Ann Coloproctol Search

CLOSE


Ann Coloproctol > Volume 38(2); 2022 > Article
An, Shim, Kim, Kim, Bang, Do, Lee, and Kim: Pretreatment inflammatory markers predicting treatment outcomes in colorectal cancer

Abstract

We aimed to review whether pretreatment inflammatory markers reflect the short- and long-term outcomes of patients with colon cancer, rectal cancer, colon and rectal cancers, and metastatic colorectal cancer (CRC). We found that pretreatment complete blood count and blood chemistry tests reflect short-term and long-term oncological outcomes in patients with CRC. Specifically, in patients with colon cancer, hypoalbuminemia was associated with worse postoperative morbidity, mortality, and inferior survival. In patients with rectal cancer, elevated neutrophil-lymphocyte ratio (NLR) and thrombocytosis were associated with postoperative complications, poor overall survival (OS), and disease-free survival (DFS). A high C-reactive protein/albumin ratio (CAR) was associated with poor OS and DFS. In patients with metastatic CRC, increased NLR and platelet-lymphocyte ratio (PLR) were associated with poor OS, DFS, and progression-free survival (PFS). In addition, high CAR and a low albumin/globulin ratio on blood chemistry tests were associated with poor OS and PFS. Although universal cut-off values were not available, various types of pretreatment laboratory markers could be utilized as adjuncts to predict prognosis in patients with CRC.

INTRODUCTION

Colorectal cancer (CRC) is the third most common malignancy and one of the leading causes of cancer-related deaths worldwide [1]. Although remarkable developments in treatment modalities have improved the prognosis of patients with CRC, long-term survival remains poor, with a 5-year survival rate of approximately 60% in patients who undergo curative resection [2, 3]. Approximately 50% of patients will eventually develop metastases, with 25% of CRC patients having distant metastasis at their initial diagnosis [4, 5]. Therefore, it is important to identify factors associated with poor prognosis in patients with CRC. The best-known prognostic factor in patients with CRC is the TNM staging system proposed by the American Joint Committee on Cancer. In addition, tumor location and genetic information are factors related to prognosis [6]. However, patients with similar characteristics and the same disease stage may have different prognoses.
There have been a number of studies investigating several pretreatment laboratory markers that can help in diagnosing cancer and predicting short-term and long-term prognoses. Such pretreatment laboratory markers can be easily assayed in routine blood tests, and their prognostic value for various types of cancer, including CRC, has been confirmed in numerous studies [7-11]. Routine blood tests performed in medical institutions include complete blood counts (CBCs) and blood chemistry panel tests. Representative available parameters include hemoglobin, neutrophils, lymphocytes, platelets, and composite markers, such as the neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) in CBCs. In addition, C-reactive protein (CRP), albumin, and composite markers, such as the Glasgow Prognostic Score (GPS) and CRP/albumin ratio (CAR) can be assessed in blood chemistry tests.
These markers can reflect the inflammatory conditions of patients. Numerous studies have suggested that systemic inflammatory responses play an important role in carcinogenesis and cancer progression, and hematologic markers reflecting systemic inflammation can be used to predict the prognosis of cancer [12-14]. The purpose of this paper was to review the currently investigated pretreatment inflammatory markers and whether they can reflect the short- and long-term outcomes of patients with CRC.

INFLAMMATORY MARKERS

Blood tests

Blood is a specialized connective tissue composed of cellular components and a fluid component referred to as plasma. Cellular elements account for approximately 45% of the circulating blood volume, and plasma constitutes approximately 55%. Cells in the blood are composed of erythrocytes known as red blood cells (RBCs), granular and agranular leukocytes, also known as white blood cells (WBCs), and thrombocytes known as platelets [15].
A CBC is a laboratory test used for screening and includes measuring the numbers of WBCs, RBCs, and platelets, the concentration of hemoglobin, hematocrit, and red cell indices, such as the mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration. Blood chemistry tests include many types of chemistry panels, such as liver, renal, lipid, electrolyte, and hormonal panels. CRP and albumin levels were measured using chemistry tests (Fig. 1).

Inflammation and cancer

Chronic inflammation induces tissue damage, and cell proliferation occurs during the healing process [12, 13]. Repeated tissue damage and regeneration in chronic inflammatory situations result in permanent genetic mutations, such as point mutations, deletions, or rearrangements. Activated inflammatory cells produce chemokines and cytokines, which can influence tumor growth, migration, and differentiation by releasing growth factors, promoting angiogenesis and lymphangiogenesis, and stimulating DNA damage. Once carcinoma arises, these tumor cells turn on the expression of chemokine receptors, such as CXCR4. The production of ligands for these receptors in other organs, such as lymph nodes, bone marrow, liver, and lung, facilitates tumor migration and invasion to secondary sites. Some tumor cells, such as those in melanoma, directly regulate the expression of chemokines involved in angiogenesis, tumor growth, progression, and metastasis [16].

MARKERS AVAILABLE THROUGH A COMPLETE BLOOD COUNT

Neutrophils

Neutrophils are the most abundant WBCs in the circulation and are produced in the bone marrow daily. Neutrophils are known to play an important role in the acute inflammatory response, and in inflammatory situations, the production rate increases up to 10 times compared to normal levels. These are the first leukocytes recruited at the site of inflammation and can eradicate pathogens through several mechanisms. In addition, neutrophils are associated with numerous pathologies, including tumors and chronic inflammation [17, 18]. Neutrophils are involved in various carcinogenic processes, such as tumor initiation, growth, and proliferation, spread to other tissues, and the formation of new blood vessels in the tumor [19]. Tumor initiation can be facilitated by the release of reactive oxygen species, or proteases. Neutrophils can weaken the immune system and promote tumor growth. In addition, neutrophils can cause metastatic spread by suppressing natural killer function and promoting extravasation of tumor cells.

Lymphocytes

Lymphocytes are a subtype of WBCs. Lymphocytes develop and mature in the bone marrow and thymus, and differentiate into natural killer cells (NK cells), T-cells, and B-cells. B and T lymphocytes are involved in the adaptive immune responses, and NK cells are involved in innate immunity [20, 21]. Lymphocytes have been recognized to play a key role in combating tumor progression. A high density of lymphocytic infiltration along the invasive margin of the tumor is an independent prognostic factor for improved survival [22-24]. In contrast, immune deficiency is associated with an increased frequency of certain malignancies [25]. Immunocompromised patients, including those infected with human immunodeficiency virus and those who received transplants, were found to have a higher incidence of cancer, especially infection-related cancers [26, 27]. In addition, several studies have revealed that lymphopenia observed in advanced cancer, including CRC, correlates with unfavorable prognostic factors in terms of progression-free survival (PFS) and overall survival (OS) [28].

Platelets

Platelets are important components of the blood and are derived from megakaryocytes in the bone marrow. The primary function of platelets is to stop bleeding by adhesion and aggregation at the site of tissue and vascular injury [29]. Platelets also play important roles in host inflammation and the immune system, in general. Platelets activate leukocytes and release numerous chemoattractants and antimicrobial mediators to directly kill pathogens [30]. Platelet count is known to have an association with tumors. Tumor cells stimulate the production of thrombopoietin by secreting interleukin (IL)-6, which contributes to thrombocytosis and hypercoagulability in patients [31]. It is defined as paraneoplastic thrombocytosis, and approximately 1/3 of cancer patients show thrombocytosis at the time of diagnosis [32]. Increased and activated platelets release growth factors that facilitate tumor growth and invasion. Platelets also contribute to tumor metastasis by facilitating cancer cell adhesion and extravasation [31, 33]. Numerous studies have shown that thrombocytosis is associated with long-term prognosis in patients [34-37]. In a multicenter study of patients with advanced rectal cancer who received neoadjuvant chemoradiation (CRT), Belluco et al. [35] demonstrated that thrombocytosis before neoadjuvant CRT was associated with worse oncologic outcome. They set the platelet count cut-off value of 300×109/L by receiver operating characteristics (ROC) curve and showed thrombocytosis was associated with poor 5-year OS, disease-free survival (DFS), and higher local and distant metastasis. In addition, thrombocytosis was confirmed as an independent prognostic factor for pathologic complete response in the study.

Neutrophil-lymphocyte ratio

The literature shows that many studies have combined these inflammatory markers to predict the tumor prognosis. The NLR is a widely used combination marker, and NLR is defined as the absolute neutrophil count divided by the absolute lymphocyte count. High NLR, that is, increased neutrophil levels and decreased lymphocyte levels, have been suggested as poor prognostic markers in CRC [38-40]. Several studies have shown that a high preoperative NLR is a useful predictive marker for short-term outcomes in patients with CRC [37, 41, 42]. A study by Xia et al. [37] analyzed the correlation between various inflammatory markers and postoperative morbidity; and among them, it was found that postoperative morbidity significantly increased when the NLR was 2.8 or higher. In a study by Gohil et al. [41], in which the preoperative NLR value of 4.3 was set as the cut-off value, patients with elevated NLR are more likely to have a prolonged hospital stay after operation. In addition, one study analyzed the relationship between preoperative NLR and the amount of ileostomy after colorectal surgery. In the study, high-output ileostomy was defined when the stoma output was more than 2,000 mL/day for 3 or more consecutive days, and the optimal cut-off value of preoperative NLR was set 3.0 by ROC curve analysis. The study revealed that a high preoperative NLR is a predictor of high-output ileostomy after colorectal surgery [43]. In addition to short-term outcomes, many studies have also shown that the NLR is a good predictor of long-term oncological outcomes [44, 45]. A study by Chiang et al. [44] demonstrated that the preoperative elevated NLR (> 3) was associated with worse 5-year DFS, and another study also revealed that the preoperative elevated NLR (> 3) could reflect poor 5-year DFS and cancer-specific survival (CSS) [45]. In several studies of patients with advanced rectal cancer who received neoadjuvant CRT, the NLR was found to be a useful marker for predicting pathological tumor regression or pathologic complete response [46, 47]. One study, which set the optimal cut-off NLR value as 3, showed that elevated NLR before CRT was significant predictor for poor pathologic response [46]. However, another study by Jeon et al. [47] suggested that the elevated NLR (> 3.23) after CRT was an independent negative predictive factor for pathologic complete response, and they also showed that elevated NLR was an independent poor prognostic factor for recurrence-free survival (RFS). In studies involving patients with resectable CRC liver metastasis, pretreatment NLR has been demonstrated to be a good predictor of long-term prognosis in terms of 5-year OS and DFS [48, 49]. Moreover, several studies have reported that pretreatment NLR could reflect the effect of palliative chemotherapy in patients with unresectable metastatic CRC [50-52] (Table 1).

Platelet-lymphocyte ratio

The PLR, defined as the ratio of platelet count to lymphocyte count, has also been suggested as a prognostic marker for CRC. Elevated PLR, that is, increased platelet count and decreased lymphocyte count, have been suggested to be poor prognostic factors in CRC [53, 54]. A retrospective study with T1 to 2 rectal cancer patients by Xia et al. [37] demonstrated that a pretreatment-elevated PLR (> 140) was correlated with a higher postoperative morbidity rate. However, the association between PLR and longterm outcomes was not demonstrated in their study. In contrast, another study by Ozawa et al. [54] set the optimal cut-off value of preoperative PLR as 25.4 by ROC curve analysis and revealed that pretreatment PLR is an independent prognostic factor for long-term oncological outcomes in terms of 5-year DFS and CSS in stage II patients with CRC. Huang et al. [55] performed a meta-analysis including 17 studies with CRC patients and reported that elevated PLR was reported to be associated with poor OS, DFS, CSS, and RFS. This systematic review also concluded that elevated PLR was associated with poor clinicopathological characteristics, such as high tumor stage and poor differentiation. In addition, in a study of patients who underwent liver resection for CRC liver metastasis, preoperative PLR was an independent prognostic factor for 5-year DFS and OS. The study determined the optimal cut-off value of preoperative PLR and NLR as 150 and 2.4 by ROC curve analysis, and this study suggested that PLR is a better predictor of long-term oncologic outcomes than NLR through multivariate analysis [56] (Table 2).

MARKERS AVAILABLE THROUGH BLOOD CHEMISTRY TESTS

Albumin

Albumin is a protein that is produced from the liver and accounts for approximately 60% of the serum protein, making up the largest proportion. It is a marker that reflects nutritional condition, and its production is regulated by osmotic colloid pressure, inflammatory state, and nutritional status. Systemic inflammation reduces albumin concentration, regardless of malnutrition [57]. Malignant neoplasms exert a great deal of stress on the human body via tissue hypoxia and necrosis, causing tissue damage. To respond to these stressful changes, the human body produces various types of proinflammatory cytokines, such as IL-6, IL-1, tumor necrosis factor alpha (TNF-α), and growth factors. Consequently, hepatocytes increase the production of acute-phase proteins, such as CRP, and decrease the production of albumin [58-60]. In addition, because patients with advanced cancer frequently present with malnutrition, there has been much interest in the relationship between hypoalbuminemia and disease status or prognosis. Many studies have suggested that preoperative hypoalbuminemia is associated with poor short-term outcomes after colorectal surgery [41, 61, 62]. Chiang et al. [61] investigated the linear relationship between preoperative albumin levels and postoperative morbidity and mortality in patients with CRC. The study reported that every 0.1-g/dL increase in albumin reduced morbidity and mortality by 7.3% and 15.6%, respectively. In another study investigating serum albumin concentrations before and after surgery for patients with CRC, it was reported that the greater the difference between albumin concentrations before and after surgery, the greater the risk of postoperative complications [63]. In addition to short-term prognosis, a number of studies have demonstrated that pretreatment albumin levels can reflect long-term oncological outcomes in patients with CRC [64-66]. In a retrospective study of 3,849 CRC patients, the 5-year OS and RFS were significantly lower in the hypoalbuminemia group with initial serum albumin of < 35 g/L than in the normal group, and postoperative morbidity was also higher in the hypoalbuminemia group [65]. Similarly, a study of CRC patients who underwent surgical treatment also found that preoperative hypoalbuminemia (< 35 g/L) was an independent poor prognostic factor of CSS [66].

C-reactive protein

CRP is a nonspecific and sensitive marker of inflammation and is produced in the liver [67]. Its synthesis is induced by proinflammatory cytokines, such as IL-6, IL-1, and TNF-α. CRP is an index that reflects the course of the inflammatory response because its value changes immediately in response to the inflammatory response. Increased CRP levels in cancer patients can predict disease severity, risk of complications, risk of tumor recurrence, and decreased survival rates [68]. According to a systematic review by Shrotriya et al. [69], increased CRP levels before surgery reflected poor prognosis in terms of short-term outcomes and oncological outcomes in patients with solid tumors in most of the studies (90%). Many studies have found that pretreatment-elevated CRP levels are associated with poor prognosis in patients with CRC patients [70-72]. Koike et al. [70] investigated the relationship between preoperative CRP level and long-term survival in patients with CRC, and the optimal cut-off value for elevated CRP was set at 0.5 mg/dL. Through multivariate analysis, the study demonstrated that CRP was an independent prognostic factor for CSS in stage I or II patients. In addition, Artaç et al. [52] analyzed whether several inflammatory markers could predict the effect of chemotherapy in metastatic CRC patients receiving chemotherapy using FOLFIRI (folinic acid, fluorouracil, and irinotecan) plus bevacizumab. In the study, CRP was measured on the day of initiation of chemotherapy, and PFS was significantly lower in the patient group above the upper limit of normal CRP level.

Glasgow Prognostic Score

GPS was introduced to enhance the effect as a prognostic predictor and is known to be a useful scoring system for predicting the prognosis of patients with CRC as well as other malignant tumors [73, 74]. GPS was based on the combination of hypoalbuminemia (< 35 g/L) and elevated CRP (> 10 mg/L); if both were abnormal, the score was 2; if one or the other was abnormal, the score was 1; if neither was abnormal, the score was 0. However, because there have been claims that hypoalbuminemia alone without elevated CRP was rare and was not associated with poor survival, GPS was modified (modified GPS [mGPS]) [74]. The mGPS was as follows: patients with CRP of ≤ 10 mg/L and albumin of ≥ 35 g/L were scored 0, those with CRP of > 10 mg/L were scored 1, and those with CRP of > 10 mg/L and albumin of < 35 g/L were scored 2. In a large retrospective study by Park et al. [75], a high score of mGPS in patients with CRC was associated with old age, emergency setting, advanced tumor stage, poor differentiation, surgical margin involvement, peritoneal involvement, and tumor perforation. The study also demonstrated that a high score of mGPS was independently associated with long-term oncologic outcomes in terms of 5-year CSS and OS. A study by Moyes et al. [76] demonstrated that preoperative mGPS was independently associated with the risk of postoperative infectious complications in patients with CRC. Besides, several studies have shown that pretreatment with high GPS or mGPS is associated with poor survival in patients with CRC [74, 75, 77]. A study by McMillan et al. [74] analyzed the relationship between mGPS and long-term survival in CRC patients who received curative resection. The study revealed that a high mGPS score is an independent prognostic factor for poor 3-year OS and CSS. In addition, a meta-analysis of 9,389 CRC patients included in 41 studies concluded that pretreatment high GPS or mGPS can reflect poor OS and CSS in CRC patients who underwent surgical resection or chemotherapy [73].

C-reactive protein/albumin ratio

Among the markers using albumin and CRP, unlike GPS and mGPS, which are classified as absolute values, CAR is a useful index indicating the ratio of albumin and CRP. Several previous studies have suggested that the CAR value can reflect the postoperative short-term outcomes in various kinds of diseases [78, 79]. There are also papers analyzing the correlation between postoperative morbidity and preoperative CAR in CRC patients. A study by Yu et al. [80] demonstrated that the preoperative CAR higher than 2.44 was an independent risk factor for anastomotic leakage. Another study by Ge et al. [81] has suggested that postoperative high CAR greater than 2.2 can be also used to predict worse postoperative outcomes in patients undergoing colorectal surgery. In addition, a study has reported that pretreatment CAR can even predict the side effects of adjuvant chemotherapy in patients with stage III CRC [82]. As well as short-term outcomes, there have also been many studies of CAR as a predictor of long-term oncological outcomes in patients with CRC [77, 83, 84]. A study by Shibutani et al. [85] demonstrated that preoperative high CAR greater than 0.0271 was independently associated with poor CSS. The study also suggested that CAR may be superior to mGPS for predicting survival. A study conducted in unresectable metastatic patients with CRC who received chemotherapy set the optimal cut-off value of pretreatment CAR to 0.183 and reported that pretreatment CAR is a useful marker for predicting OS and effectiveness of chemotherapy. The study also suggested that CAR had a better predictive value than mGPS [50]. These studies said that although mGPS is a simple and useful prognostic scoring system, it has a weak point of not reflecting the linear change. In addition, a meta-analysis with 2,492 CRC patients who received curative resection or chemotherapy in 9 retrospective studies concluded that pretreatment high CAR is a powerful prognostic indicator predicting poor OS and DFS [86-90] (Table 3).

SUMMARY AND CONCLUSION

In summary, we have reviewed that currently-available pretreatment laboratory markers could be utilized as an adjunct to predict prognosis in patients with CRC (Table 4). In literature, study subjects were colon cancer only, rectal cancer only, colon and rectal cancer together, and metastatic CRC.
In patients with colon cancer, pretreatment blood chemistry reflects short-term and long-term oncological outcomes. Hypoalbuminemia correlated with inferior OS and RFS as well as worse postoperative morbidity and mortality. In patients with rectal cancer, pretreatment CBC can predict short-term and long-term outcomes, and blood chemistry can reflect long-term oncologic outcomes. Elevated NLR and thrombocytosis correlated with higher postoperative complication rates and poor OS and DFS. In addition, high CAR was associated with poor OS and DFS.
According to studies enrolling both colon and rectal cancer patients, pretreatment CBC and chemistry tests can also reflect short- and long-term oncologic outcomes. Elevated NLR was correlated with increased length of hospital stay, poor CSS and RFS, while thrombocytosis was related to poor CSS and DFS. Hypoalbuminemia was associated with an increased hospital stay, increased postoperative 30-day mortality, and poor OS and DFS. Preoperative high mGPS predicted increased postoperative infectious complications and worse OS and DFS. Moreover, a high CAR was associated with a poor OS.
In patients with metastatic CRC, pretreatment CBC and chemistry tests can also predict long-term oncologic outcomes. Increased NLR and PLR were associated with poor OS, DFS, and PFS. High CAR and low albumin/globulin ratio on blood chemistry tests were related to poor OS and PFS.
In conclusion, a higher inflammatory status reflected by pretreatment CBC and chemistry tests can predict poor surgical and oncological outcomes in patients with either nonmetastatic or metastatic CRC. However, to be incorporated in clinical practice as a helpful adjunct, the optimal cut-off values of these laboratory markers need to be defined in future clinical studies.

Notes

CONFLICT OF INTEREST

No potential conflict of interest relevant to this article was reported.

ACKNOWLEDGMENTS

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF 2017R1D1A3-B03032301).

Fig. 1.
Preoperative inflammatory markers.
ac-2021-01004-0143f1.jpg
Table 1.
Neutrophil related markers from a complete blood count
Marker Year Main outcome Study N Design, center Surgical disease TNM stage Cut-off Therapy Statistics and comments
NLR 2020 Postoperative complications OS DFS Xia et al. [37] 154 Retrospective, single center RC T1–2 2.8 Resection NLR was associated with postoperative CTx.
NLR was an independent prognostic factor for DFS (HR, 6.656, P = 0.009).
NLR 2019 Ileostomy amount: 2,000 mL/day Hiramoto et al. [43] 45 Retrospective, single center CRC 1–4 3.0 Resection Preoperative NLR was significantly higher in the high-output ileostomy group (P = 0.004).
NLR 2019 OS DFS Kim et al. [48] 83 Retrospective, 2 centers CRLM 4 1.94 Resection, CTx HR, 2.23; independent prognostic factor
NLR 2019 pCR OS Mao et al. [49] 183 Retrospective, single center CRLM 4 2.3 Preoperative CTx, resection OR, 2.40; P = 0.015
RFS HR, 2.43; P < 0.001
HR, 1.53; P = 0.017
NLR 2019 OS PFS Casadei-Gardini et al. [51] 276 RCT, multi-center mCRC 4 3 CTx (± bevacizumab) HR, 2.38
HR, 2.27
NLR + LDH is a good prognostic marker for mCRC with 1st-line CTx.
NLR 2016 OS Ishizuka et al. [77] 627 Retrospective, single center CRC 1–4 2.9 Resection HR, 1.811; P = 0.003
NLR 2016 OS Shibutani et al. [50] 99 Retrospective, single center Unresectable mCRC 4 3 Palliative CTx HR, 2.706; P = 0.001
NLR 2016 Tumor regression grade Caputo et al. [42] 151 Retrospective, single center RC 2, 3 2.8 nCRT followed by total mesorectal excision Preoperative high NLR was associated with overall postoperative complications.
Postoperative complications
NLR 2014 pCR Kim et al. [46] 102 Retrospective, single center CRC 1–4 3 Preoperative CRT, resection HR, 5.2; P = 0.04
CSS HR, 6.6; P = 0.02
RFS HR, 2.8; P = 0.03
NLR 2014 OS Neofytou et al. [56] 140 Retrospective, single center CRLM 4 2.4 Resection NLR was not an independent prognostic factor for OS or DFS.
DFS
NLR 2014 Hospital LOS Gohil et al. [41] 196 Retrospective, single center CRC 1–4 4.3 Resection Preoperative NLR was associated with increased LOS (OR, 1.702).

NLR, neutrophil-lymphocyte ratio; OS, overall survival; DFS, disease-free survival; RC, rectal cancer; CTx, chemotherapy; HR, hazard ratio; CRC, colorectal cancer; CRLM, colorectal cancer with liver metastasis; pCR, pathologic complete response; RFS, relapse-free survival; OR, odds ratio; PFS, progression-free survival; RCT, randomized controlled trial; mCRC, metastatic colorectal cancer; LDH, lactate dehydrogenase; FOLFIRI, folinic acid, fluorouracil, and irinotecan; nCRT, neoadjuvant chemoradiotherapy; CSS, cancer-specific survival; LOS, length of stay.

Table 2.
Platelet related markers from a complete blood count
Marker Year Main outcome Author N Design, center Surgical disease TNM stage Cut-off Therapy Statistics and comments
PLT 2018 pCR Belluco et al. [35] 965 Retrospective, multicenter RC with nCRT 2, 3 300K nCRT followed by TME OR, 0.51; P=0.001
OS HR, 1.45; P=0.009
DFS HR, 1.39; P=0.009
PLT 2012 OS Ishizuka et al. [36] 453 Retrospective, single center CRC 1–4 300K Resection OR, 1.642; P=0.039
Thrombocytosis is associated with OS.
PLR 2020 Postoperative complication Xia et al. [37] 154 Retrospective, single center RC T1–2 140.0 Resection PLR was associated with postoperative complications. However, PLR was not an independent prognostic factor for OS, DFS.
OS
DFS
PLR 2015 CSS Ozawa et al. [54] 234 Retrospective, single center CRC 2 25.4 Resection HR, 3.61; P=0.038
DFS HR, 2.65; P=0.011
PLR 2014 OS Neofytou et al. [56] 140 Retrospective, single center CRLM 4 15 Resection HR, 2.17; P=0.027
DFS HR, 1.68; P=0.034

PLT, platelet; OS, overall survival; DFS, disease-free survival; RC, rectal cancer; pCR, pathologic complete response; nCRT, neoadjuvant chemoradiotherapy; TME, total mesorectal excision; OR, odds ratio; HR, hazard ratio; CSS, cancer-specific survival; CRC, colorectal cancer; PLR, platelet-to-lymphocyte ratio; CRLM, colorectal cancer with liver metastasis.

Table 3.
Markers available through blood chemistry tests
Marker Year Main outcome Author N Design, center Surgical disease TNM stage Cut-off Therapy Statistics and comments
Alb 2019 OS Yamamoto et al. [64] 523 Retrospective, single center CRC 1–4 3.85 Resection Kaplan-Meier test, P < 0.0001
DFS
ΔAlba 2018 Postoperative complication Wang et al. [63] 193 Retrospective, single center CRC 1–3 17.3% Resection OR, 1.66/+
Alb 2017 Postoperative morbidity, mortality Chiang et al. [61] 3,732 Retrospective, single center CC 1–3 3.5 Resection OR, 0.927
OR, 0.844
Morbidity and mortality were inversely related to Alb.
Alb 2015 30-Day mortality Montomoli et al. [62] 9,339 Prospective, population-based cohort CRC 1–4 2.5 Resection HR, 7.59
2.6–3.0 HR, 5.19
3.1–3.5 HR, 2.58
3.6–4.0 4.0 HR, 1.75
HR, 1.00
Preoperative Alb was inversely associated with 30-day mortality.
Alb 2014 Hospital LOS Gohil et al. [41] 196 Retrospective, single center CRC 1–4 34.5 g/dL Resection OR, 0.47/+
Alb 2011 Short-term outcomes Lai et al. [65] 3,849 Retrospective, single center CC 1–3 3.5 Resection OR, 2.15; P < 0.001
OS HR, 1.75; P < 0.001
RFS HR, 1.28; P = 0.020
Alb 2009 OS Sun et al. [66] 1,367 Retrospective, single center CRC 1–4 3.5 Resection HR, 1.45; P = 0.011
CSS Alb was associated only with CSS.
CRP 2017 PFS Artaç et al. [52] 90 Retrospective, 2 centers mCRC 4 ULN Bevacizumab + FOLFIRI Only CRP was confirmed to be associated with PFS in the multivariate analysis.
CRP 2008 CSS Koike et al. [70] 300 Retrospective, single center CRC 1, 2 0.5 mg/dL Resection HR, 1.88/+
GPS 2016 OS Ishizuka et al. [77] 627 Retrospective, single center CRC 1–4 0, 1, 2 Resection HR, 1.809; P = 0.006
mGPS 2009 Postoperative infection Moyes et al. [76] 455 Retrospective, single center CRC 1–3 0, 1, 2 Resection HR, 1.76; P = 0.003
mGPS 2007 OS (3 yr) McMillan et al. [74] 316 Retrospective, single center CRC 1–3 0, 1, 2 Resection Univariate analysis/+
CSS (3 yr)
CAR 2017 Postoperative complication Ge et al. [81] 359 Retrospective, single center CRC 1–3 0.022 Resection OR, 4.413; P < 0.001
CAR 2017 OS Haruki et al. [84] 112 Retrospective, single center CRLM 4 0.04 Resection HR, 2.559; P = 0.021
DFS HR, 1.731; P = 0.002
CAR 2017 OS DFS Ide et al. [83] 115 Retrospective, single center RC with nCRT 1–3 0.049 nCRT followed by TME HR, 5.09; P < 0.01
HR, 4.98; P < 0.01
CAR 2016 RFS Shibutani et al. [85] 705 Retrospective, single center CRC 1–3 0.0271 Resection HR, 1.503/+
CSS HR, 1.672/+
CAR 2016 OS Ishizuka et al. [77] 627 Retrospective, single center CRC 1–4 0.038 Resection HR, 2.613; P < 0.001
CAR 2016 OS Shibutani et al. [50] 99 Retrospective, single center Unresectable mCRC 4 0.183 Palliative CTx HR, 1.866; P = 0.031
CAR 2016 Side effects (G3 or G4) Tominaga et al. [82] 136 Retrospective, single center CRC 3 0.1 Adjuvant CTx HR, 7.06; P < 0.001
GAR 2018 OS Hachiya et al. [87] 941 Retrospective, single center CRC 1–4 0.83 Resection HR, 1.979; P=0.001
NLR was also associated with OS.
AGR 2015 OS Shibutani et al. [88] 66 Retrospective, single center Unresectable mCRC 4 1.25 CTx HR, 2.247
PFS HR, 2.662
The high-AGR group showed superior chemotherapeutic response
AGR 2013 OS Azab et al. [89] 534 Retrospective, single center CRC 1–4 < 1.029 Resection, CTx High AGR was associated with improved survival (HR, 0.25).
1.028–1.321
> 1.321
LCR 2020 Postoperative complication (SSI) Okugawa et al. [90] 477 Retrospective, 2 centers CRC 1–4 6,000 Resection Low LCR was associated with postoperative complication and survival.
OS
DFS

Alb, albumin; OS, overall survival; DFS, disease-free survival; CRC, colorectal cancer; OR, odds ratio; CC, colon cancer; HR, hazard ratio; LOS, length of stay; RFS, relapse-free survival; CSS, cancer-specific survival; PFS, progression-free survival; mCRC, metastatic CRC; ULN, upper limit of normal; FOLFIRI, folinic acid, fluorouracil and irinotecan; CRP, C-reactive protein; GPS, Glasgow Prognostic Score; mGPS, modified GPS; CAR, CRP-albumin ratio; CRLM, colorectal cancer with liver metastasis; RC, rectal cancer; nCRT, neoadjuvant chemoradiotherapy; TME, total mesorectal excision; CTx, chemotherapy; GAR, globulin-albumin ration; AGR, albumin-globulin ration; SSI, surgical site infection; LCR, lymphocyte-CRP ratio.

a (Preoperative Alb−Alb at postoperative day 2)/preoperative Alb.

Table 4.
Association with inflammatory markers for each disease group
Disease and serologic test Short-term outcome Overall survival Disease-freesurvival Progression-free survival
Colon cancer
 CBC
 Chemistry Alb [61, 65] Alb [65] Alb [65]
Rectal cancer
 CBC NLR, PLR [37] PLT [35] NLR [37]
NLR [42] PLT [35]
 Chemistry CAR [83] CAR [83]
Colorectal cancer
 CBC NLR [41] PLT [36], NLR [46] NLR [46]
CAR [77]
 Chemistry Alb, NLR [41] Alb [64] Alb [64]
Alb [62, 63] GPS [74] LCR [90]
mGPS [76] CAR [77]
LCR [90] GAR [87]
LCR [90]
Metastatic colorectal cancer
 CBC NLR [48-51] NLR [48, 49] NLR [51, 52]
PLR [56] PLR [56]
 Chemistry CAR [50] CAR [84] CRP [52]
CAR [84] AGR [88]
AGR [88]

CBC, complete blood count; Alb, albumin; NLR, neutrophil-lymphocyte ratio; PLR, platelet-lymphocyte ratio; PLT, platelet; CAR, CRP-albumin ratio; mGPS, modified GPS; GPS, Glasgow Prognostic Score; GAR, globulin-albumin ration; LCR, lymphocyte-CRP ratio; AGR, albumin-globulin ration; CRP, C-reactive protein.

REFERENCES

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394–424.
crossref pmid
2. Park S, Kang J, Park EJ, Baik SH, Lee KY. Laparoscopic and robotic surgeries for patients with colorectal cancer who have had a previous abdominal surgery. Ann Coloproctol 2017;33:184–91.
crossref pmid pmc
3. Park IJ, Lee JL, Yoon YS, Kim CW, Lim SB, Yu CS, et al. Oncologic outcomes of organ preserving approaches in patients with rectal cancer treated with preoperative chemoradiotherapy. Ann Coloproctol 2019;35:65–71.
crossref pmid pmc
4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30.
crossref pmid
5. Kim YW, Kim IY. The role of surgery for asymptomatic primary tumors in unresectable stage IV colorectal cancer. Ann Coloproctol 2013;29:44–54.
crossref pmid pmc
6. Kim K, Castro EJT, Shim H, Advincula JVG, Kim YW. Differences regarding the molecular features and gut microbiota between right and left colon cancer. Ann Coloproctol 2018;34:280–5.
crossref pmid pmc
7. Dolan RD, Laird BJ, Horgan PG, McMillan DC. The prognostic value of the systemic inflammatory response in randomised clinical trials in cancer: a systematic review. Crit Rev Oncol Hematol 2018;132:130–7.
crossref pmid
8. Watt DG, Ramanathan ML, McSorley ST, Walley K, Park JH, Horgan PG, et al. Clinicopathological determinants of an elevated systemic inflammatory response following elective potentially curative resection for colorectal cancer. Ann Surg Oncol 2017;24:2588–94.
crossref pmid pmc
9. Stojkovic Lalosevic M, Pavlovic Markovic A, Stankovic S, Stojkovic M, Dimitrijevic I, Radoman Vujacic I, et al. Combined diagnostic efficacy of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and mean platelet volume (MPV) as biomarkers of systemic inflammation in the diagnosis of colorectal cancer. Dis Markers 2019;2019:6036979.
crossref pmid pmc
10. Park JH, McMillan DC, Horgan PG, Roxburgh CS. The impact of anti-inflammatory agents on the outcome of patients with colorectal cancer. Cancer Treat Rev 2014;40:68–77.
crossref pmid
11. Dolan RD, McSorley ST, Park JH, Watt DG, Roxburgh CS, Horgan PG, et al. The prognostic value of systemic inflammation in patients undergoing surgery for colon cancer: comparison of composite ratios and cumulative scores. Br J Cancer 2018;119:40–51.
crossref pmid pmc
12. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420:860–7.
crossref pmid pmc
13. Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol 2014;15:e493–503.
crossref pmid
14. Lee YN, Lee JL, Yu CS, Kim JB, Lim SB, Park IJ, et al. Clinicopathological characteristics and surgical outcomes of Crohn disease-associated colorectal malignancy. Ann Coloproctol 2021;37:101–8.
crossref pmid pmc
15. Ovalle WK, Nahirney PC. Blood and bone marrow. In: Ovalle WK. editor. Netter’s essential histology: with student consult access. 2nd ed. Philadelphia: Elsevier Health Sciences; 2013. p.157–72.

16. Owen JD, Strieter R, Burdick M, Haghnegahdar H, Nanney L, Shattuck-Brandt R, et al. Enhanced tumor-forming capacity for immortalized melanocytes expressing melanoma growth stimulatory activity/growth-regulated cytokine beta and gamma proteins. Int J Cancer 1997;73:94–103.
crossref pmid
17. Liew PX, Kubes P. The neutrophil’s role during health and disease. Physiol Rev 2019;99:1223–48.
crossref pmid
18. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 2013;13:159–75.
crossref pmid
19. Ocana A, Nieto-Jiménez C, Pandiella A, Templeton AJ. Neutrophils in cancer: prognostic role and therapeutic strategies. Mol Cancer 2017;16:137.
crossref pmid pmc
20. Olsen Saraiva Camara N, Lepique AP, Basso AS. Lymphocyte differentiation and effector functions. Clin Dev Immunol 2012;2012:510603.
crossref pmid pmc
21. Sakai Y, Kobayashi M. Lymphocyte ‘homing’ and chronic inflammation. Pathol Int 2015;65:344–54.
crossref pmid
22. Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012;12:298–306.
crossref pmid
23. Ohtani H. Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human colorectal cancer. Cancer Immun 2007;7:4.
pmid pmc
24. Mei Z, Liu Y, Liu C, Cui A, Liang Z, Wang G, et al. Tumour-infiltrating inflammation and prognosis in colorectal cancer: systematic review and meta-analysis. Br J Cancer 2014;110:1595–605.
crossref pmid pmc
25. Ménétrier-Caux C, Ray-Coquard I, Blay JY, Caux C. Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer 2019;7:85.
crossref pmid pmc
26. Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet 2007;370:59–67.
crossref pmid
27. Ettorre GM, Piselli P, Galatioto L, Rendina M, Nudo F, Sforza D, et al. De novo malignancies following liver transplantation: results from a multicentric study in central and southern Italy, 1990-2008. Transplant Proc 2013;45:2729–32.
crossref pmid
28. Cézé N, Thibault G, Goujon G, Viguier J, Watier H, Dorval E, et al. Pre-treatment lymphopenia as a prognostic biomarker in colorectal cancer patients receiving chemotherapy. Cancer Chemother Pharmacol 2011;68:1305–13.
crossref pmid
29. Ruggeri ZM, Mendolicchio GL. Adhesion mechanisms in platelet function. Circ Res 2007;100:1673–85.
crossref pmid
30. Jenne CN, Kubes P. Platelets in inflammation and infection. Platelets 2015;26:286–92.
crossref pmid
31. Haemmerle M, Stone RL, Menter DG, Afshar-Kharghan V, Sood AK. The platelet lifeline to cancer: challenges and opportunities. Cancer Cell 2018;33:965–83.
crossref pmid pmc
32. Lin RJ, Afshar-Kharghan V, Schafer AI. Paraneoplastic thrombocytosis: the secrets of tumor self-promotion. Blood 2014;124:184–7.
crossref pmid pmc
33. Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer 2011;11:123–34.
crossref pmid pmc
34. Gu D, Szallasi A. Thrombocytosis portends adverse prognosis in colorectal cancer: a meta-analysis of 5,619 patients in 16 individual studies. Anticancer Res 2017;37:4717–26.
pmid
35. Belluco C, Forlin M, Delrio P, Rega D, Degiuli M, Sofia S, et al. Elevated platelet count is a negative predictive and prognostic marker in locally advanced rectal cancer undergoing neoadjuvant chemoradiation: a retrospective multi-institutional study on 965 patients. BMC Cancer 2018;18:1094.
crossref pmid pmc
36. Ishizuka M, Nagata H, Takagi K, Iwasaki Y, Kubota K. Preoperative thrombocytosis is associated with survival after surgery for colorectal cancer. J Surg Oncol 2012;106:887–91.
crossref pmid
37. Xia LJ, Li W, Zhai JC, Yan CW, Chen JB, Yang H. Significance of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, lymphocyte-to-monocyte ratio and prognostic nutritional index for predicting clinical outcomes in T1-2 rectal cancer. BMC Cancer 2020;20:208.
crossref pmid pmc
38. Hung HY, Chen JS, Yeh CY, Changchien CR, Tang R, Hsieh PS, et al. Effect of preoperative neutrophil-lymphocyte ratio on the surgical outcomes of stage II colon cancer patients who do not receive adjuvant chemotherapy. Int J Colorectal Dis 2011;26:1059–65.
crossref pmid
39. Carruthers R, Tho LM, Brown J, Kakumanu S, McCartney E, McDonald AC. Systemic inflammatory response is a predictor of outcome in patients undergoing preoperative chemoradiation for locally advanced rectal cancer. Colorectal Dis 2012;14:e701–7.
crossref pmid
40. Guo J, Chok AY, Lim HJ, Tay WX, Lye WK, Samarakoon LB, et al. Prognostic value of neutrophil-to-lymphocyte ratio in obstructing colorectal cancer treated by endoscopic stenting as a bridge to surgery. Ann Coloproctol 2021;37:159–65.
crossref pmid pmc
41. Gohil R, Rishi M, Tan BH. Pre-operative serum albumin and neutrophil-lymphocyte ratio are associated with prolonged hospital stay following colorectal cancer surgery. Br J Med Med Res 2014;4:481–7.
crossref pmid pmc
42. Caputo D, Caricato M, Coppola A, La Vaccara V, Fiore M, Coppola R. Neutrophil to lymphocyte ratio (NLR) and derived neutrophil to lymphocyte ratio (d-NLR) predict non-responders and postoperative complications in patients undergoing radical surgery after neo-adjuvant radio-chemotherapy for rectal adenocarcinoma. Cancer Invest 2016;34:440–51.
crossref pmid
43. Hiramoto Y, Kawahara H, Matsumoto T, Takeda M, Misawa T, Yanaga K. Preoperative neutrophil-lymphocyte ratio is a predictor of high-output ileostomy after colorectal surgery. Anticancer Res 2019;39:3265–8.
crossref pmid
44. Chiang SF, Hung HY, Tang R, Changchien CR, Chen JS, You YT, et al. Can neutrophil-to-lymphocyte ratio predict the survival of colorectal cancer patients who have received curative surgery electively? Int J Colorectal Dis 2012;27:1347–57.
crossref pmid
45. Shin JS, Suh KW, Oh SY. Preoperative neutrophil to lymphocyte ratio predicts survival in patients with T1-2N0 colorectal cancer. J Surg Oncol 2015;112:654–7.
crossref pmid
46. Kim IY, You SH, Kim YW. Neutrophil-lymphocyte ratio predicts pathologic tumor response and survival after preoperative chemoradiation for rectal cancer. BMC Surg 2014;14:94.
crossref pmid pmc
47. Jeon BH, Shin US, Moon SM, Choi JI, Kim MS, Kim KH, et al. Neutrophil to lymphocyte ratio: a predictive marker for treatment outcomes in patients with rectal cancer who underwent neoadjuvant chemoradiation followed by surgery. Ann Coloproctol 2019;35:100–6.
crossref pmid pmc
48. Kim H, Jung HI, Kwon SH, Bae SH, Kim HC, Baek MJ, et al. Preoperative neutrophil-lymphocyte ratio and CEA is associated with poor prognosis in patients with synchronous colorectal cancer liver metastasis. Ann Surg Treat Res 2019;96:191–200.
crossref pmid pmc
49. Mao R, Zhao JJ, Bi XY, Zhang YF, Li ZY, Huang Z, et al. A low neutrophil to lymphocyte ratio before preoperative chemotherapy predicts good outcomes after the resection of colorectal liver metastases. J Gastrointest Surg 2019;23:563–70.
crossref pmid
50. Shibutani M, Maeda K, Nagahara H, Iseki Y, Hirakawa K, Ohira M. The significance of the C-reactive protein to albumin ratio as a marker for predicting survival and monitoring chemotherapeutic effectiveness in patients with unresectable metastatic colorectal cancer. Springerplus 2016;5:1798.
crossref pmid pmc
51. Casadei-Gardini A, Scarpi E, Ulivi P, Palladino MA, Accettura C, Bernardini I, et al. Prognostic role of a new inflammatory index with neutrophil-to-lymphocyte ratio and lactate dehydrogenase (CII: Colon Inflammatory Index) in patients with metastatic colorectal cancer: results from the randomized Italian Trial in Advanced Colorectal Cancer (ITACa) study. Cancer Manag Res 2019;11:4357–69.
pmid pmc
52. Artaç M, Uysal M, Karaağaç M, Korkmaz L, Er Z, Güler T, et al. Prognostic impact of neutrophil/lymphocyte ratio, platelet count, CRP, and albumin levels in metastatic colorectal cancer patients treated with FOLFIRI-bevacizumab. J Gastrointest Cancer 2017;48:176–80.
crossref pmid
53. Jia J, Zheng X, Chen Y, Wang L, Lin L, Ye X, et al. Stage-dependent changes of preoperative neutrophil to lymphocyte ratio and platelet to lymphocyte ratio in colorectal cancer. Tumour Biol 2015;36:9319–25.
crossref pmid
54. Ozawa T, Ishihara S, Nishikawa T, Tanaka T, Tanaka J, Kiyomatsu T, et al. The preoperative platelet to lymphocyte ratio is a prognostic marker in patients with stage II colorectal cancer. Int J Colorectal Dis 2015;30:1165–71.
crossref pmid
55. Huang XZ, Chen WJ, Zhang X, Wu CC, Zhang CY, Sun SS, et al. An elevated platelet-to-lymphocyte ratio predicts poor prognosis and clinicopathological characteristics in patients with colorectal cancer: a meta-analysis. Dis Markers 2017;2017:1053125.
crossref pmid pmc
56. Neofytou K, Smyth EC, Giakoustidis A, Khan AZ, Cunningham D, Mudan S. Elevated platelet to lymphocyte ratio predicts poor prognosis after hepatectomy for liver-only colorectal metastases, and it is superior to neutrophil to lymphocyte ratio as an adverse prognostic factor. Med Oncol 2014;31:239.
crossref pmid
57. McMillan DC, Watson WS, O’Gorman P, Preston T, Scott HR, McArdle CS. Albumin concentrations are primarily determined by the body cell mass and the systemic inflammatory response in cancer patients with weight loss. Nutr Cancer 2001;39:210–3.
crossref pmid
58. McMillan DC. Systemic inflammation, nutritional status and survival in patients with cancer. Curr Opin Clin Nutr Metab Care 2009;12:223–6.
crossref pmid
59. Kowalski-Saunders PW, Winwood PJ, Arthur MJ, Wright R. Reversible inhibition of albumin production by rat hepatocytes maintained on a laminin-rich gel (Engelbreth-Holm-Swarm) in response to secretory products of Kupffer cells and cytokines. Hepatology 1992;16:733–41.
crossref pmid
60. Nazha B, Moussaly E, Zaarour M, Weerasinghe C, Azab B. Hypoalbuminemia in colorectal cancer prognosis: Nutritional marker or inflammatory surrogate? World J Gastrointest Surg 2015;7:370–7.
crossref pmid pmc
61. Chiang JM, Chang CJ, Jiang SF, Yeh CY, You JF, Hsieh PS, et al. Pre-operative serum albumin level substantially predicts post-operative morbidity and mortality among patients with colorectal cancer who undergo elective colectomy. Eur J Cancer Care (Engl) 2017;26:e12403.
crossref
62. Montomoli J, Erichsen R, Antonsen S, Nilsson T, Sørensen HT. Impact of preoperative serum albumin on 30-day mortality following surgery for colorectal cancer: a population-based cohort study. BMJ Open Gastroenterol 2015;2:e000047.
crossref pmid pmc
63. Wang Y, Wang H, Jiang J, Cao X, Liu Q. Early decrease in postoperative serum albumin predicts severe complications in patients with colorectal cancer after curative laparoscopic surgery. World J Surg Oncol 2018;16:192.
crossref pmid pmc
64. Yamamoto M, Saito H, Uejima C, Tanio A, Tada Y, Matsunaga T, et al. Combination of serum albumin and cholinesterase levels as prognostic indicator in patients ith colorectal cancer. Anticancer Res 2019;39:1085–90.
crossref pmid
65. Lai CC, You JF, Yeh CY, Chen JS, Tang R, Wang JY, et al. Low preoperative serum albumin in colon cancer: a risk factor for poor outcome. Int J Colorectal Dis 2011;26:473–81.
crossref pmid
66. Sun LC, Chu KS, Cheng SC, Lu CY, Kuo CH, Hsieh JS, et al. Preoperative serum carcinoembryonic antigen, albumin and age are supplementary to UICC staging systems in predicting survival for colorectal cancer patients undergoing surgical treatment. BMC Cancer 2009;9:288.
crossref pmid pmc
67. Morley JJ, Kushner I. Serum C-reactive protein levels in disease. Ann N Y Acad Sci 1982;389:406–18.
crossref pmid
68. Mahmoud FA, Rivera NI. The role of C-reactive protein as a prognostic indicator in advanced cancer. Curr Oncol Rep 2002;4:250–5.
crossref pmid
69. Shrotriya S, Walsh D, Bennani-Baiti N, Thomas S, Lorton C. Creactive protein is an important biomarker for prognosis tumor recurrence and treatment response in adult solid tumors: a systematic review. PLoS One 2015;10:e0143080.
crossref pmid pmc
70. Koike Y, Miki C, Okugawa Y, Yokoe T, Toiyama Y, Tanaka K, et al. Preoperative C-reactive protein as a prognostic and therapeutic marker for colorectal cancer. J Surg Oncol 2008;98:540–4.
crossref pmid
71. Nozoe T, Matsumata T, Sugimachi K. Preoperative elevation of serum C-reactive protein is related to impaired immunity in patients with colorectal cancer. Am J Clin Oncol 2000;23:263–6.
crossref pmid
72. Nozoe T, Matsumata T, Kitamura M, Sugimachi K. Significance of preoperative elevation of serum C-reactive protein as an indicator for prognosis in colorectal cancer. Am J Surg 1998;176:335–8.
crossref pmid
73. Lu X, Guo W, Xu W, Zhang X, Shi Z, Zheng L, et al. Prognostic value of the Glasgow prognostic score in colorectal cancer: a meta-analysis of 9,839 patients. Cancer Manag Res 2018;11:229–49.
crossref pmid pmc
74. McMillan DC, Crozier JE, Canna K, Angerson WJ, McArdle CS. Evaluation of an inflammation-based prognostic score (GPS) in patients undergoing resection for colon and rectal cancer. Int J Colorectal Dis 2007;22:881–6.
crossref pmid
75. Park JH, Watt DG, Roxburgh CS, Horgan PG, McMillan DC. Colorectal cancer, systemic inflammation, and outcome: staging the tumor and staging the host. Ann Surg 2016;263:326–36.
pmid
76. Moyes LH, Leitch EF, McKee RF, Anderson JH, Horgan PG, McMillan DC. Preoperative systemic inflammation predicts postoperative infectious complications in patients undergoing curative resection for colorectal cancer. Br J Cancer 2009;100:1236–9.
crossref pmid pmc
77. Ishizuka M, Nagata H, Takagi K, Iwasaki Y, Shibuya N, Kubota K. Clinical significance of the C-reactive protein to albumin ratio for survival after surgery for colorectal cancer. Ann Surg Oncol 2016;23:900–7.
crossref pmid
78. Lee JW, Sharma AR, Lee SS, Chun WJ, Kim HS. The C-reactive protein to albumin ratio predicts postoperative complication in patients who undergo gastrectomy for gastric cancer. Heliyon 2020;6:e04220.
crossref pmid pmc
79. Faris FM, Fattah AH, Ali M, Gaber S. C-reactive protein/albumin ratio in correlation to sequential organ failure assessment score: predictive value in early postoperative abdominal sepsis. Egypt J Crit Care Med 2021;8:73–7.

80. Yu Y, Wu Z, Shen Z, Cao Y. Preoperative C-reactive protein-to-albumin ratio predicts anastomotic leak in elderly patients after curative colorectal surgery. Cancer Biomark 2020;27:295–302.
crossref pmid
81. Ge X, Cao Y, Wang H, Ding C, Tian H, Zhang X, et al. Diagnostic accuracy of the postoperative ratio of C-reactive protein to albumin for complications after colorectal surgery. World J Surg Oncol 2017;15:15.
crossref pmid pmc
82. Tominaga T, Nonaka T, Sumida Y, Hidaka S, Sawai T, Nagayasu T. The C-reactive protein to albumin ratio as a predictor of severe side effects of adjuvant chemotherapy in stage III colorectal cancer patients. PLoS One 2016;11:e0167967.
crossref pmid pmc
83. Ide S, Toiyama Y, Okugawa Y, Oki S, Yasuda H, Fujikawa H, et al. Clinical significance of C-reactive protein-to-albumin ratio with rectal cancer patient undergoing chemoradiotherapy followed by surgery. Anticancer Res 2017;37:5797–804.
pmid
84. Haruki K, Shiba H, Horiuchi T, Sakamoto T, Gocho T, Fujiwara Y, et al. Impact of the C-reactive protein to albumin ratio on long-term outcomes after hepatic resection for colorectal liver metastases. Am J Surg 2017;214:752–6.
crossref pmid
85. Shibutani M, Maeda K, Nagahara H, Iseki Y, Ikeya T, Hirakawa K. Prognostic significance of the preoperative ratio of C-reactive protein to albumin in patients with colorectal cancer. Anticancer Res 2016;36:995–1001.
pmid
86. Fan Y, Xiang S, Dai Z, Zou C, Wang X, Gao Z. Prognostic significance of C-reactive protein to albumin ratio in colorectal cancer patients: a meta-analysis. Int J Colorectal Dis 2019;34:1105–11.
crossref pmid
87. Hachiya H, Ishizuka M, Takagi K, Iwasaki Y, Shibuya N, Nishi Y, et al. Clinical significance of the globulin-to-albumin ratio for prediction of postoperative survival in patients with colorectal cancer. Ann Gastroenterol Surg 2018;2:434–41.
crossref pmid pmc
88. Shibutani M, Maeda K, Nagahara H, Ohtani H, Iseki Y, Ikeya T, et al. The pretreatment albumin to globulin ratio predicts chemotherapeutic outcomes in patients with unresectable metastatic colorectal cancer. BMC Cancer 2015;15:347.
crossref pmid pmc
89. Azab B, Kedia S, Shah N, Vonfrolio S, Lu W, Naboush A, et al. The value of the pretreatment albumin/globulin ratio in predicting the long-term survival in colorectal cancer. Int J Colorectal Dis 2013;28:1629–36.
crossref pmid
90. Okugawa Y, Toiyama Y, Yamamoto A, Shigemori T, Ide S, Kitajima T, et al. Lymphocyte-C-reactive protein ratio as promising new marker for predicting surgical and oncological outcomes in colorectal cancer. Ann Surg 2020;272:342–51.
crossref pmid


ABOUT
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
AUTHOR INFORMATION
Editorial Office
Room 1519, Suseo Hyundai Venture-vill, 10 Bamgogae-ro 1-gil, Gangnam-gu, Seoul 06349, Korea
Tel: +82-2-2040-7737    Fax: +82-2-2040-7735    E-mail: editor@coloproctol.org                

Copyright © 2024 by Korean Society of Coloproctology.

Developed in M2PI

Close layer