative resection of gastric cancer. The American Journal of Surgery. 2012;204(3):294–299. doi: 10.1016/j.amjsurg.2011.10.015. [DOI] [PubMed] [Google Scholar]
  • 13.Jiang X., Hiki N., Nunobe S., et al. Prognostic importance of the inflammation-based Glasgow prognostic score in patients with gastric cancer. British Journal of Cancer. 2012;107(2):275–279. doi: 10.1038/bjc.2012.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hashimoto K., Ikeda Y., Korenaga D., et al. The impact of preoperative serum C-reactive protein on the prognosis of patients with hepatocellular carcinoma. Cancer. 2005;103(9):1856–1864. doi: 10.1002/cncr.20976. [DOI] [PubMed] [Google Scholar]
  • 15.Kinoshita A., Onoda H., Imai N., et al. Comparison of the prognostic value of inflammation-based prognostic scores in patients with hepatocellular carcinoma. British Journal of Cancer. 2012;107(6):988–993. doi: 10.1038/bjc.2012.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Crumley A. B. C., McMillan D. C., McKernan M., Going J. J., Shearer C. J., Stuart R. C. An elevated C-reactive protein concentration, prior to surgery, predicts poor cancer-specific survival in patients undergoing resection for gastro-oesophageal cancer. British Journal of Cancer. 2006;94(11):1568–1571. doi: 10.1038/sj.bjc.6603150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Crumley A. B. C., McMillan D. C., McKernan M., McDonald A. C., Stuart R. C. Evaluation of an inflammation-based prognostic score in patients with inoperable gastro-oesophageal cancer. British Journal of Cancer. 2006;94(5):637–641. doi: 10.1038/sj.bjc.6602998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Vashist Y. K., Loos J., Dedow J., et al. Glasgow prognostic score is a predictor of perioperative and long-term outcome in patients with only surgically treated esophageal cancer. Annals of Surgical Oncology. 2011;18(4):1130–1138. doi: 10.1245/s10434-010-1383-7. [DOI] [PubMed] [Google Scholar]
  • 19.Lis C. G., Grutsch J. F., Vashi P. G., Lammersfeld C. A. Is serum albumin an independent predictor of survival in patients with breast cancer? Journal of Parenteral and Enteral Nutrition. 2003;27(1):10–15. doi: 10.1177/014860710302700110. [DOI] [PubMed] [Google Scholar]
  • 20.Al Murri A. M., Bartlett J. M. S., Canney P. A., Doughty J. C., Wilson C., McMillan D. C. Evaluation of an inflammation-based prognostic score (GPS) in patients with metastatic breast cancer. British Journal of Cancer. 2006;94(2):227–230. doi: 10.1038/sj.bjc.6602922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Azab B., Bhatt V. R., Phookan J., et al. Usefulness of the neutrophil-to-lymphocyte ratio in predicting short- and long-term mortality in breast cancer patients. Annals of Surgical Oncology. 2012;19(1):217–224. doi: 10.1245/s10434-011-1814-0. [DOI] [PubMed] [Google Scholar]
  • 22.Parker D., Bradley C., Bogle S. M., et al. Serum albumin and CA125 are powerful predictors of survival in epithelial ovarian cancer. BJOG. 1994;101(10):888–893. doi: 10.1111/j.1471-0528.1994.tb13550.x. [DOI] [PubMed] [Google Scholar]
  • 23.Hefler L. A., Concin N., Hofstetter G., et al. Serum C-reactive protein as independent prognostic variable in patients with ovarian cancer. Clinical Cancer Research. 2008;14(3):710–714. doi: 10.1158/1078-0432.ccr-07-1044. [DOI] [PubMed] [Google Scholar]
  • 24.Cho H., Hur H. W., Kim S. W., et al. Pre-treatment neutrophil to lymphocyte ratio is elevated in epithelial ovarian cancer and predicts survival after treatment. Cancer Immunology, Immunotherapy. 2009;58(1):15–23. doi: 10.1007/s00262-008-0516-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Polterauer S., Grimm C., Seebacher V., et al. The inflammation-based glasgow prognostic score predicts survival in patients with cervical cancer. International Journal of Gynecological Cancer. 2010;20(6):1052–1057. doi: 10.1111/igc.0b013e3181e64bb1. [DOI] [PubMed] [Google Scholar]
  • 26.Lee Y.-Y., Choi C. H., Kim H.-J., et al. Pretreatment neutrophil:lymphocyte ratio as a prognostic factor in cervical carcinoma. Anticancer Research. 2012;32(4):1555–1561. [PubMed] [Google Scholar]
  • 27.La Torre M., Nigri G., Cavallini M., Mercantini P., Ziparo V., Ramacciato G. The glasgow prognostic score as a predictor of survival in patients with potentially resectable pancreatic adenocarcinoma. Annals of Surgical Oncology. 2012;19(9):2917–2923. doi: 10.1245/s10434-012-2348-9. [DOI] [PubMed] [Google Scholar]
  • 28.Lehrer S., Diamond E. J., Mamkine B., Droller M. J., Stone N. N., Stock R. G. C-reactive protein is significantly associated with prostate-specific antigen and metastatic disease in prostate cancer. BJU International. 2005;95(7):961–962. doi: 10.1111/j.1464-410x.2005.05447.x. [DOI] [PubMed] [Google Scholar]
  • 29.Beer T. M., Lalani A. S., Lee S., et al. C-reactive protein as a prognostic marker for men with androgen-independent prostate cancer. Cancer. 2008;112(11):2377–2383. doi: 10.1002/cncr.23461. [DOI] [PubMed] [Google Scholar]
  • 30.McArdle P. A., Qayyum T., McMillan D. C. Systemic inflammatory response and survival in patients with localised prostate cancer: 10-year follow-up. Urologia Internationalis. 2010;84(4):430–435. doi: 10.1159/000313364. [DOI] [PubMed] [Google Scholar]
  • 31.Liu Z.-Q., Chu L., Fang J.-M., et al. Prognostic role of C-reactive protein in prostate cancer: a systematic review and meta-analysis. Asian Journal of Andrology. 2014;16(3):467–471. doi: 10.4103/1008-682x.123686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Karakiewicz P. I., Hutterer G. C., Trinh Q.-D., et al. C-reactive protein is an informative predictor of renal cell carcinoma-specific mortality: a European study of 313 patients. Cancer. 2007;110(6):1241–1247. doi: 10.1002/cncr.22896. [DOI] [PubMed] [Google Scholar]
  • 33.Ramsey S., Lamb G. W. A., Aitchison M., Graham J., McMillan D. C. Evaluation of an inflammation-based prognostic score in patients with metastatic renal cancer. Cancer. 2007;109(2):205–212. doi: 10.1002/cncr.22400. [DOI] [PubMed] [Google Scholar]
  • 34.Lamb G. W. A., Aitchison M., Ramsey S., Housley S. L., McMillan D. C. Clinical utility of the glasgow prognostic score in patients undergoing curative nephrectomy for renal clear cell cancer: basis of new prognostic scoring systems. British Journal of Cancer. 2012;106(2):279–283. doi: 10.1038/bjc.2011.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hilmy M., Bartlett J. M. S., Underwood M. A., McMillan D. C. The relationship between the systemic inflammatory response and survival in patients with transitional cell carcinoma of the urinary bladder. British Journal of Cancer. 2005;92(4):625–627. doi: 10.1038/sj.bjc.6602406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yoshida S., Saito K., Koga F., et al. C-reactive protein level predicts prognosis in patients with muscle-invasive bladder cancer treated with chemoradiotherapy. BJU International. 2008;101(8):978–981. doi: 10.1111/j.1464-410X.2007.07408.x. [DOI] [PubMed] [Google Scholar]
  • 37.Gakis G., Todenhöfer T., Renninger M., et al. Development of a new outcome prediction model in carcinoma invading the bladder based on preoperative serum C-reactive protein and standard pathological risk factors: the TNR-C score. BJU International. 2011;108(11):1800–1805. doi: 10.1111/j.1464-410x.2011.10234.x. [DOI] [PubMed] [Google Scholar]
  • 38.Gondo T., Nakashima J., Ohno Y., et al. Prognostic value of neutrophil-to-lymphocyte ratio and establishment of novel preoperative risk stratification model in bladder cancer patients treated with radical cystectomy. Urology. 2012;79(5):1085–1091. doi: 10.1016/j.urology.2011.11.070. [DOI] [PubMed] [Google Scholar]
  • 39.Hwang E. C., Hwang I. S., Yu H. S., et al. Utility of inflammation-based prognostic scoring in patients given systemic chemotherapy first-line for advanced inoperable bladder cancer. Japanese Journal of Clinical Oncology. 2012;42(10):955–960. doi: 10.1093/jjco/hys124. [DOI] [PubMed] [Google Scholar]
  • 40.Demirtaş A., Sabur V., Aknsal E. C., et al. Can neutrophil-lymphocyte ratio and lymph node density be used as prognostic factors in patients undergoing radical cystectomy? The Scientific World Journal. 2013;2013:5. doi: 10.1155/2013/703579.703579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hermanns T., Bhindi B., Wei Y., et al. Pre-treatment neutrophil-to-lymphocyte ratio as predictor of adverse outcomes in patients undergoing radical cystectomy for urothelial carcinoma of the bladder. British Journal of Cancer. 2014;111(3):444–451. doi: 10.1038/bjc.2014.305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kaynar M., Yıldırım M. E., Badem H., et al. Bladder cancer invasion predictability based on preoperative neutrophil-lymphocyte ratio. Tumor Biology. 2014;35(7):6601–6605. doi: 10.1007/s13277-014-1889-x. [DOI] [PubMed] [Google Scholar]
  • 43.Potretzke A., Hillman L., Wong K., et al. NLR is predictive of upstaging at the time of radical cystectomy for patients with urothelial carcinoma of the bladder. Urologic Oncology: Seminars and Original Investigations. 2014;32(5):631–636. doi: 10.1016/j.urolonc.2013.12.009. [DOI] [PubMed] [Google Scholar]
  • 44.Viers B. R., Boorjian S. A., Frank I., et al. Pretreatment neutrophil-to-lymphocyte ratio is associated with advanced pathologic tumor stage and increased cancer-specific mortality among patients with urothelial carcinoma of the bladder undergoing radical cystectomy. European Urology. 2014;66(6):1157–1164. doi: 10.1016/j.eururo.2014.02.042. [DOI] [PubMed] [Google Scholar]
  • 45.Kang M., Jeong C. W., Kwak C., Kim H. H., Ku J. H. The prognostic significance of the early postoperative neutrophil-to-lymphocyte ratio in patients with urothelial carcinoma of the bladder undergoing radical cystectomy. Annals of Surgical Oncology. 2015:1–8. doi: 10.1245/s10434-015-4708-8. [DOI] [PubMed] [Google Scholar]
  • 46.Ku J. H., Kang M., Kim H. S., Jeong C. W., Kwak C., Kim H. H. The prognostic value of pretreatment of systemic inflammatory responses in patients with urothelial carcinoma undergoing radical cystectomy. British Journal of Cancer. 2015;112(3):461–467. doi: 10.1038/bjc.2014.631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mano R., Baniel J., Shoshany O., et al. Neutrophil-to-lymphocyte ratio predicts progression and recurrence of non–muscle-invasive bladder cancer. Urologic Oncology: Seminars and Original Investigations. 2015;33:67.e1–67.e7. doi: 10.1016/j.urolonc.2014.06.010. [DOI] [PubMed] [Google Scholar]
  • 48.Seah J.-A., Leibowitz-Amit R., Atenafu E. G., et al. Neutrophil-lymphocyte ratio and pathological response to neoadjuvant chemotherapy in patients with muscle-invasive bladder cancer. Clinical Genitourinary Cancer. 2015;13(4):e229–e233. doi: 10.1016/j.clgc.2015.02.001. [DOI] [PubMed] [Google Scholar]
  • 49.Saito K., Kawakami S., Ohtsuka Y., et al. The impact of preoperative serum C-reactive protein on the prognosis of patients with upper urinary tract urothelial carcinoma treated surgically. BJU International. 2007;100(2):269–273. doi: 10.1111/j.1464-410x.2007.06934.x. [DOI] [PubMed] [Google Scholar]
  • 50.Azuma T., Matayoshi Y., Odani K., et al. Preoperative neutrophil-lymphocyte ratio as an independent prognostic marker for patients with upper urinary tract urothelial carcinoma. Clinical Genitourinary Cancer. 2013;11(3):337–341. doi: 10.1016/j.clgc.2013.04.003. [DOI] [PubMed] [Google Scholar]
  • 51.Hashimoto T., Ohno Y., Nakashima J., Gondo T., Ohori M., Tachibana M. Clinical significance of preoperative peripheral blood neutrophil count in patients with non-metastatic upper urinary tract carcinoma. World Journal of Urology. 2013;31(4):953–958. doi: 10.1007/s00345-012-0942-x. [DOI] [PubMed] [Google Scholar]
  • 52.Obata J., Kikuchi E., Tanaka N., et al. C-reactive protein: a biomarker of survival in patients with localized upper tract urothelial carcinoma treated with radical nephroureterectomy. Urologic Oncology. 2013;31(8):1725–1730. doi: 10.1016/j.urolonc.2012.05.008. [DOI] [PubMed] [Google Scholar]
  • 53.Dalpiaz O., Ehrlich G. C., Mannweiler S., et al. Validation of pretreatment neutrophil-lymphocyte ratio as a prognostic factor in a European cohort of patients with upper tract urothelial carcinoma. BJU International. 2014;114(3):334–339. doi: 10.1111/bju.12441. [DOI] [PubMed] [Google Scholar]
  • 54.Dalpiaz O., Pichler M., Mannweiler S., et al. Validation of the pretreatment derived neutrophil-lymphocyte ratio as a prognostic factor in a European cohort of patients with upper tract urothelial carcinoma. British Journal of Cancer. 2014;110(10):2531–2536. doi: 10.1038/bjc.2014.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ku J. H., Kim M., Choi W. S., Kwak C., Kim H. H. Preoperative serum albumin as a prognostic factor in patients with upper urinary tract urothelial carcinoma. International Brazilian Journal of Urology. 2014;40(6):753–762. doi: 10.1590/s1677-5538.ibju.2014.06.06. [DOI] [PubMed] [Google Scholar]
  • 56.Luo H.-L., Chen Y.-T., Chuang Y.-C., et al. Subclassification of upper urinary tract urothelial carcinoma by the neutrophil-to-lymphocyte ratio (NLR) improves prediction of oncological outcome. BJU International. 2014;113(5):E144–E149. doi: 10.1111/bju.12582. [DOI] [PubMed] [Google Scholar]
  • 57.Tanaka N., Kikuchi E., Kanao K., et al. A multi-institutional validation of the prognostic value of the neutrophil-to-lymphocyte ratio for upper tract urothelial carcinoma treated with radical nephroureterectomy. Annals of Surgical Oncology. 2014;21(12):4041–4048. doi: 10.1245/s10434-014-3830-3. [DOI] [PubMed] [Google Scholar]
  • 58.Tanaka N., Kikuchi E., Shirotake S., et al. The predictive value of C-reactive protein for prognosis in patients with upper tract urothelial carcinoma treated with radical nephroureterectomy: a multi-institutional study. European Urology. 2014;65(1):227–234. doi: 10.1016/j.eururo.2012.11.050. [DOI] [PubMed] [Google Scholar]
  • 59.Kim M., Moon K. C., Choi W. S., et al. Prognostic value of systemic inflammatory responses in patients with upper urinary tract urothelial carcinoma. World Journal of Urology. 2015;33(10):1439–1457. doi: 10.1007/s00345-015-1484-9. [DOI] [PubMed] [Google Scholar]
  • 60.Sung H. H., Gyun Jeon H., Jeong B. C., et al. Clinical significance of prognosis using the neutrophil-lymphocyte ratio and erythrocyte sedimentation rate in patients undergoing radical nephroureterectomy for upper urinary tract urothelial carcinoma. BJU International. 2015;115:587–594. doi: 10.1111/bju.12846. [DOI] [PubMed] [Google Scholar]
  • 61.Babjuk M., Burger M., Zigeuner R., et al. EAU guidelines on non-muscle-invasive urothelial carcinoma of the bladder: update 2013. European Urology. 2013;64(4):639–653. doi: 10.1016/j.eururo.2013.06.003. [DOI] [PubMed] [Google Scholar]
  • 62.Rouprêt M., Babjuk M., Compérat E., et al. European guidelines on upper tract urothelial carcinomas: 2013 update. European Urology. 2013;63(6):1059–1071. doi: 10.1016/j.eururo.2013.03.032. [DOI] [PubMed] [Google Scholar]
  • 63.Witjes J. A., Compérat E., Cowan N. C., et al. EAU guidelines on muscle-invasive and metastatic bladder cancer: summary of the 2013 guidelines. European Urology. 2014;65(4):778–792. doi: 10.1016/j.eururo.2013.11.046. [DOI] [PubMed] [Google Scholar]
  • 64.Gabay C., Kushner I. Acute-phase proteins and other systemic responses to inflammation. The New England Journal of Medicine. 1999;340(6):448–454. doi: 10.1056/nejm199902113400607. [DOI] [PubMed] [Google Scholar]
  • 65.Mahmoud F. A., Rivera N. I. The role of C-reactive protein as a prognostic indicator in advanced cancer. Current Oncology Reports. 2002;4(3):250–255. doi: 10.1007/s11912-002-0023-1. [DOI] [PubMed] [Google Scholar]
  • 66.Allin K. H., Nordestgaard B. G. Elevated C-reactive protein in the diagnosis, prognosis, and cause of cancer. Critical Reviews in Clinical Laboratory Sciences. 2011;48(4):155–170. doi: 10.3109/10408363.2011.599831. [DOI] [PubMed] [Google Scholar]
  • 67.Seebacher V., Grimm C., Reinthaller A., et al. The value of serum albumin as a novel independent marker for prognosis in patients with endometrial cancer. European Journal of Obstetrics Gynecology and Reproductive Biology. 2013;171(1):101–106. doi: 10.1016/j.ejogrb.2013.07.044. [DOI] [PubMed] [Google Scholar]
  • 68.McMillan D. C. The systemic inflammation-based Glasgow Prognostic Score: a decade of experience in patients with cancer. Cancer Treatment Reviews. 2013;39(5):534–540. doi: 10.1016/j.ctrv.2012.08.003. [DOI] [PubMed] [Google Scholar]
  • 69.Proctor M. J., Morrison D. S., Talwar D., et al. A comparison of inflammation-based prognostic scores in patients with cancer. A Glasgow Inflammation Outcome study. European Journal of Cancer. 2011;47(17):2633–2641. doi: 10.1016/j.ejca.2011.03.028. [DOI] [PubMed] [Google Scholar]
  • 70.Keizman D., Gottfried M., Ish-Shalom M., et al. Pretreatment neutrophil-to-lymphocyte ratio in metastatic castration-resistant prostate cancer patients treated with ketoconazole: association with outcome and predictive nomogram. Oncologist. 2012;17(12):1508–1514. doi: 10.1634/theoncologist.2012-0125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Keizman D., Ish-Shalom M., Huang P., et al. The association of pre-treatment neutrophil to lymphocyte ratio with response rate, progression free survival and overall survival of patients treated with sunitinib for metastatic renal cell carcinoma. European Journal of Cancer. 2012;48(2):202–208. doi: 10.1016/j.ejca.2011.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wu Y., Fu X., Zhu X., et al. Prognostic role of systemic inflammatory response in renal cell carcinoma: a systematic review and meta-analysis. Journal of Cancer Research and Clinical Oncology. 2011;137(5):887–896. doi: 10.1007/s00432-010-0951-3. [DOI] [PubMed] [Google Scholar]
  • 73.Chang S. S. Non-muscle invasive bladder cancer. Urologic Clinics of North America. 2013;40(2) doi: 10.1016/j.ucl.2013.01.011. [DOI] [PubMed] [Google Scholar]
  • 74.Seo K. W., Kim B. H., Park C. H., Kim C. I., Chang H. S. The efficacy of the EORTC scoring system and risk tables for the prediction of recurrence and progression of non-muscle-invasive bladder cancer after intravesical bacillus calmette-guerin instillation. Korean Journal of Urology. 2010;51(3):165–170. doi: 10.4111/kju.2010.51.3.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hernández V., De La Peña E., Martin M. D., Blázquez C., Diaz F. J., Llorente C. External validation and applicability of the EORTC risk tables for non-muscle-invasive bladder cancer. World Journal of Urology. 2011;29(4):409–414. doi: 10.1007/s00345-010-0635-2. [DOI] [PubMed] [Google Scholar]
  • 76.Hong S. J., Cho K. S., Han M., et al. Nomograms for prediction of disease recurrence in patients with primary Ta, T1 transitional cell carcinoma of the bladder. Journal of Korean Medical Science. 2008;23(3):428–433. doi: 10.3346/jkms.2008.23.3.428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fernandez-Gomez J., Madero R., Solsona E., et al. Predicting nonmuscle invasive bladder cancer recurrence and progression in patients treated with bacillus calmette-guerin: the CUETO scoring model. The Journal of Urology. 2009;182(5):2195–2203. doi: 10.1016/j.juro.2009.07.016. [DOI] [PubMed] [Google Scholar]
  • 78.Bassi P., Ferrante G. D., Piazza N., et al. Prognostic factors of outcome after radical cystectomy for bladder cancer: a retrospective study of a homogeneous patient cohort. The Journal of Urology. 1999;161(5):1494–1497. doi: 10.1016/s0022-5347(05)68936-7. [DOI] [PubMed] [Google Scholar]
  • 79.Honma I., Masumori N., Sato E., et al. Local recurrence after radical cystectomy for invasive bladder cancer: an analysis of predictive factors. Urology. 2004;64(4):744–748. doi: 10.1016/j.urology.2004.05.003. [DOI] [PubMed] [Google Scholar]
  • 80.Türkölmez K., Tokgöz H., Reşorlu B., Köse K., Bedük Y. Muscle-invasive bladder cancer: predictive factors and prognostic difference between primary and progressive tumors. Urology. 2007;70(3):477–481. doi: 10.1016/j.urology.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 81.International Bladder Cancer Nomogram Consortium. Postoperative nomogram predicting risk of recurrence after radical cystectomy for bladder cancer. Journal of Clinical Oncology. 2006;24(24):3967–3972. doi: 10.1200/JCO.2005.05.3884. [DOI] [PubMed] [Google Scholar]
  • 82.Karakiewicz P. I., Shariat S. F., Palapattu G. S., et al. Nomogram for predicting disease recurrence after radical cystectomy for transitional cell carcinoma of the bladder. Journal of Urology. 2006;176(4):1354–1362. doi: 10.1016/j.juro.2006.06.025. [DOI] [PubMed] [Google Scholar]
  • 83.Shariat S. F., Karakiewicz P. I., Palapattu G. S., et al. Nomograms provide improved accuracy for predicting survival after radical cystectomy. Clinical Cancer Research. 2006;12(22):6663–6676. doi: 10.1158/1078-0432.ccr-06-0372. [DOI] [PubMed] [Google Scholar]
  • 84.Svatek R. S., Shariat S. F., Novara G., et al. Discrepancy between clinical and pathological stage: external validation of the impact on prognosis in an international radical cystectomy cohort. BJU International. 2011;107(6):898–904. doi: 10.1111/j.1464-410x.2010.09628.x. [DOI] [PubMed] [Google Scholar]

Articles from Disease Markers are provided here courtesy of Wiley

RESOURCES