The prognostic significance of Lung Immune Prognostic Index (LIPI) and Pan-immune-inflammation value (PIV) in patients with large cell neuroendocrine carcinoma

DOI: https://doi.org/10.21203/rs.3.rs-2363112/v1

Abstract

Large cell neuroendocrine carcinomas (LCNEC) are rare tumors of the lung. Due to their rarity, there is no standard follow-up and treatment. Inflammation and immune systems play an important role in the pathogenesis, development, and progression of lung and other cancers. Lung Immune Prognostic Index (LIPI) and Pan-immune-inflammation value (PIV) are markers of inflammation and immune marker, and there is no study with LCNEC. Therefore, in our study to evaluate the relationship between these markers and LCNEC prognosis; We retrospectively analyzed 109 patients diagnosed with metastatic LCNEC in our center between 2009 and 2022 by calculating the LIPI and PIV values at the time of diagnosis. Median OS  was 7.8 (95% Confidence interval (CI), 6.20-9.39) months. Respectively median OS times were 9.6 (95% CI, 7.28-11.91) months versus 4.5 (95% CI, 2.87-6.13) months in low and high LIPI scores. Median OS times were 10.25 months (95 CI, 9.12-11.32) compared to 3.95 months (95 CI, 2.5-6.36) in low and high PIV, respectively.  In multivariate analysis ECOG performance score ( p=0.005), metastasis time (de novo vs recurrent) (p=0.008), platelet (p=0.04), albumin (p=0.026), lymphocyte count (p =0.037), LIPI (p =0.002)  score and PIV (p =0.001) were related with survival time. In conclusion LIPI, PIV, ECOG performance score, de novo metastases, albumin levels, lymphocyte and platelet counts are associated with prognosis. These factors can be used in patient monitoring and treatment as simple and inexpensive biomarkers.

Introduction

Neuroendocrine tumors constitute 20% of lung cancers [1]. LCNEC is extremely rare and constitutes 2–3% of all lung cancers [1]. They were first defined by Travis et al [2]. They have a high grade, aggressive course, and poor prognosis. Considering the 5-year survival of the patients in all stages, it varies between 15–57% [35]. In 2015, the World Health Organization classified small cell lung cancer (NSCLC) and LCNEC together as separate groups of high-grade lung neuroendocrine tumors [6]. However, they show clinical, biological, and pathological features similar to SCLC and non-small cell lung cancer (NSCLC). Since LCNEC is rarely seen and therefore randomized clinical trials cannot be performed, there is no standard approach for its follow-up and treatment. Information on follow-up and treatment of patients is based on retrospective data, and case reports, and is often derived from LCNEC adaptation of results from SCLC and NSCLC.

In recent years, numerous studies have been reported on blood parameters showing inflammation, the immune system, and the prognosis of SCLC, NSCLC, and many other cancers[78]. By using blood parameters, new markers, scoring and nomograms were developed to obtain information about cancer prognosis. LIPI and PIV are among these scorings and be associated with prognosis in many cancers. However, there are no studies on the prognostic and surveillance of LIPI and PIV with LCNEC. In our study, we aimed to examine the relationship between LIPI and PIV results at the time of metastasis, prognosis, and surveillance in LCNEC patients.

Material And Methods

Data collection

Patients, who were followed up with a diagnosis of LCNEC in the Medical Oncology Department of Manisa City Hospital between 2009 and 2022, were retrospectively analyzed. The demographic characteristics of the patients, such as age and sex, sites of metastasis, metastasis time( de novo vs recurrent) Eastern Cooperative Oncology Group (ECOG) performance score, smoking habit, smoking pack year ((packs of cigarettes smoked per day) multiply (year)), albumin, hemoglobin, lactate dehydrogenase( LDH), uric acid levels, white blood cell count, lymphocyte and platelet counts. LIPI, PIV, and overall survival (OS) time were recorded, and the correlation between OS and other parameters was retrospectively analyzed. OS time referred to the time from the date of diagnosis to the death of the patient. The PIV was calculated using the following formula: neutrophil (103/mm3) x monocyte (103/mm3) x platelet (103/mm3)/lymphocyte (103/mm3). LIPI is a marker combining the derived neutrophil-to-lymphocyte ratio (dNLR, calculated as neutrophil count/white blood cell- neutrophil count) and serum lactate dehydrogenase (LDH). dNLR ≥ 3 and LDH higher than the upper limit of normal (ULN) are defined as “Poor (LIPI 2)”, patients with dNLR ≥ 3 and LDH lower than ULN or dNLR < 3 and LDH higher than ULN are defined as “Intermediate (LIPI 1)”, and patients with dNLR < 3 and LDH lower than ULN are defined as “Good (LIPI 0). LIPI values were defined as LIPI 0–1 low LIPI and LIPI 2 Hıgh LIPI. The median PIV value of the patients was 600 (103/mm3). PIV values were defined as 600 > low PIV and 600 ≤ high PIV. Patients according to ECOG score (3 > or 3≤), Metastasis time (de novo or recurrent), PIV(Low or High), and LIPI ( Low or High) divided into groups.

Statistical analysis

Descriptive statistics were presented as mean, standard deviation, median, minimum and maximum values for numerical variables, and as numbers and percentages for categorical variables. The comparison of numerical variables between two independent groups was performed using Student’s t-test in case of normal distribution and the Mann-Whitney U test otherwise. Rates were compared between the groups using the chi-square analysis and Fisher’s exact test. Survival analyses were undertaken with the Kaplan-Meier method. Determinative factors were examined using the Cox regression analysis. P < 0.05 was considered significant in all statistical analyses.

Results

A total of 109 patients, 55 (50.46%) male, and 54 (49.54%) female, with a mean age of 61.86 ± 9.97 years, were retrospectively analyzed. All patients had a habit of smoking. The median packs of cigarettes smoked per year was 50 (10–150). 77 (70.64%) patients were metastatic at the time of diagnosis and metastasis was developed in 32 patients during follow-up (Table 1). Median OS was 7.8 (95% CI, 6.20–9.39) months. Respectively median OS was 9.6 (95% CI, 7.28–11.91) months vs 4.5 (95% CI, 2.87–6.13) months in low and high LIPI scores and 10.25 (95% CI, 9.12–11.32) months vs 3.95 (95% CI, 2.5–6.36) months in low and high PIV. ECOG performance score, smoking habit (smoking pack year), metastasis time ( de novo vs recurrent), liver, bone, adrenal metastasis, LDH, uric acid, albumin level, lymphocyte, platelet, LIPI score, and PIV values were associated with median OS. And In multivariate analysis ECOG performance score( p = 0.005), metastasis time (de novo vs recurrent)(p = 0.008), platelet(p = 0.04), albumin(p = 0.026), lymphocyte count(p = 0.037), LIPI(p = 0.002) score and PIV(p = 0.001) were related with survival time. ( Table 2) (Fig. 1).

Table 1

Demographic, clinic and laboratory features of all patients.

   

All

n(%) = 109(100%)

mean ± SD

median( min-max)

LIPI(High)

n(%) = 55(100%)

mean ± SD

median( min-max)

LIPI(Low)

n(%) = 54(100%)

mean ± SD

median( min-max)

p

value

PIV (High)

n(%) = 55(100%)

mean ± SD

median( min-max)

PIV (Low)

n(%) = 54(100%)

mean ± SD

median( min-max)

P

value

Age

 

61.86 ± 9.97

62.13 ± 9.82

60.87 ± 10.69

0.593

59.59 ± 9.10

62.89 ± 1023

0.109

Sex

Male

55 (50.46%)

28 (50.90%)

27 (49.1%)

0.92

24 (43.4%)

31 (56.6%)

0.15

Female

54 (49.54%)

27 (50%)

27 (50%)

 

31 (57.4%)

23 (42.6%)

 

ECOG

≥ 3

53 (48.62%)

23 (43.4%)

30 (56.6%)

0.58

20 (37.74%)

33 (62.26%)

0.096

< 3

56 (51.38%)

22 (39.29%)

34 (60.71%)

 

35 (53.03%)

31 (46.97%)

 

Metastasis

Recurrent

32 (29.36%)

17 (53.13%)

15 (46.87%)

0.72

20 (62.5%)

12 (37.5%)

0.11

De novo

77 (70.64%)

38 (49.35%)

39 (50.64%)

 

35 (45.45%)

65 (54.55%)

 

Smoking

 

50 (10–150)

50 (10–150)

60 (30–120)

0.03

45 (30–150)

60 (10–120)

0.011

Metastassite

Liver

31 (28.4%)

6 (19.4%)

25 (80.6%)

0.118

7 (22.6%)

24 (77.4%)

0.12

 

Bone

53 (48.6%)

10 (18.9%)

43 (81.1%)

0.012

11 (20.75%)

42 (79.25%)

0.22

Adrenal

40 (29%)

6 (15%)

34 (85%)

0.008

11 (27.5%)

29 (72.5%)

0.52

Brain

48 (44%)

13 (27.1%)

35 (72.9%)

0.52

12 (25%)

36 (75%)

0.216

Lung

39 (35.8%)

10 (25.6%)

29 (74.4%)

0.43

11 (28.20%)

28 (71.80%)

0.61

LN

51 (46.8%)

13 (25.5%)

38 (74.5%)

0.308

16 (31.4%)

35 (68.6%)

0.97

Laboratory

Hb (g/dL)

12 ± 1.64

13.09 ± 1.56

12.73 ± 1.928

0.64

13.64 ± 1.57

12.64 ± 1.58

0.003

LDH(U/L)

336 (135–9800)

276 (135–1091)

698 (157–9800)

< 0.001

221(135–608)

365 (145–9800)

< 0.0001

Uric acid(mg/dL)

4.8 (2.5-9)

4.8 (2.5-9)

5.8 (2.9–7.7)

0.001

4.4 (2.8-9)

5 (2.5–7.7)

0.084

Albumin(g/dL)

3.7 (2.6-5.0)

3.8 (2.6–4.7)

3.4 (2.6-5)

0.003

4 (3-4.7)

3.6 (2.6-5)

< 0.0001

WBC(103/µL)

9.3 (4.3–20.3)

9.3 (4.3–20.3)

9.2 (5.8–14.5)

0.65

9.35 (4.7–20.3)

9.2 (4.3–16.2)

0.638

Neutrophil (103/µL)

7.0 (2.5–18.00)

6.8 (2.5–18.0)

7.8 (4.5–12.5)

0.61

6.85 (3.2–18.0)

7 (2.5–14)

0.132

Lymphocyte(103/µL)

1.5 (0.7–3.8)

1.6 (1.0-3.8)

1.2 (0.7–2.6)

< 0.0001

1.8 (1-3.8)

1.4 (0.7–2.7)

0.015

Platelet(103/µL)

300 (134–614)

282 (134–557)

334 (109–614)

0.004

282 (134–557)

324 (109–614)

0.005

SD: standart deviasion min: minumum, max(maximum), LIPI; Lung immun prognostic index, PIV; pan-immune-inflamation value, ECOG; Eastern Cooperative Oncology Groub, LN;Lymph node, Hb; Hemoglobin, LDH: lactate dehydrogenase, WBC; White Blood Cell,

Table 2

Univariate and multivariate analyses of overall survival

 

Univariate analysis

(HR, 95% CI)

p value

Multivariate analysis

(HR, 95% CI)

p value

Age

1.006(0.985–1.027)

0.571

   

Sex

1.051(0.653–1.692)

0.836

   

ECOG(< 3 vs 3≤)

2.178(1.438–3.293)

< 0.0001

1.937(1.165–3.219)

0.005

Smoking

1.012(1.005–1.020)

0.001

1.008(0.997–1.019)

0.174

Metastasis

(de novo vs recurrent)

2.885(1.913–4.361)

< 0.0001

1.797(1.094–2.952)

0.008

Liver metastas

1.856(1.200-2.872)

0.008

1.449(0.853–2.463)

0.170

Bone metastas

1.873(1.257–2.790)

0.002

1.352(0.840–2.177)

0.215

Adrenal metastas

2.178(1.438–3.299)

0.001

1.622(0.949–2.771)

0.77

Brain metastas

1.301(0.936–2.066)

0.102

   

Lung metastas

1.354(0895-2.047)

0.151

   

LN metastas

1.046(0.703–1.558)

0.825

   

Hb

-0.933(0.821–1.060)

0.286

   

LDH

1.000(1.00-1.001

0.001

1.000(1.000–1.000)

0.959

Uric acid

1.284(1.100-1.499)

0.002

1.000(0.870–1.365)

0.455

Albumin

-0.348(0.218–0.557)

< 0.0001

-0.607(0.379–0.970)

0.026

WBC

1.000(0.997–1.004)

0.607

   

Neutrophil

1.010(0.985–1.020)

0.259

   

Lymphocyte

-1.000 (1.000-1.010)

0.033

-1.000(0.999-1.000)

0.037

Platelet

1.004(1.001–1.006)

0.02

1.004(1.001–1.005)

0.04

LIPI

1.873(1.257–2.793)

< 0.0001

2.393(1.499–3.820)

0.002

PIV

2.662(1.731–4.095)

< 0.0001

2.143(1.332–3.452)

0.001

HR: hazard ratio, CI: confidence interval, ECOG; Eastern Cooperative Oncology Groub, LN;Lymph node, Hb; Hemoglobin, LDH: lactate dehydrogenase, WBC; White Blood Cell, LIPI; Lung immun prognostic index, PIV; pan-immune-inflamation value

Discussıon

This study was conducted to evaluate the relationship between PIV and LIPI score, survival time, and prognosis in patients with LCNECs. In our study, LIPI score, PIV, ECOG performance score, de novo metastasis, albumin level, and lymphocyte and platelet counts are associated with prognosis.

Inflammatory status and immune status play a key role in determining cancer formation, progression, invasion, metastasis, response to treatment, and survival [9]. In peripheral blood, biomarkers formed by the values of neutrophils, lymphocytes, monocytes, and platelets reflecting the circulating inflammatory and immune status have shown prognostically significant results [10].

LCNEC has an aggressive course but is rare and constitutes 2–3% of all lung cancers [1]. Because of its high grade, aggressive behavior, frequent recurrence after surgery, and positive neuroendocrine markers, it is similar to SCLC and has been classified among high-grade neuroendocrine cancers of the lung together with SCLC by the World Health Organization [36].

However, they are similar to NSCLC due to features such as being peripherally located, detecting mutations such as EGFR ALK RAF, etc., although rare, and patients benefiting from treatments targeting these mutations and immunotherapies such as nivolumab pembrolizumab in case-level reports [1115]. The most important prognostic marker in follow-up and treatment is the stage. However, the prognosis of patients with the same stages may be different. Therefore, there is a need for other prognostic markers beside the stage. Due to the rarity of the information with LCNEC, there is no controlled study, and it is obtained from retrospective data, case reports, or LCNEC adaptation of this information due to similarities between both SCLC and NSCLC. Many studies have been showing between many markers reflecting inflammation and immune status and the prognosis of SCLC, NSCLC, and other cancers. Therefore, prognostic markers reflecting inflammation and the immune system in NSCLC and SCLC may be useful in LCNEC patients.

LIPI score is an inflammatory marker that includes the neutrophil-to-lymphocyte ratio (dNLR) and serum lactate dehydrogenase (LDH). Significant results were obtained in studies on prognosis and survival in studies with LIPI scores in SCLC and NSCLC [1618]. LIPI score can predict prognosis as in SCLC and NSCLC. To our knowledge, there is no study in the literature showing the benefit of the LIPI score in predicting the prognosis of LCNEC.

Peripheral blood neutrophilia and relative lymphopenia before treatment are potential prognostic indicators of the tumor-related inflammatory response [19]. In studies conducted in SCLC and NSCLC, a high neutrophil-lymphocyte ratio was found to be associated with short progression-free survival and short overall survival [2021]. LDH is a prognostic marker in lung cancer [22]. Normal differentiated cells obtain energy through mitochondrial oxidative phosphorylation, while tumor cells rely on aerobic energy to support cell proliferation. This phenomenon is called the Warburg effect [23]. It promotes the expression and activation of glycolytic enzymes in cancer. LDH is one of them and plays a vital role in the glycolysis pathway process that can convert pyruvate to lactate [24]. High LDH levels are associated with poor prognosis in lung cancer, renal cell carcinoma, breast cancer, and colorectal cancer [2529]. Therefore, as the LIPI score is calculated using lymphocyte neutrophil and LDH values, it may be useful in predicting the prognosis in LCNEC

In previous studies, a relationship was found between LDH, lymphocytes, neutrophils, LIPI, and many cancers. However, the relationship between LIPI and prognosis in patients with LCNEC has not been investigated until now.. In our study, median OS was associated with LIPI scores. Our study is the first in the literature to demonstrate a relationship between LIPI and LCNEC prognosis.

PIV is an evaluation of inflammation and the immune system in cancer patients and is calculated according to neutrophil, platelet, monocyte, and lymphocyte levels [30]. There are many studies evaluating prognosis, treatment response, and survival in colorectal, skin, lung, breast, kidney, esophagus, and Merkel cell cancers [3135]. In studies conducted in SCLC and NSCLC, a relationship was found between high PIV values and short survival [32, 36]. There is no study conducted on PIV in patients with LCNEC.

Cancer cells and platelets have important interactions in circulation [37]. It has been found that platelets may play a key role in tumor growth and metastasis [38].To escape the immune system effect, tumor cells form a thrombus with Platelets. In addition, activated platelets form various growth factors that help tumor invasion and development [38]. Monocytes are associated with cancer prognosis, and after affecting circulating macrophages, they affect angiogenesis, invasion, and immunosuppression due to the release of endothelial growth factor (VEGF), tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-10 [3940]. Neutrophils are associated with tumor growth by the generation of reactive oxygen species and secretion of pre-tumor IL 1 and IL 6 tumor necrotic factor alpha [4142]. Lymphocytes are involved in the basic defense against cancer [43]. So PIV may predict a prognosis similar to SCLC and NSCLC.

Earlier studies have established a relationship between PIV and many cancers. However, the relationship between PIV and prognosis in patients with LCNEC has not been examined. In our study, median OS was linked to PIV. Our research is the first in the literature to show a link between PIV and the LCNEC prognosis.

Despite limitations, such as the retrospective and single-center design, and the small sample size, our study is important in that it is the first to show prognostic factors in LCNEC with LIPI score and PIV

In conclusion, due to their rarity, there is no randomized clinical study, standard follow-up, and treatment for LCNECs. In LCNEC patients, LIPI and PIV are simply relevant parameters that demonstrate inflammation and the immune system and can guide treatment selection and patient prognosis. LIPI, PIV, ECOG performance score, de novo metastases, albumin levels, lymphocytes, and platelet counts are related to prognosis. These factors can be used for patient monitoring and treatment as simple and inexpensive biomarkers.

Declarations

Author contributions

All authors contributed to the study's conception and design. Concept: S.M., Design: S.M , Supervision: S.M, Resources: S.M. and E.K., Materials S.M and E.K., Data Collection and/or Processing: S.M and E.K., Analysis and/or Interpretation: E.K Literature Searc: S.M and E.K. The first draft of the manuscript was written by S.M. and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Funding

The authors declare that they did not receive any funding for this study.

Competing interests

The authors declare no competing interests.

Data Availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Ethics Approval

The study was conducted by the principles of the Declaration of Helsinki and reviewed and approved by the Health Sciences Ethics Committee of Manisa Celal Bayar University (decision number: 10.478.486, date: 05/02/2020). 

References

  1. Fasano, M., Della Corte, C.M., Papaccio, F., Ciardiello, F., Morgillo, F. Pulmonary Large-Cell Neuroendocrine Carcinoma: From Epidemiology to Therapy. J Thorac Oncol. 10(8):1133–41 (2015).
  2. Travis, W.D., et all. Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol. 15(6):529 – 53 (1991).
  3. Battafarano, R.J., et all. Large cell neuroendocrine carcinoma: an aggressive form of non-small cell lung cancer. J Thorac Cardiovasc Surg. 130(1):166 – 72 (2005).
  4. Dresler, C.M., Ritter, J.H., Patterson, G.A., Ross, E., Bailey, M.S., Wick, M.R. Clinical-pathologic analysis of 40 patients with large cell neuroendocrine carcinoma of the lung. Ann Thorac Surg.63(1):180–5 (1997).
  5. Travis W.D., et all. Survival analysis of 200 pulmonary neuroendocrine tumors with clarification of criteria for atypical carcinoid and its separation from typical carcinoid. Am J Surg Pathol. 22(8):934 – 44 (1998).
  6. Travis W.D., et all. WHO Panel. The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J Thorac Oncol. 10(9):1243–1260 (2015).
  7. Wang D., et all. Prognostic Nutritional Index and Systemic Immune-Inflammation Index Predict the Prognosis of Patients with HCC. J Gastrointest Surg.;25(2):421–427 (2021).
  8. Liu J., et all. Systemic immune-inflammation index, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio can predict clinical outcomes in patients with metastatic non-small-cell lung cancer treated with nivolumab. J Clin Lab Anal. Oct;33(8):e22964 (2019).
  9. Grivennikov, S.I., Greten, F.R., Karin, M. Immunity, inflammation, and cancer. Cell.19;140(6):883 – 99 (2010).
  10. Gao, Y., Zhang, H., Li, Y., Wang, D., Ma, Y., Chen, Q. Preoperative increased systemic immune-inflammation index predicts poor prognosis in patients with operable non-small cell lung cancer. Clin Chim Acta. 484:272–277 (2018).
  11. Fu, Z., Zhu, G., Wang, L., Hu, S., Cheng, L., Liu, F. Case Report: A Pregnant Woman Diagnosed as ALK-Rearrangement Lung Large Cell Neuroendocrine Cancer With Brain Metastasis. Front Oncol. 25;12:823813 (2022).
  12. Kadota, N., et all. Complete and durable response of pulmonary large-cell neuroendocrine carcinoma to pembrolizumab. Cancer Rep (Hoboken). 5(8):e1589 (2022).
  13. Levra, M.G., et all. Efficacy of Immune Checkpoint Inhibitors in Large Cell Neuroendocrine Lung Cancer: Results from a French Retrospective Cohort. J. Thorac. Oncol. 12:702–703 (2017).
  14. Muto, S., et all. Successful Treatment of Combined Large Cell Neuroendocrine Carcinoma Harboring an EGFR Mutation with EGFR-TKIs plus Bevacizumab: A Case Report. Case Rep Oncol. 30;13(3):1387–1392 (2020).
  15. Sakamoto, T., Arai, K., Makishima, K., Yamasaki, A. BRAF V600E-mutated combined large cell neuroendocrine carcinoma and adenocarcinoma responding to targeted therapy. BMJ Case Rep. 30;14(12):e243295 (2021).
  16. Mezquita, L., et all. Association of the Lung Immune Prognostic Index With Immune Checkpoint Inhibitor Outcomes in Patients With Advanced Non-Small Cell Lung Cancer. JAMA Oncol. 1;4(3):351–357 (2018).
  17. Sonehara, K., Tateishi, K., Komatsu, M., Yamamoto, H., Hanaoka, M. Lung immune prognostic index as a prognostic factor in patients with small cell lung cancer. Thorac Cancer. 11(6):1578–1586 (2020).
  18. Minami, S., Ihara, S., Komuta, K. Pretreatment Lung Immune Prognostic Index Is a Prognostic Marker of Chemotherapy and Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor. World J Oncol. 10(1):35–45 (2019).
  19. Bonomi, M., Patsias, A., Posner, M., Sikora, A. The role of inflammation in head and neck cancer. Adv Exp Med Biol. 816:107–27 (2014).
  20. Wang, Z., et all. Prognostic role of pretreatment neutrophil-to-lymphocyte ratio in non-small cell lung cancer patients treated with systemic therapy: a meta-analysis. Transl Lung Cancer Res. 8(3):214–226 (2019).
  21. Yu, Y., Qian, L., Cui, J.Value of neutrophil-to-lymphocyte ratio for predicting lung cancer prognosis: A meta-analysis of 7,219 patients. Mol Clin Oncol;7(3):498–506 (2017).
  22. Zhang, X. et all. Prognostic significance of serum LDH in small cell lung cancer: A systematic review with meta-analysis. Cancer Biomark. 16(3):415 – 23 8 (2016).
  23. Vander Heiden, M.G., Cantley L.C., Thompson C.B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. May 22;324(5930):1029-33 (2019).
  24. Doherty, J.R., Cleveland, J.L. Targeting lactate metabolism for cancer therapeutics. J Clin Invest.123(9):3685–92 (2013).
  25. Armstrong, A.J., George, D.J., Halabi, S. Serum lactate dehydrogenase predicts for overall survival benefit in patients with metastatic renal cell carcinoma treated with inhibition of mammalian target of rapamycin. J Clin Oncol. 20;30(27):3402–7 (2012).
  26. Brown, J.E., Cook, R.J., Lipton, A., Coleman, R.E. Serum lactate dehydrogenase is prognostic for survival in patients with bone metastases from breast cancer: a retrospective analysis in bisphosphonate-treated patients. Clin Cancer Res.15;18(22):6348–55 (2012).
  27. Koukourakis, M.I., et all. Prognostic and predictive role of lactate dehydrogenase 5 expression in colorectal cancer patients treated with PTK787/ZK 222584 (vatalanib) antiangiogenic therapy. Clin Cancer Res. Jul 15;17(14):4892 – 900 (2011).
  28. Zhu, L., et all. A new prognostic score based on the systemic inflammatory response in patients with inoperable non-small-cell lung cancer. Onco Targets Ther. 8;9:4879-86 (2016).
  29. Liu, X., et all. The prognostic value of the serum neuron specific enolase and lactate dehydrogenase in small cell lung cancer patients receiving first-line platinum-based chemotherapy. Medicine (Baltimore). Nov;96(46):e8258 (2017).
  30. Fucà, G., et all. The Pan-Immune-Inflammation Value is a new prognostic biomarker in metastatic colorectal cancer: results from a pooled-analysis of the Valentino and TRIBE first-line trials. Br J Cancer. 123(3):403–409 (2020).
  31. Baba, Y., et all. Pan-immune-inflammation Value and Prognosis in Patients With Esophageal Cancer. Annals of Surgery Open. 3;1 - p e113 (2022).
  32. Chen, X., et all. The Pan-Immune-Inflammation Value predicts the survival of patients with anaplastic lymphoma kinase-positive non-small cell lung cancer treated with first-line ALK inhibitor. Transl Oncol. 6;17:101338 (2022).
  33. Fucà, G., et all. The Pan-Immune-Inflammation Value in Patients with Metastatic Melanoma Receiving First-Line Therapy. Target Oncol.(4):529–536 (2021).
  34. Gambichler, T., et all. Pan-immune-inflammation value independently predicts disease recurrence in patients with Merkel cell carcinoma. J Cancer Res Clin Oncol. 148(11):3183–3189 (2022).
  35. Ligorio, F., et all.The Pan-Immune-Inflammation-Value Predicts the Survival of Patients with Human Epidermal Growth Factor Receptor 2 (HER2)-Positive Advanced Breast Cancer Treated with First-Line Taxane-Trastuzumab-Pertuzumab. Cancers (Basel). 19;13(8):1964 (2021).
  36. Zeng, R., et all. PIV and PILE Score at Baseline Predict Clinical Outcome of Anti-PD-1/PD-L1 Inhibitor Combined With Chemotherapy in Extensive-Stage Small Cell Lung Cancer Patients. Front Immunol.29;12:724443(2021).
  37. Stone, R.L., Nick, A.M., McNeish, I.A., Balkwill, F., Han, H.D., Bottsford-Miller, J., et all. Paraneoplastic thrombocytosis in ovarian cancer. N Engl J Med. 16;366(7):610–8 (2012).
  38. Haemmerle, M., Taylor, M.L., Gutschner, T., Pradeep, S., Cho, M.S., Sheng, J., et all. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat Commun. 21;8(1):310 (2017).
  39. Petty, A.J., Yang, Y. Tumor-associated macrophages: implications in cancer immunotherapy. Immunotherapy. Mar;9(3):289–302 (2017).
  40. Shibutani, M., et all. The peripheral monocyte count is associated with the density of tumor-associated macrophages in the tumor microenvironment of colorectal cancer: a retrospective study. BMC Cancer. 5;17(1):404 (2017).
  41. Dumitru, C.A., Lang, S., Brandau, S. Modulation of neutrophil granulocytes in the tumor microenvironment: mechanisms and consequences for tumor progression. Semin Cancer Biol. 23(3):141–8 (2013).
  42. Deryugina, E.I., Zajac, E., Juncker-Jensen, A., Kupriyanova, T.A., Welter. L., Quigley, J.P. Tissue-infiltrating neutrophils constitute the major in vivo source of angiogenesis-inducing MMP-9 in the tumor microenvironment. Neoplasia. 23;16(10):771 – 88 (2014).
  43. Galli, F., Aguilera, J.V., Palermo, B., Markovic, S.N., Nisticò, P., Signore, A. Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. J Exp Clin Cancer Res.18;39(1):89 (2020).