Skip to main content

ORIGINAL RESEARCH article

Front. Nutr., 16 August 2024
Sec. Clinical Nutrition

Phase angle is a predictor for postoperative complications in colorectal cancer

Xiao-Yu Liu&#x;Xiao-Yu Liu1Bing Kang&#x;Bing Kang2Quan Lv&#x;Quan Lv1Zi-Wei Wang
Zi-Wei Wang1*
  • 1Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
  • 2Department of Clinical Nutrition, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China

Aim: The aim of this study was to develop a validated nomogram to predict the risk of postoperative complications in colorectal cancer (CRC) patients by analyzing the factors that contribute to these complications.

Methods: We retrospectively collected clinical information on patients who underwent CRC surgery at a single clinical center from January 2021 to December 2021. Univariate and multivariate logistic regression analysis to identify independent risk factors for postoperative complications and to develop a predictive model. A receiver operating characteristic (ROC) curve was used to calculate the area under the curve (AUC) to assess the predicted probability. Calibration curve was drawn to compare the predicted probability of the nomogram with the actual probability, and decision curve analysis (DCA) was employed to evaluate the clinical utility of the nomogram.

Results: A total of 190 CRC patients were included in this study. We retrospectively collected baseline information, clinical information, surgical information, and nutrition-related indicators for all patients. Through multivariate logistic regression analysis, preoperative albumin (p = 0.041, OR = 0.906, 95% CI = 0.824–0.996), surgical time (p = 0.009, OR = 1.006, 95% CI = 1.001–1.010), waistline (p = 0.049, OR = 1.011, 95% CI = 1.002–1.020) and phase angle (PA) (p = 0.022, OR = 0.615, 95% CI = 0.405–0.933) were identified as independent risk factors for postoperative complications in CRC, and a nomogram prediction model was established using the above four variables. The AUC of 0.706 for the ROC plot and the high agreement between predicted and actual probabilities in the calibration curves suggested that the prediction model has good predictive power. The DCA also confirmed the good clinical performance of the nomogram.

Conclusion: This study developed a nomogram to predict the risk of postoperative complications in CRC patients, providing surgeons with a reliable reference to personalized patient management in the perioperative period and preoperative nutritional interventions.

Introduction

According to Global Cancer Data 2020, colorectal cancer (CRC) has become the third most common new cancer diagnosis and the second leading cause of cancer-related deaths worldwide (13). In 2020, approximately 1,931,590 people were diagnosed with CRC and 935,173 people died from CRC (4, 5). The World Health Organization (WHO) estimates that by 2030, new cases of CRC will increase by 77% and deaths from CRC will increase by 80% (6, 7). The main treatments for CRC include surgery, local radiotherapy and systemic treatments such as chemotherapy and targeted therapy (8, 9).

Currently, radical surgery remains the cornerstone of CRC treatment (10, 11). Several previous studies have shown that postoperative complications affect the prognosis of CRC patients (1214). Because CRC affects the intake and absorption of food and nutrients, it often leads to malnutrition in patients (15, 16). Malnutrition can cause changes in immune, respiratory and renal function and impairs wound healing (17, 18). Therefore, comprehensive preoperative assessment of the patient’s nutritional status and timely intervention should receive the full attention of clinicians.

Various objective measures are often used to assess the nutritional status of patients, mainly including anthropometric measures, Subjective Gross Assessment (SGA), Nutritional Risk Score 2002 (NRS2002) and Bioelectrical impedance analysis (BIA) related indicators (19, 20). Anthropometric measurements mainly include body mass index (BMI), waistline, hipline, calf line and triceps skin fold (TSF).

SGA is a nutritional assessment of patients based on subjective patient interviews combined with objective measurement data. Patient-generated SGA (PG-SGA) is an adaptation of SGA and is specifically used as a reference method to assess nutritional status in cancer patients (21, 22). Several studies have shown that PG-SGA is associated with a poorer clinical prognosis in cancer patients (2325). The PG-SGA primarily assesses the patient’s weight change over a 2-week period, changes in dietary intake, symptoms of nutritional effects, activity and function, and physical examination findings (26).

NRS-2002 is a screening tool for malnutrition, endorsed by the European Society for Clinical Nutrition and Metabolism (ESPEN) for used in hospitalized patients (27, 28). The NRS-2002 is divided into two sections: the patient’s nutritional status and current illness and severity, which are assessed on the basis of the patient’s age, weight loss, body mass index (BMI), food intake and severity of illness (29).

The aim of this study was to collect clinical information and preoperative nutrition-related indicators from CRC patients, analyze the factors leading to postoperative complications, and to establish a valid nomogram to predict the risk of these complications.

Materials and methods

Patient selection

We retrospectively collected information on patients who underwent CRC surgery from January 2021 to December 2021 at a single clinical center, including baseline information, clinical information, and surgical information. Inclusion criteria were patients with preoperative pathologically confirmed colorectal malignancy who underwent laparoscopic radical surgery. Exclusion criteria were as follows: 1. Patients who underwent CRC surgery after recurrence; 2. Patients with metastatic CRC; and 3. Patients with incomplete baseline or surgical information. Finally, 190 patients were included.

This study was reviewed by the Ethics Committee of the First Affiliated Hospital of Chongqing Medical University (2022–135-2). It complied with the principles of medical ethics and the Declaration of Helsinki, and all patients participating in the study signed an informed consent form.

Data elements

We retrospectively collected baseline information, clinical information, surgical information, and nutrition-related indicators from patients. Baseline information included age, sex, BMI, smoking and drinking history, and preoperative comorbidities. Clinical information included preoperative albumin and hemoglobin, tumor location, tumor stage and tumor size. Surgical information included surgical time, blood loss and postoperative complications. Nutrition-related indicators included waistline, hipline, calf line, TSF, NRS2002, PG-SGA and BIA related indicators.

Preoperative comorbidities included hypertension and type 2 diabetes mellitus (T2DM). The tumor was comprehensively staged according to the 8th edition of the AJCC guidelines and was classified as stage I-IV (30).

BIA

BIA is a non-invasive, practical, objective and simple method of assessing body composition and measuring nutritional status that can be applied to both healthy and patient populations (18, 19, 3134). BIA uses the measurement of impedance as low amplitude (800 mA) and high frequency (50 kHz) electrical currents pass through the body to calculate indicators of body composition such as muscle, fat, cell mass and volumetric loading status (35, 36).

BIA is a body composition analysis technique that quantifies various body tissue components and directly measures impedance, phase angle (PA) and other metrics (37, 38). The indices measured by BIA in this study included: appendicular skeletal muscle mass index (ASMMI), body cell mass (BCM), body cell mass index (BCMI), extracellular water (ECW), fat free mass (FFM), fat free mass index (FFMI), fat mass (FM), fat mass index (FMI), PA, skeletal muscle index (SMI), skeletal muscle mass (SMM), total body water (TBW) (Figure 1).

Figure 1
www.frontiersin.org

Figure 1. Univariate logistic regression analysis of the BIA related indicators. BIA, Bioelectrical impedance analysis; ASMMI, appendicular skeletal muscle mass index; BCM, body cell mass; BCMI, body cell mass index; ECW, extracellular water; FFM, fat free mass; FFMI, fat free mass index; FM, fat mass; FMI, fat mass index; PA, phase angle; SMI, skeletal muscle index; SMM, skeletal muscle mass; TBW, total body water. (A-L) are the results of a univariate logistic analysis of the above indicators of the development of postoperative complications.

Statistical analysis

All data in this study were processed using SPSS (version 22.0) and R (version 4.1.2). Continuous variables that followed a normal distribution were expressed as mean ± standard deviation (SD) and comparisons were made using t-test; continuous variables that did not follow a normal distribution were expressed as median and interquartile range (IQR) and assessed using chi-squared or Fisher’s exact test; and categorical variables were expressed as numbers and percentages. Univariate logistic regression analysis was performed on all variables, and variables with p < 0.05 were considered potential risk factors for the occurrence of postoperative complications in CRC patients. The screened potential risk factors were then subjected to multivariate logistic regression analysis to identify independent predictors of complications after CRC surgery. Finally, variables with p < 0.05 in the multivariate logistic regression analysis were included and a nomogram was created to predict the risk of postoperative complications in CRC.

The predictive model was evaluated in three ways. First, the predictive value of the risk factors was verified using the receiver operating characteristic (ROC) curve, and the area under the curve (AUC) was calculated to assess the performance of the nomogram. The AUC took values between 0 and 1, with 1 indicating perfect agreement, 0.5 indicating no better than chance, and greater than 0.7 indicating that the model has relatively good predictive power (39, 40). Secondly, prediction curves were plotted to test the correction of the postoperative complication risk map, comparing the predicted and actual probabilities of the nomogram, using the 45 degree line as a perfect model with 100% accuracy (41). Finally, decision curve analysis (DCA) was used to analyze the net benefit based on different threshold probabilities to determine the clinical applicability of the nomogram (42, 43).

Results

Baseline information

Based on the above inclusion and exclusion criteria, a total of 190 patients who underwent CRC surgery were included in this study. This included 42 patients in the complication group and 148 patients in the no complication group. Comparing the baseline information of the two groups of patients, we found a statistical difference (p < 0.05), including preoperative albumin (p = 0.005), surgical time (p = 0.004), blood loss (p = 0.010), and waistline (p = 0.043; Table 1).

Table 1
www.frontiersin.org

Table 1. Baseline information between the complications group and no complications group.

Nomogram variable screening

Univariate and multivariate logistic regression analysis were performed on a total of 32 potential factors, including baseline information, clinical information, surgical information, and nutrition-related indicators, to find the risk factors affecting the occurrence of complications after CRC surgery. The results of univariate logistic regression analysis showed that preoperative albumin (p = 0.005, OR = 0.879, 95% CI = 0.804–0.962), surgical time (p = 0.009, OR = 1.006, 95% CI = 1.001–1.010), waistline (p = 0.046, OR = 1.042, 95% CI = 1.001–1.084) and PA (p = 0.016, OR = 0.622, 95% CI = 0.424–0.914) were the potential risk factors for postoperative complications of CRC (Table 1; Figure 1). Further multivariate logistic regression analysis of the four potential risk factors showed that preoperative albumin (p = 0.041, OR = 0.906, 95% CI = 0.824–0.996), surgical time (p = 0.009, OR = 1.006, 95% CI = 1.001–1.010), waistline (p = 0.049, OR = 1.011, 95% CI = 1.002–1.020) and PA (p = 0.022, OR = 0.615, 95% CI = 0.405–0.933) were independent risk factors for the occurrence of postoperative complications in CRC (Table 2).

Table 2
www.frontiersin.org

Table 2. Multivariate logistic regression analysis of the complications.

Nomogram construction and validation

A nomogram model for predicting the risk of postoperative complications in CRC patients was constructed using four independent risk factors identified by multivariate logistic regression analysis. As shown in Figure 2, the corresponding scores of each factor were obtained according to the patients’ own actual situation, and the total score was obtained by adding the four scores, and the final predicted risk of postoperative complications was the probability corresponding to an individual patient’s total score.

Figure 2
www.frontiersin.org

Figure 2. Nomogram for predicting the risk of postoperative complications after CRC surgery. CRC, colorectal cancer.

By ROC analysis, AUC of the nomogram over time was 0.706, indicating that the model has good predictive performance (Figure 3). The calibration curve showed that there was a high degree of agreement between the predicted and observed results of the nomogram model constructed in this study (Figure 4). Finally, DCA was used to evaluate the clinical application value of the prediction model, as shown in Figure 5.

Figure 3
www.frontiersin.org

Figure 3. ROC curves of the nomogram. ROC, receiver operating characteristic; AUC, area under the curve.

Figure 4
www.frontiersin.org

Figure 4. Calibration curve for the nomogram.

Figure 5
www.frontiersin.org

Figure 5. DCA for the nomogram. DCA, decision curve analysis.

Discussion

A total of 190 patients undergoing CRC surgery were included in this study, including 42 patients in the complication group and 148 patients in the no complication group. Multivariate logistic regression analysis revealed that preoperative albumin, surgical time, waistline and PA were independent risk factors for postoperative complications of CRC. Using these four independent risk factors, we constructed a nomogram model to predict the risk of postoperative complications in CRC patients.

The results of this study suggested that lower preoperative albumin, longer surgical time, larger waistline and smaller PA were independent risk factors for postoperative complications in CRC. Several previous studies have shown that low preoperative albumin levels were a risk factor for postoperative complications and survival in several types of cancer, including CRC (4447). This might be because low preoperative albumin was a marker of malnutrition, cancer cachexia and chronic inflammatory activity, among other factors (48, 49).

The results of the multivariate logistic regression analysis showed that the duration of surgery was an independent risk factor for the development of postoperative complications in CRC, which was consistent with previous literature (50, 51). Prolonged surgical time not only increased tissue contraction, causing tissue ischaemia, desiccation and necrosis, but also led to coagulation, hemostasis and endothelial damage, thereby increasing the risk of venous thromboembolism (52, 53). In addition, the effectiveness of antimicrobial prophylaxis decreased over time and the likelihood of aseptic technique violations increases (54). Longer surgical time also meant longer anesthesia time and increased fatigue for the surgical team (55).

Waistline was an indicator of central obesity and was often considered a more useful perioperative risk assessment tool than BMI in CRC surgery (56, 57). Increased waistline affected the incidence of postoperative complications in CRC, which might be due to prolonged surgical time, relative wound hypoxia due to reduced subcutaneous tissue oxygenation, impaired collagen synthesis and immune system function (58, 59). Moreover, abdominal obesity was often associated with high levels of inflammatory mediators such as interleukin-6, c-reactive protein and tumor necrosis factor-α, which often led to insulin resistance and postoperative complications (60, 61).

PA was a BIA correlated that was directly related to the quality and integrity of cell membranes, and was often considered a marker of cellular health (62, 63). PA was closely related to the level of inflammation in the body, and inflammation and low albumin were associated with altered fluid distribution in the extracellular space, which could reduce the PA (64, 65). In addition, PA was a marker of oxidative stress, whereby an imbalance between oxidants and antioxidants produced oxidative stress, leading to the destruction of cellular components such as proteins or lipids, which could lead to cellular damage and rupture of cell membranes (6567).

However, this study still has some limitations. First, this was a single-center retrospective study. Secondly, the nomogram prediction model developed in this study was not internally validated, and we will continue to collect clinical information and nutritional indicators from the patients concerned to improve internal validation. The application of deep learning algorithms has the potential to improve the accuracy and effectiveness of CRC detection (68, 69), however, due to the small sample size of this study, the application of deep learning algorithms was not carried out and it is expected that in the future appropriate validation can be carried out when large data sets are available.

In conclusion, this study created a nomogram to predict the risk of postoperative complications in CRC patients, providing surgeons with a reliable reference to personalized patient management in the perioperative period and preoperative nutritional interventions.

Data availability statement

The raw data supporting the conclusions of this article will be made available by the authors, without undue reservation.

Ethics statement

The studies involving humans were approved by this study was reviewed by the Ethics Committee of the First Affiliated Hospital of Chongqing Medical University (2022-135-2), and all patients in this study signed an informed consent form. The studies were conducted in accordance with the local legislation and institutional requirements. Written informed consent for participation was not required from the participants or the participants' legal guardians/next of kin because the present study was performed according to the World Medical Association Declaration of Helsinki. Ethical approval from the institutional review board (K2024-002-01) was obtained. In addition, we applied for a waiver of informed consent.

Author contributions

X-YL: Conceptualization, Data curation, Methodology, Writing – original draft. QL: Data curation, Investigation, Software, Writing – original draft, Project administration. BK: Conceptualization, Investigation, Project administration, Software, Writing – original draft. Z-WW: Formal analysis, Software, Validation, Writing – review & editing, Methodology.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Acknowledgments

We acknowledge all the authors whose publications are referred in our article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Xia, C, Dong, X, Li, H, Cao, M, Sun, D, He, S, et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chin Med J. (2022) 135:584–90. doi: 10.1097/CM9.0000000000002108

PubMed Abstract | Crossref Full Text | Google Scholar

2. Miller, KD, Nogueira, L, Mariotto, AB, Rowland, JH, Yabroff, KR, Alfano, CM, et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. (2019) 69:363–85. doi: 10.3322/caac.21565

Crossref Full Text | Google Scholar

3. Liu, XY, Zhang, B, Cheng, YX, Tao, W, Yuan, C, Wei, ZQ, et al. Does chronic kidney disease affect the complications and prognosis of patients after primary colorectal cancer surgery? World J Gastrointest Oncol. (2022) 14:1199–209. doi: 10.4251/wjgo.v14.i6.1199

PubMed Abstract | Crossref Full Text | Google Scholar

4. Siegel, RL, Wagle, NS, Cercek, A, Smith, RA, and Jemal, A. Colorectal cancer statistics, 2023. CA Cancer J Clin. (2023) 73:233–54. doi: 10.3322/caac.21772

Crossref Full Text | Google Scholar

5. Engholm, G, Storm, HH, Ferlay, J, Christensen, N, Johannesen, TB, Klint, Å, et al. NORDCAN: Cancer incidence, mortality, prevalence and survival in the Nordic countries, version 8.2 (26.03.2019). (2019).

Google Scholar

6. Sung, H, Ferlay, J, Siegel, RL, Laversanne, M, Soerjomataram, I, Jemal, A, et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2021) 71:209–49. doi: 10.3322/caac.21660

PubMed Abstract | Crossref Full Text | Google Scholar

7. Binefa, G, Rodríguez-Moranta, F, Teule, A, and Medina-Hayas, M. Colorectal cancer: from prevention to personalized medicine. World J Gastroenterol. (2014) 20:6786–808. doi: 10.3748/wjg.v20.i22.6786

PubMed Abstract | Crossref Full Text | Google Scholar

8. Liu, XY, Li, ZW, Zhang, B, Liu, F, Zhang, W, and Peng, D. Effects of preoperative bicarbonate and lactate levels on short-term outcomes and prognosis in elderly patients with colorectal cancer. BMC Surg. (2023) 23:127. doi: 10.1186/s12893-023-02039-x

Crossref Full Text | Google Scholar

9. Kuipers, EJ, Grady, WM, Lieberman, D, Seufferlein, T, Sung, JJ, Boelens, PG, et al. Colorectal cancer. Nat Rev Dis Primers. (2015) 1:15065. doi: 10.1038/nrdp.2015.65

PubMed Abstract | Crossref Full Text | Google Scholar

10. Chern, YJ, You, JF, Cheng, CC, Jhuang, JR, Yeh, CY, Hsieh, PS, et al. Decreasing postoperative pulmonary complication following laparoscopic surgery in elderly individuals with colorectal Cancer: a competing risk analysis in a propensity score-weighted cohort study. Cancers (Basel). (2021) 14:131. doi: 10.3390/cancers14010131

Crossref Full Text | Google Scholar

11. Dekker, E, Tanis, PJ, Vleugels, JLA, Kasi, PM, and Wallace, MB. Colorectal cancer. Lancet. (2019) 394:1467–80. doi: 10.1016/S0140-6736(19)32319-0

Crossref Full Text | Google Scholar

12. Liu, XY, Li, ZW, Kang, B, Cheng, YX, Tao, W, Zhang, B, et al. Does preoperative waiting time affect the short-term outcomes and prognosis of colorectal Cancer patients? A retrospective study from the west of China. Can J Gastroenterol Hepatol. (2022) 2022:8235736. doi: 10.1155/2022/8235736

Crossref Full Text | Google Scholar

13. Nowakowski, M, Pisarska, M, Rubinkiewicz, M, Torbicz, G, Gajewska, N, Mizera, M, et al. Postoperative complications are associated with worse survival after laparoscopic surgery for non-metastatic colorectal cancer - interim analysis of 3-year overall survival. Wideochir Inne Tech Maloinwazyjne. (2018) 13:326–32. doi: 10.5114/wiitm.2018.76179

PubMed Abstract | Crossref Full Text | Google Scholar

14. Liu, XY, Zhang, B, Kang, B, Cheng, YX, Yuan, C, Tao, W, et al. The effect of complications on oncological outcomes of colorectal Cancer patients after primary surgery: a propensity score matching analysis. Front Oncol. (2022) 12:857062. doi: 10.3389/fonc.2022.857062

Crossref Full Text | Google Scholar

15. Vandewoude, M. Nutritional assessment in geriatric cancer patients. Support Care Cancer. (2010) 18:S51–6. doi: 10.1007/s00520-009-0755-7

Crossref Full Text | Google Scholar

16. Dewys, WD, Begg, C, Lavin, PT, Band, PR, Bennett, JM, Bertino, JR, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern cooperative oncology group. Am J Med. (1980) 69:491–7. doi: 10.1016/s0149-2918(05)80001-3

Crossref Full Text | Google Scholar

17. Cavagnari, MAV, Silva, TD, Pereira, MAH, Sauer, LJ, Shigueoka, D, Saad, SS, et al. Impact of genetic mutations and nutritional status on the survival of patients with colorectal cancer. BMC Cancer. (2019) 19:644. doi: 10.1186/s12885-019-5837-4

Crossref Full Text | Google Scholar

18. Barbosa-Silva, MC, and Barros, AJ. Bioelectric impedance and individual characteristics as prognostic factors for post-operative complications. Clin Nutr. (2005) 24:830–8. doi: 10.1016/j.clnu.2005.05.005

Crossref Full Text | Google Scholar

19. Gupta, D, Lammersfeld, CA, Burrows, JL, Dahlk, SL, Vashi, PG, Grutsch, JF, et al. Bioelectrical impedance phase angle in clinical practice: implications for prognosis in advanced colorectal cancer. Am J Clin Nutr. (2004) 80:1634–8. doi: 10.1093/ajcn/80.6.1634

PubMed Abstract | Crossref Full Text | Google Scholar

20. Bauer, J, Capra, S, and Ferguson, M. Use of the scored patient-generated subjective global assessment (PG-SGA) as a nutrition assessment tool in patients with cancer. Eur J Clin Nutr. (2002) 56:779–85. doi: 10.1038/sj.ejcn.1601412

Crossref Full Text | Google Scholar

21. Barao, K, Abe Vicente Cavagnari, M, Silva Fucuta, P, and Manoukian Forones, N. Association between nutrition status and survival in elderly patients with colorectal Cancer. Nutr Clin Pract. (2017) 32:658–63. doi: 10.1177/0884533617706894

Crossref Full Text | Google Scholar

22. Ottery, FD. Definition of standardized nutritional assessment and interventional pathways in oncology. Nutrition. (1996) 12:S15–9. doi: 10.1016/0899-9007(96)90011-8

Crossref Full Text | Google Scholar

23. Mota, AP, Aredes, MA, De Oliveira Miguel, J, and Chaves, GV. Nutritional status assessed by Patient-Generated Subjective Global Assessment is associated with toxicity to chemoradiotherapy in women with cervical cancer: a prospective study. Eur J Clin Nutr. (2022) 76:1740–7. doi: 10.1038/s41430-022-01180-9

Crossref Full Text | Google Scholar

24. Ge, T, Lin, T, Yang, J, and Wang, M. Nutritional status and related factors of patients with advanced lung cancer in northern China: a retrospective study. Cancer Manag Res. (2019) 11:2225–31. doi: 10.2147/CMAR.S193567

PubMed Abstract | Crossref Full Text | Google Scholar

25. Kim, HS, Lee, JY, Lim, SH, Cho, J, Kim, SJ, Jang, JH, et al. Patient-generated subjective global assessment as a prognosis tool in patients with multiple myeloma. Nutrition. (2017) 36:67–71. doi: 10.1016/j.nut.2016.06.009

Crossref Full Text | Google Scholar

26. Jager-Wittenaar, H, and Ottery, FD. Assessing nutritional status in cancer: role of the patient-generated subjective global assessment. Curr Opin Clin Nutr Metab Care. (2017) 20:322–9. doi: 10.1097/MCO.0000000000000389

Crossref Full Text | Google Scholar

27. Kyle, UG, Kossovsky, MP, Karsegard, VL, and Pichard, C. Comparison of tools for nutritional assessment and screening at hospital admission: a population study. Clin Nutr. (2006) 25:409–17. doi: 10.1016/j.clnu.2005.11.001

PubMed Abstract | Crossref Full Text | Google Scholar

28. Velasco, C, García, E, Rodríguez, V, Frias, L, Garriga, R, Álvarez, J, et al. Comparison of four nutritional screening tools to detect nutritional risk in hospitalized patients: a multicentre study. Eur J Clin Nutr. (2011) 65:269–74. doi: 10.1038/ejcn.2010.243

PubMed Abstract | Crossref Full Text | Google Scholar

29. Bakkaloglu, OK, Bektas, M, Ince, B, Amikishiyev, S, Tor, YB, Altınkaynak, M, et al. Malnutrition risk in hospitalized patients measured with nutrition risk screening 2002 tool and its association with in-hospital mortality. Eur Rev Med Pharmacol Sci. (2023) 27:5812–21. doi: 10.26355/eurrev_202306_32820

Crossref Full Text | Google Scholar

30. Weiser, MR. AJCC 8th edition: colorectal Cancer. Ann Surg Oncol. (2018) 25:1454–5. doi: 10.1245/s10434-018-6462-1

Crossref Full Text | Google Scholar

31. Paiva, SI, Borges, LR, Halpern-Silveira, D, Assunção, MCF, Barros, AJD, and Gonzalez, MC. Standardized phase angle from bioelectrical impedance analysis as prognostic factor for survival in patients with cancer. Support Care Cancer. (2010) 19:187–92. doi: 10.1007/s00520-009-0798-9

PubMed Abstract | Crossref Full Text | Google Scholar

32. Gupta, D, Lis, CG, Dahlk, SL, King, J, Vashi, PG, Grutsch, JF, et al. The relationship between bioelectrical impedance phase angle and subjective global assessment in advanced colorectal cancer. Nutr J. (2008) 7:19. doi: 10.1186/1475-2891-7-19

Crossref Full Text | Google Scholar

33. Kroc, Ł, Fife, E, Piechocka-Wochniak, E, Sołtysik, B, and Kostka, T. Comparison of nutrition risk screening 2002 and subjective global assessment form as short nutrition assessment tools in older hospitalized adults. Nutrients. (2021) 13:225. doi: 10.3390/nu13010225

PubMed Abstract | Crossref Full Text | Google Scholar

34. Barbosa-Silva, MC, Barros, AJ, Wang, J, Heymsfield, SB, and Pierson, RN. Bioelectrical impedance analysis: population reference values for phase angle by age and sex. Am J Clin Nutr. (2005) 82:49–52. doi: 10.1093/ajcn.82.1.49

Crossref Full Text | Google Scholar

35. Kyle, UG, Bosaeus, I, de Lorenzo, AD, Deurenberg, P, Elia, M, Gómez, JM, et al. Bioelectrical impedance analysis--part I: review of principles and methods. Clin Nutr. (2004) 23:1226–43. doi: 10.1016/j.clnu.2004.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

36. Wang, W, Meng, X, Liu, J, Lou, X, Zhang, P, He, P, et al. Study on the correlation between bioelectrical impedance analysis index and protein energy consumption in maintenance dialysis patients. Nutr J. (2023) 22:56. doi: 10.1186/s12937-023-00890-5

Crossref Full Text | Google Scholar

37. Ruperto, M, and Barril, G. Extracellular mass to body cell mass ratio as a potential index of wasting and fluid overload in hemodialysis patients. Clin Nutr. (2020) 39:316–7. doi: 10.1016/j.clnu.2019.11.019

Crossref Full Text | Google Scholar

38. Rymarz, A, Bartoszewicz, Z, Szamotulska, K, and Niemczyk, S. The associations between body cell mass and nutritional and inflammatory markers in patients with chronic kidney disease and in subjects without kidney disease. J Ren Nutr. (2016) 26:87–92. doi: 10.1053/j.jrn.2015.09.005

PubMed Abstract | Crossref Full Text | Google Scholar

39. Liu, Q, Zhou, Q, Song, M, Zhao, F, Yang, J, Feng, X, et al. A nomogram for predicting the risk of sepsis in patients with acute cholangitis. J Int Med Res. (2020) 48:66100. doi: 10.1177/0300060519866100

Crossref Full Text | Google Scholar

40. Yu, P, Kan, R, Meng, X, Wang, Z, Xiang, Y, Mao, B, et al. A nomogram for predicting the risk of CKD based on Cardiometabolic risk factors. Int J Gen Med. (2023) 16:4143–54. doi: 10.2147/IJGM.S425122

PubMed Abstract | Crossref Full Text | Google Scholar

41. Lin, Z, Li, Y, Wu, J, Zheng, H, and Yang, C. Nomogram for prediction of prolonged postoperative ileus after colorectal resection. BMC Cancer. (2022) 22:1273. doi: 10.1186/s12885-022-10377-x

Crossref Full Text | Google Scholar

42. Shi, B, Shen, L, Huang, W, Cai, L, Yang, S, Zhang, Y, et al. A nomogram for predicting surgical timing in neonates with necrotizing Enterocolitis. J Clin Med. (2023) 12:3062. doi: 10.3390/jcm12093062

PubMed Abstract | Crossref Full Text | Google Scholar

43. Huai, J, Ye, X, and Ding, J. Nomogram for the prediction of delayed colorectal post-polypectomy bleeding. Turk J Gastroenterol. (2021) 32:727–34. doi: 10.5152/tjg.2021.20842

Crossref Full Text | Google Scholar

44. Uppal, S, al-Niaimi, A, Rice, LW, Rose, SL, Kushner, DM, Spencer, RJ, et al. Preoperative hypoalbuminemia is an independent predictor of poor perioperative outcomes in women undergoing open surgery for gynecologic malignancies. Gynecol Oncol. (2013) 131:416–22. doi: 10.1016/j.ygyno.2013.08.011

PubMed Abstract | Crossref Full Text | Google Scholar

45. Lee, KG, Lee, HJ, Yang, JY, Oh, SY, Bard, S, Suh, YS, et al. Risk factors associated with complication following gastrectomy for gastric cancer: retrospective analysis of prospectively collected data based on the Clavien-Dindo system. J Gastrointest Surg. (2014) 18:1269–77. doi: 10.1007/s11605-014-2525-1

PubMed Abstract | Crossref Full Text | Google Scholar

46. Kudsk, KA, Tolley, EA, DeWitt, RC, Janu, PG, Blackwell, AP, Yeary, S, et al. Preoperative albumin and surgical site identify surgical risk for major postoperative complications. JPEN J Parenter Enteral Nutr. (2003) 27:1–9. doi: 10.1177/014860710302700101

PubMed Abstract | Crossref Full Text | Google Scholar

47. 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 Color Dis. (2011) 26:473–81. doi: 10.1007/s00384-010-1113-4

Crossref Full Text | Google Scholar

48. Braga, M, Ljungqvist, O, Soeters, P, Fearon, K, Weimann, A, Bozzetti, F, et al. ESPEN guidelines on parenteral nutrition: surgery. Clin Nutr. (2009) 28:378–86. doi: 10.1016/j.clnu.2009.04.002

Crossref Full Text | Google Scholar

49. Ataseven, B, du Bois, A, Reinthaller, A, Traut, A, Heitz, F, Aust, S, et al. Pre-operative serum albumin is associated with post-operative complication rate and overall survival in patients with epithelial ovarian cancer undergoing cytoreductive surgery. Gynecol Oncol. (2015) 138:560–5. doi: 10.1016/j.ygyno.2015.07.005

PubMed Abstract | Crossref Full Text | Google Scholar

50. Cheng, H, Clymer, JW, Po-Han Chen, B, Sadeghirad, B, Ferko, NC, Cameron, CG, et al. Prolonged operative duration is associated with complications: a systematic review and meta-analysis. J Surg Res. (2018) 229:134–44. doi: 10.1016/j.jss.2018.03.022

Crossref Full Text | Google Scholar

51. Haddock, NT, Wen, YE, Steppe, C, Shang, Z, and Teotia, SS. Operative time predicts postoperative outcomes in bilateral DIEP flap reconstruction: multivariate 1000 flap analysis. Plast Reconstr Surg Glob Open. (2022) 10:e4713. doi: 10.1097/GOX.0000000000004713

Crossref Full Text | Google Scholar

52. Bekelis, K, Labropoulos, N, and Coy, S. Risk of venous thromboembolism and operative duration in patients undergoing neurosurgical procedures. Neurosurgery. (2017) 80:787–92. doi: 10.1093/neuros/nyw129

Crossref Full Text | Google Scholar

53. Kim, JY, Khavanin, N, Rambachan, A, McCarthy, RJ, Mlodinow, AS, de Oliveria, GS, et al. Surgical duration and risk of venous thromboembolism. JAMA Surg. (2015) 150:110–7. doi: 10.1001/jamasurg.2014.1841

Crossref Full Text | Google Scholar

54. Korol, E, Johnston, K, Waser, N, Sifakis, F, Jafri, HS, Lo, M, et al. A systematic review of risk factors associated with surgical site infections among surgical patients. PLoS One. (2013) 8:e83743. doi: 10.1371/journal.pone.0083743

Crossref Full Text | Google Scholar

55. Shtarbanov, P, Ioannidi, L, Hamilton, S, Ghali, S, Mosahebi, A, Ahmed, Z, et al. Prolonged operative time is a risk factor for adverse postoperative outcomes in the unilateral deep inferior epigastric perforator (DIEP) flap surgery: a retrospective cohort study. J Plast Reconstr Aesthet Surg. (2023) 87:180–6. doi: 10.1016/j.bjps.2023.07.048

Crossref Full Text | Google Scholar

56. Kartheuser, AH, Leonard, DF, Penninckx, F, Paterson, HM, Brandt, D, Remue, C, et al. Waist circumference and waist/hip ratio are better predictive risk factors for mortality and morbidity after colorectal surgery than body mass index and body surface area. Ann Surg. (2013) 258:722–30. doi: 10.1097/SLA.0b013e3182a6605a

Crossref Full Text | Google Scholar

57. Balentine, CJ, Robinson, CN, Marshall, CR, Wilks, J, Buitrago, W, Haderxhanaj, K, et al. Waist circumference predicts increased complications in rectal cancer surgery. J Gastrointest Surg. (2010) 14:1669–79. doi: 10.1007/s11605-010-1343-3

PubMed Abstract | Crossref Full Text | Google Scholar

58. Pierpont, YN, Dinh, TP, Salas, RE, Johnson, EL, Wright, TG, Robson, MC, et al. Obesity and surgical wound healing: a current review. ISRN Obes. (2014) 2014:638936. doi: 10.1155/2014/638936

Crossref Full Text | Google Scholar

59. Kurmann, A, Vorburger, SA, Candinas, D, and Beldi, G. Operation time and body mass index are significant risk factors for surgical site infection in laparoscopic sigmoid resection: a multicenter study. Surg Endosc. (2011) 25:3531–4. doi: 10.1007/s00464-011-1753-7

PubMed Abstract | Crossref Full Text | Google Scholar

60. Weisberg, SP, McCann, D, Desai, M, Rosenbaum, M, Leibel, RL, Ferrante, AW, et al. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. (2003) 112:1796–808. doi: 10.1172/JCI19246

Crossref Full Text | Google Scholar

61. Gurunathan, U, Rapchuk, IL, Dickfos, M, Larsen, P, Forbes, A, Martin, C, et al. Association of Obesity with Septic Complications after Major Abdominal Surgery: a secondary analysis of the RELIEF randomized clinical trial. JAMA Netw Open. (2019) 2:e1916345. doi: 10.1001/jamanetworkopen.2019.16345

PubMed Abstract | Crossref Full Text | Google Scholar

62. Dittmar, M. Reliability and variability of bioimpedance measures in normal adults: effects of age, gender, and body mass. Am J Phys Anthropol. (2003) 122:361–70. doi: 10.1002/ajpa.10301

Crossref Full Text | Google Scholar

63. Cimmino, F, Petrella, L, Cavaliere, G, Ambrosio, K, Trinchese, G, Monda, V, et al. A bioelectrical impedance analysis in adult subjects: the relationship between phase angle and body cell mass. J Funct Morphol Kinesiol. (2023) 8:107. doi: 10.3390/jfmk8030107

PubMed Abstract | Crossref Full Text | Google Scholar

64. Lukaski, HC, Kyle, UG, and Kondrup, J. Assessment of adult malnutrition and prognosis with bioelectrical impedance analysis: phase angle and impedance ratio. Curr Opin Clin Nutr Metab Care. (2017) 20:330–9. doi: 10.1097/MCO.0000000000000387

Crossref Full Text | Google Scholar

65. Lee, GR, and Kim, EY. Usefulness of phase angle on bioelectrical impedance analysis as a surveillance tool for postoperative infection in critically ill patients. Front Med (Lausanne). (2023) 10:1111727. doi: 10.3389/fmed.2023.1111727

Crossref Full Text | Google Scholar

66. Da Silva, BR, Gonzalez, MC, Cereda, E, and Prado, CM. Exploring the potential role of phase angle as a marker of oxidative stress: a narrative review. Nutrition. (2022) 93:111493. doi: 10.1016/j.nut.2021.111493

PubMed Abstract | Crossref Full Text | Google Scholar

67. Rosas-Carrasco, O, Núñez-Fritsche, G, López-Teros, MT, Acosta-Méndez, P, Cruz-Oñate, JC, Navarrete-Cendejas, AY, et al. Low muscle strength and low phase angle predicts greater risk to mortality than severity scales (APACHE, SOFA, and CURB-65) in adults hospitalized for SARS-CoV-2 pneumonia. Front Nutr. (2022) 9:965356. doi: 10.3389/fnut.2022.965356

PubMed Abstract | Crossref Full Text | Google Scholar

68. Bousis, D, Verras, GI, Bouchagier, K, Antzoulas, A, Panagiotopoulos, I, Katinioti, A, et al. The role of deep learning in diagnosing colorectal cancer. Prz Gastroenterol. (2023) 18:266–73. doi: 10.5114/pg.2023.129494

PubMed Abstract | Crossref Full Text | Google Scholar

69. Chlorogiannis, DD, Verras, G, Tzelepi, V, Chlorogiannis, A, Filippiadis, DK, Kelekis, A, et al. Tissue classification and diagnosis of colorectal cancer histopathology images using deep learning algorithms. Is the time ripe for clinical practice implementation? Prz Gastroenterol. (2023) 18:353–67. doi: 10.5114/pg.2023.130337

Crossref Full Text | Google Scholar

Keywords: colorectal cancer, complications, surgery, phase angle, nomogram

Citation: Liu X-Y, Kang B, Lv Q and Wang Z-W (2024) Phase angle is a predictor for postoperative complications in colorectal cancer. Front. Nutr. 11:1446660. doi: 10.3389/fnut.2024.1446660

Received: 10 June 2024; Accepted: 02 August 2024;
Published: 16 August 2024.

Edited by:

Francesk Mulita, General University Hospital of Patras, Greece

Reviewed by:

Nallely Bueno-Hernández, General Hospital of Mexico, Mexico
Angelis Peteinaris, University of Patras, Greece
Andreas Antzoulas, General University Hospital of Patras, Greece

Copyright © 2024 Liu, Kang, Lv and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zi-Wei Wang, wangziwe571@sina.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.