リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Integrated analysis of tertiary lymphoid structures in relation to tumor-infiltrating lymphocytes and patient survival in pancreatic ductal adenocarcinoma」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Integrated analysis of tertiary lymphoid structures in relation to tumor-infiltrating lymphocytes and patient survival in pancreatic ductal adenocarcinoma

Tanaka, Takeshi Masuda, Atsuhiro Inoue, Jun Hamada, Tsuyoshi Ikegawa, Takuya Toyama, Hirochika Sofue, Keitaro Shiomi, Hideyuki Sakai, Arata Kobayashi, Takashi Tanaka, Shunta Nakano, Ryota Yamada, Yasutaka Ashina, Shigeto Tsujimae, Masahiro Yamakawa, Kohei Abe, Shohei Gonda, Masanori Masuda, Shigeto Inomata, Noriko Uemura, Hisahiro Kohashi, Shinya Nagao, Kae Kanzawa, Maki Itoh, Tomoo Ueda, Yoshihide Fukumoto, Takumi Kodama, Yuzo 神戸大学

2023.03

概要

Background Tertiary lymphoid structure (TLS) reflects an intense immune response against cancer, which correlates with favorable patient survival. However, the association of TLS with tumor-infiltrating lymphocytes (TILs) and clinical outcomes has not been investigated comprehensively in pancreatic ductal adenocarcinoma (PDAC). Methods We utilized an integrative molecular pathological epidemiology database on 162 cases with resected PDAC, and examined TLS in relation to levels of TILs, patient survival, and treatment response. In whole-section slides, we assessed the formation of TLS and conducted immunohistochemistry for tumor-infiltrating T cells (CD4, CD8, CD45RO, and FOXP3). As confounding factors, we assessed alterations of four main driver genes (KRAS, TP53, CDKN2A [p16], and SMAD4) using next-generation sequencing and immunohistochemistry, and tumor CD274 (PD-L1) expression assessed by immunohistochemistry. Results TLSs were found in 112 patients with PDAC (69.1%). TLS was associated with high levels of CD4⁺ TILs (multivariable odds ratio [OR], 3.50; 95% confidence interval [CI] 1.65–7.80; P = 0.0002), CD8⁺ TILs (multivariable OR, 11.0; 95% CI 4.57–29.7, P < 0.0001) and CD45RO⁺ TILs (multivariable OR, 2.65; 95% CI 1.25–5.80, P = 0.01), but not with levels of FOXP3⁺ TILs. TLS was associated with longer pancreatic cancer-specific survival (multivariable hazard ratio, 0.37; 95% CI 0.25–0.56, P < 0.0001) and favorable outcomes of adjuvant S-1-treatment. TLS was not associated with driver gene alterations but tumor CD274 negative expression. Conclusions Our comprehensive data supports the surrogacy of TLS for vigorous anti-tumor immune response characterized by high levels of helper and cytotoxic T cells and their prognostic role.

この論文で使われている画像

参考文献

1. Mizrahi JD, Surana R, Valle JW, et al. Pancreatic cancer. Lancet

2020;395:2008–20.

2. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and

metastasis. Nat Med 2013;19:1423-37.

3. Ziani L, Chouaib S, Thiery J. Alteration of the Antitumor Immune Response by

Cancer-Associated Fibroblasts. Front Immunol 2018;9:414.

4. Foucher ED, Ghigo C, Chouaib S, et al. Pancreatic ductal adenocarcinoma: A

strong imbalance of Good and bad immunological cops in the tumor

microenvironment. Front Immunol 2018;9:1044.

5. Ino Y, Yamazaki-Itoh R, Shimada K, et al. Immune cell infiltration as an indicator

of the immune microenvironment of pancreatic cancer. Br J Cancer

2013;108:914–23.

6. Yang JJ, Hu ZG, Shi WX, et al. Prognostic significance of neutrophil to

lymphocyte ratio in pancreatic cancer: a meta-analysis. World J Gastroenterol

2015;21:2807–15.

7. Colbeck EJ, Ager A, Gallimore A, et al. Tertiary Lymphoid Structures in Cancer:

drivers of antitumor Immunity, immunosuppression, or Bystander Sentinels in

37

Disease? Front Immunol 2017;8:1830.

8. Dieu-Nosjean MC, Giraldo NA, Kaplon H, et al. Tertiary lymphoid structures,

drivers of the anti-tumor responses in human cancers. Immunol Rev

2016;271:260–75.

9. Ager A. High endothelial venules and other blood vessels: critical regulators of

lymphoid organ development and function. Front Immunol 2017;8:45.

10. Martinet L, Garrido I, Filleron T, et al. Human solid tumors contain high

endothelial venules: association with T− and B- lymphocyte infiltration and

favorable prognosis in breast cancer. Cancer Res 2011;71:5678–87.

11. Lee HJ, Kim JY, Park IA, et al. Prognostic significance of tumor-infiltrating

lymphocytes and the tertiary lymphoid structures in HER2-positive breast

cancer treated with adjuvant trastuzumab. Am J Clin Pathol 2015;144:278–

88.

12.Germain C, Gnjatic S, Tamzalit F, et al. Presence of B cells in tertiary lymphoid

structures is associated with a protective immunity in patients with lung cancer.

Am J Respir Crit Care Med 2014;189:832–44.

13. Goc J, Germain C, Vo-Bourgais TKD, et al. Dendritic cells in tumorassociated tertiary lymphoid structures signal a Th1 cytotoxic immune

38

contexture and license the positive prognostic value of infiltrating CD8+T cells.

Cancer Res 2014;74:705–15.

14. Di Caro GD, Bergomas F, Grizzi F, et al. Occurrence of tertiary lymphoid tissue

is associated with T-cell infiltration and predicts better prognosis in early-stage

colorectal cancers. Clin Cancer Res 2014;20:2147–58.

15. Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and

recurrence of stage II and III colorectal cancer. Oncoimmunology

2018;7:e1378844.

16. Hiraoka N, Ino Y, Yamazaki-Itoh R, et al. Intratumoral tertiary lymphoid organ

is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer

2015;112:1782–90.

17. Kuwabara S, Tsuchikawa T, Nakamura T, et al. Prognostic relevance of

tertiary lymphoid organs following neoadjuvant chemoradiotherapy in

pancreatic ductal adenocarcinoma. Cancer Sci 2019;110:1853–62.

18. Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational

landscape of pancreatic cancer. Nature 2015;518:495–501.

19. Qian ZR, Rubinson DA, Nowak JA, et al. Association of alterations in main

driver genes with outcomes of patients with resected pancreatic ductal

39

adenocarcinoma. JAMA Oncol 2018;4:e173420.

20. Chen H, Chong W, Teng C, et al. The immune response-related mutational

signatures and driver genes in non-small-cell lung cancer. Cancer Sci

2019;110:2348–56.

21. Luen S, Virassamy B, Savas P, et al. The genomic landscape of breast cancer

and its interaction with host immunity. Breast 2016;29:241–50.

22. Giannakis M, Mu XJ, Shukla SA, et al. Genomic correlates of immune-cell

infiltrates in colorectal carcinoma. Cell Rep 2016;15:857–65.

23. Jiang X, Wang J, Deng X, et al. Role of the tumor microenvironment in PDL1/PD-1-mediated tumor immune escape. Mol Cancer 2019;18:10.

24. Iwatate Y, Hoshino I, Yokota H, et al. Radiogenomics for predicting p53 status,

PD-L1 expression, and prognosis with machine learning in pancreatic cancer.

Br J Cancer 2020;123:1253–61.

25. Petrackova A, Vasinek M, Sedlarikova L, et al. Standardization of sequencing

coverage depth in NGS: recommendation for detection of clonal and

subclonal mutations in Cancer Diagnostics. Front Oncol 2019;9:851.

26. Cancer Genome Atlas Research Network. Electronic address:andrew_a

guirre@dfci. harvard.edu, Cancer Genome Atlas Research Network.

40

Integrated genomic characterization of pancreatic ductal adenocarcinoma.

Cancer Cell 2017;32:185–203.e13.

27. Yang SR, Lin CY, Stehr H, et al. Comprehensive genomic profiling of

malignant effusions in patients with metastatic lung adenocarcinoma. J Mol

Diagn 2018;20:184–94.

28. Suzuki H, Aoki K, Chiba K, et al. Mutational landscape and clonal

architecture in grade II and III gliomas. Nat Genet 2015;47:458–68.

29. Mazaika E, Homsy J. Digital droplet PCR: CNV analysis and other

applications. Curr Protoc Hum Genet 2014;82:7.24.1–13.

30. Lassaletta A, Zapotocky M, Mistry M, et al. Therapeutic and prognostic

implications of BRAF V600E in pediatric low-grade gliomas. J Clin Oncol

2017;35:2934–41.

31. Tsujimae M, Masuda A, Ikegawa T, et al. Comprehensive Analysis of

Molecular Biologic Characteristics of Pancreatic Ductal Adenocarcinoma

Concomitant with Intraductal Papillary Mucinous Neoplasm. Ann Surg Oncol

2022.

32. Yachida S, White CM, Naito Y, et al. Clinical significance of the genetic

landscape of pancreatic cancer and implications for identification of potential

41

long-term survivors. Clin Cancer Res 2012;18:6339–47.

33. Ohtsubo K, Watanabe H, Yamaguchi Y, et al. Abnormalities of tumor

suppressor gene p16 in pancreatic carcinoma: immunohistochemical and

genetic

findings

compared

with

clinicopathological

parameters.

Gastroenterol 2003;38:663–71.

34. Tascilar M, Skinner HG, Rosty C, et al. The SMAD4 protein and prognosis of

pancreatic ductal adenocarcinoma. Clin Cancer Res 2001;7:4115–21.

35. Ying S, Humbert M, Barkans J, et al. Expression of IL-4 and IL-5 mRNA and

protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in

bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics.

J Immunol. 1997;158:3539-44.

36. Vonderheide RH. The Immune Revolution: A Case for Priming, Not

Checkpoint. Cancer Cell. 2018;33:563-9.

37. Foucher ED, Ghigo C, Chouaib S, et al. Pancreatic Ductal Adenocarcinoma:

A Strong Imbalance of Good and Bad Immunological Cops in the Tumor

Microenvironment. Front Immunol. 2018;9:1044.

38. Zitvogel L, Galluzzi L, Kepp O, et al. Type I interferons in anticancer immunity.

Nat Rev Immunol. 2015;15:405-14.

42

39. Hotta K, Sho M, Fujimoto K, et al. Prognostic significance of CD45RO+

memory T cells in renal cell carcinoma. Br J Cancer. 2011;105:1191-6.

40. Tan AH, Goh SY, Wong SC, et al. T helper cell-specific regulation of inducible

costimulator expression via distinct mechanisms mediated by T-bet and

GATA-3. J Biol Chem. 2008;283:128-136.

41. Masugi Y, Abe T, Ueno A, et al. Characterization of spatial distribution of

tumor-infiltrating CD8 + T cells refines their prognostic utility for pancreatic

cancer survival. Mod Pathol. 2019;32:1495-1507.

42. Takada K, Kashiwagi S, Asano Y, et al. Clinical verification of the relationship

between smoking and the immune microenvironment of breast cancer. J

Transl Med. 2019;17:13.

43. Liu C, Xu B, Li Q, et al. Smoking history influences the prognostic value of

peripheral naïve CD4+ T cells in advanced non-small cell lung cancer. Cancer

Cell Int. 2019;19:176.

44. Pineda S, Maturana EL, Yu K, et al. Tumor-Infiltrating B- and T-Cell

Repertoire in Pancreatic Cancer Associated With Host and Tumor Features.

Front Immunol.2021;12:730746.

45. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune

43

set point. Nature 2017;541:321–30.

46. Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 promotes

T-cell apoptosis: a potential mechanism of immune evasion. Nat Med

2002;8:793–800.

47. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity

cycle. Immunity 2013;39:1–10.

48. Mundry CS, Eberle KC, Singh PK, et al. Local and systemic

immunosuppression in pancreatic cancer: targeting the stalwarts in tumor’s

arsenal. Biochim Biophys Acta Rev Cancer 2020;1874:188387.

49. Sautès-Fridman C, Petitprez F, Calderaro J, et al. Tertiary lymphoid structures

in the era of cancer immunotherapy. Nat Rev Cancer 2019;19:307–25.

50. Farren MR, Mace TA, Geyer S, et al. Systemic immune activity predicts

overall survival in treatment-naïve patients with metastatic pancreatic cancer.

Clin Cancer Res 2016;22:2565–74.

51. Lee KH, Kim EY, Yun JS, et al. The prognostic and predictive value of tumorinfiltrating lymphocytes and hematologic parameters in patients with breast

cancer. BMC Cancer 2018;18:938.

52. Huang Y, Lou XY, Zhu YX, et al. Local environment in biopsy better predict the

44

pathological response to neoadjuvant chemoradiotherapy in rectal cancer.

Biosci Rep 2019;39.

53. Uesaka K, Boku N, Fukutomi A, et al. Adjuvant chemotherapy of S-1 versus

gemcitabine for resected pancreatic cancer: a phase 3, open-label,

randomised, non-inferiority trial (JASPAC 01). Lancet 2016;388:248–57.

54. Zitvogel L, Apetoh L, Ghiringhelli F, et al. Immunological aspects of cancer

chemotherapy. Nat Rev Immunol 2008;8:59–73.

55. Adachi Y, Taketani S, Oyaizu H, et al. Apoptosis of colorectal

adenocarcinoma induced by 5-FU and/or IFN-gamma through caspase 3 and

caspase 8. Int J Oncol. 1999;15:1191-6.

56. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer

Immunoediting. Nat Rev Immunol. 2006;6:836-48.

57. Kursunel MA, Esendagli G. The untold story of IFN-γ in cancer biology.

Cytokine Growth Factor Rev. 2016;31:73-81.

58. Choudhry H, Helmi N, Abdulaal WH, Prospects of IL-2 in Cancer

Immunotherapy. Biomed Res Int. 2018;9056173.

59. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy.

Science 2015;348:69–74.

45

60. Pu N, Chen Q, Gao S, Liu G, Zhu Y, Yin L, et al. Genetic landscape of

prognostic value in pancreatic ductal adenocarcinoma microenvironment. Ann

Transl Med 2019;7:645.

61. Wartenberg M, Cibin S, Zlobec I, et al. Integrated genomic and

immunophenotypic classification of pancreatic cancer reveals three distinct

subtypes

with

prognostic/predictive

significance.

Clin

Cancer

Res

2018;24:4444–54.

62. Cheng H, Fan K, Luo G, et al. Kras G12D mutation contributes to regulatory

T cell conversion through activation of the MEK/ERK pathway in pancreatic

cancer. Cancer Lett. 2019;446:103-111.

46

Figure legends

Fig. 1 Characterization of TLS in PDAC. a Representative images of H&E-stained

sections were obtained from resected PDAC. The dotted line indicates the margin

of the tumor, and the black arrow indicates the TLS beside the tumor (left panel).

TLS within the tumor is shown, and black arrowheads indicate cancer cells (right

panel). b & c Components and proportions of immune cells within TLS. In the

IHC analysis, the TLSs were composed of T cells (CD4, CD8, CD45RO, and

FOXP3), B cells (CD20), and macrophages (CD68). d The association between

the presence of TLS and tumor characteristics (pTNM stage, pT stage, and

histological grade). Statistical significance was set at P < 0.05. Original

magnification (a, 40×; b, 200×). Scale bars, 200 μm (a); 50 μm (b). H&E,

haematoxylin and eosin; IHC, immunohistochemistry; PDAC, pancreatic ductal

adenocarcinoma; TLS, tertiary lymphoid structure

Fig. 2 CSS of the PDAC patients according to the presence or absence of TLS.

The CSS of PDAC patients according to the presence or absence of TLS was

investigated. The differences of the CSS in adjuvant chemotherapy among the

groups treated with S-1, gemcitabine, and non-treated groups according to the

47

presence or absence of TLS were also examined. a All patients b TLS-present,

and c TLS-absent patients. The CSS curves were estimated using the KaplanMeier method, and the significant differences between the two groups were

evaluated using a log-rank test. The number of patients at risk is shown in the

CSS curves. CSS, cancer-specific survival; PDAC, pancreatic ductal

adenocarcinoma; TLS, tertiary lymphoid structure

Supplementary Fig. 1 Representative immunohistochemistry images. a Typical

images of high or low levels of CD4+, CD8+, CD45RO+ and FOXP3+ TILs. b

Typical images of presence or absence of CD274 (PD-L1) expression. c Typical

images of intact or altered (lost or over expression) of protein expression of TP53,

CDKN2A and SMAD4. Original magnification (a-c, 200×). Scale bars, 50 μm (ac). PDL1, Programmed death-ligand 1; TILs, tumor infiltrating lymphocytes

Supplementary Fig. 2 CSS of PDAC patients according to the intratumoral TLS

and peritumoral TLS. Intratumoral TLSs were detected within and around the

tumor; peritumoral TLSs were detected only around the tumor. The CSS curves

were estimated using the Kaplan-Meier method, and the significant differences

48

between the two groups were evaluated using a log-rank test. The number of

patients at risk is shown in the CSS curves. CSS, cancer-specific survival; PDAC,

pancreatic ductal adenocarcinoma; TLS, tertiary lymphoid structure

Supplementary Fig. 3 CSS of PDAC patients according to the level of NLR or

extent of CD8+ TILs. The CSS of PDAC patients according to high or low NLR

were investigated. The differences of the CSS in adjuvant chemotherapy among

the groups treated with S-1, gemcitabine, and non-treated groups according to

the high or low NLR were also examined. a All patients b Patients with low

NLR c Patients with high NLR. The CSS of PDAC according to the high or low

levels of CD8+ TILs were investigated. The differences of the CSS in adjuvant

chemotherapy among the groups treated with S-1, gemcitabine, and non-treated

groups according to the high or low levels of CD8+ TILs were also examined. d

All patients e Patients with high levels of CD8+ TILs f Patients with low levels

of CD8+ TILs. The CSS curves were estimated using the Kaplan-Meier method,

and the significant differences between the two groups were evaluated by a logrank test. The number of patients at risk is shown in the CSS curves. CSS,

cancer-specific survival; NLR, neutrophil-to lymphocyte ratio; PDAC, pancreatic

49

ductal adenocarcinoma; TILs, tumor infiltrating lymphocytes

Supplementary Fig. 4 In situ hybridization for IFN-gamma and IL-2, and

apoptosis assay. a Typical images of IFN-gamma and IL-2 expression in TLSpresent and absent patients. IFN-gamma-positive cells and IL-2-positive

cells show distinct red staining. b The expression levels of IFN-gamma or IL-2

per cm2 in TLS-present patients (n=5) and TLS-absent patients (n=5) were

counted. Mean ± SD (bars). *, P < 0.05. c Double immunohistochemistry-in situ

hybridization. To distinguish between IHC of CD4+ and CD8+ T cells (brown) and

ISH of IFN-gamma (pink) more clearly, brown is changed to green using color

replacement tools of Photoshop. IFN-gamma mRNA (pink arrowhead) was

localized to CD4+ and CD8+ T cells (green). d TUNEL assays were performed 7

days after the addition of IFN-gamma (100 units/mL), IL-2 (500 units/ml), 5-FU

(0.05 mg/mL), and their combination to detect the apoptotic cells. TUNEL analysis

showed a significant difference in cell numbers at day 7 between the combination

therapy group and other treatment groups. Mean ± SD (bars). **, P < 0.001, ***,

P < 0.01. Original magnification (a, c, 400×). Scale bars, 20 μm. IFN, interferon;

IL, interleukin; PDAC, pancreatic ductal adenocarcinoma; SD, standard

50

deviation; TLS, tertiary lymphoid structure

51

Table 1. Patient’s characteristics according to presence or absence of TLS in PDAC

Characteristics

Age (years), Median (range)

BMI (kg/m2), Median (range)

Sex

Male

Female

Family history of PDAC

Present

Absent

Alcohol consumption

< 50 (g/day)

≥ 50 (g/day)

History of smoking

Present

Absent

Diabetes mellitus

Present

Absent

CA19-9

< 37 (U/ml)

≥ 37 (U/ml)

CEA

< 5 (ng/ml)

≥ 5 (ng/ml)

Pathological Stagea

Ia/Ib

IIa/IIb

III

Neoadjuvant chemotherapy

Present

Absent

Adjuvant chemotherapy

Present

Absent

Histology grade

Well/Moderate

Poorly

Residual tumor status

R0

R1

KRAS mutation

Present

Absent

TP53 alteration

Present

Absent

CDKN2A alteration

Present

Absent

SMAD4 alteration

Present

Absent

CD274 expression

Positive

Negative

All patients (N=162)

69 (40-85)

21.0 (14.3-33.2)

Present (N=112)

70 (40-85)

21.0 (14.3-32.3)

TLS

Absent (N=50)

69 (48-82)

20.7 (16.5-33.2)

90 (55.6%)

72 (44.4%)

64 (57.1%)

48 (42.9%)

26 (52.0%)

24 (48.0%)

14 (8.6%)

148 (91.4%)

11 (9.8%)

101 (90.2%)

3 (6.0%)

47 (94.0%)

146 (90.1%)

16 (9.9%)

98 (87.5%)

14 (12.5%)

48 (96.0%)

2 (4.0%)

78 (48.1%)

84 (51.9%)

61 (54.5%)

51 (45.5%)

17 (34.0%)

33 (66.0%)

64 (39.5%)

98 (60.5%)

44 (39.3%)

68 (60.7%)

20 (40.0%)

30 (60.0%)

40 (24.7%)

122 (75.3%)

34 (30.4%)

78 (69.6%)

6 (12.0%)

44 (88.0%)

116 (71.6%)

46 (28.4%)

79 (70.5%)

33 (29.5%)

37 (74.0%)

13 (26.0%)

35 (21.6%)

89 (54.9%)

38 (23.5%)

27 (24.1%)

62 (55.4%)

23 (20.5%)

8 (16.0%)

27 (54.0%)

15 (30.0%)

18 (11.1%)

144 (88.9%)

13 (11.6%)

99 (88.4%)

5 (10.0%)

45 (90.0%)

117 (72.2%)

45 (27.8%)

83 (74.1%)

29 (25.9%)

34 (68.0%)

16 (32.0%)

143 (88.3%)

19 (11.7%)

99 (88.4%)

13 (11.6%)

44 (88.0%)

6 (12.0%)

118 (72.8%)

44 (27.2%)

84 (75.0%)

28 (25.0%)

34 (68.0%)

16 (32.0%)

150 (92.6%)

12 (7.4%)

104 (92.9%)

8 (7.1%)

46 (92.0%)

4 (8.0%)

111 (68.5%)

51 (31.5%)

76 (67.9%)

36 (32.1%)

35 (70.0%)

15 (30.0%)

104 (64.2%)

58 (35.8%)

68 (60.7%)

44 (39.3%)

36 (72.0%)

14 (28.0%)

62 (38.3%)

100 (61.7%)

39 (34.8%)

73 (65.2%)

23 (46.0%)

27 (54.0%)

23 (14.2%)

139 (85.8%)

9 (8.0%)

103 (92.0%)

14 (28.0%)

36 (72.0%)

P value

0.66

0.92

0.61

0.55

0.15

0.02

0.99

0.02

0.71

0.30

0.99

0.45

0.99

0.44

0.99

0.86

0.21

0.22

0.002

(%) indicates the proportion of cases with a specific clinical, pathological, or tumor

molecular characteristic according to the presence or absence of TLS.

a Pathological stage was diagnosed based on TNM Classification of Malignant Tumors,

8th Edition. BMI, body mass index; CA19-9, carbohydrate antigen 19-9; CEA,

carcinoembryonic antigen; PDAC, pancreatic ductal adenocarcinoma; PD-L1,

programmed cell death ligand-1; TLS, tertiary lymphoid structure.

Table 2. The association of TLS with TILs in PDAC

No. of

Cases

TLS

No. of cases

with high levels of

CD4+ TILs

162

80 (49.4%)

Absence

50

14 (28.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

66 (58.9%)

3.69 (1.82-7.80)

4.21 (1.93-9.74)

3.50 (1.65-7.80)

0.0002

0.0002

0.001

P value

TLS

No. of

Cases

162

Absence

50

9 (18.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

72 (64.3%)

8.20 (3.76-19.6)

9.15 (3.98-23.1)

11.0 (4.57-29.7)

<0.0001

<0.0001

<0.0001

High levels of CD45RO+ TILs

Univariate

Multivariate

Multivariate

OR (95%CI)

ORa (95%CI)

ORb (95%CI)

No. of

Cases

162

No. of cases

with high levels of

CD45RO+ TILs

81 (50.0%)

Absence

50

17 (34.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

64 (57.1%)

2.59 (1.31-5.27)

2.51 (1.19-5.46)

2.65 (1.25-5.80)

0.006

0.02

0.01

P value

TLS

High levels of CD8+ TILs

Univariate

Multivariate

Multivariate

OR (95%CI)

ORa (95%CI)

ORb (95%CI)

No. of cases

with high levels of

CD8+ TILs

81 (50.0%)

P value

TLS

High levels of CD4+ TILs

Univariate

Multivariate

Multivariate

OR (95%CI)

ORa (95%CI)

ORb (95%CI)

High levels of FOXP3+ TILs

Univariate

Multivariate

Multivariate

OR (95%CI)

ORa (95%CI)

ORb (95%CI)

No. of

Cases

162

No. of cases

with high levels of

FOXP3+ TILs

76 (46.9%)

Absence

50

23 (46.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

53 (47.3%)

1.05 (0.54-2.07)

0.86 (0.40-1.81)

0.80 (0.45-1.83)

0.88

0.68

0.80

P value

Lymphocyte counts were divided into high and low group by the median split. The median of CD4+,

CD8+, CD45RO+ and FOXP3+ T cells are 48, 140, 110 and 33 count/mm2, respectively.

a The odds ratio was initially adjusted for age, sex, BMI, amount of alcohol consumption, family

history of pancreatic cancer, history of smoking, diabetes mellitus, serum CA19-9, serum CEA.

b The odds ratio was initially adjusted for stage, histology, adjuvant, neo-adjuvant, residual tumor

status, gene alterations (KRAS, TP53, CDKN2A, SMAD4).

OR, odds ratio; PDAC, pancreatic ductal adenocarcinoma; TILs, tumor-infiltrating lymphocytes; TLS,

tertiary lymphoid structure.

Table 3. The association of TLS with serum ALC, ANC and NLR in PDAC

TLS

No. of cases with

high levels of ALC

81 (50.0%)

Univariate

OR (95%CI)

Absence

50

17 (34.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

64 (57.1%)

2.59 (1.31-5.27)

2.84 (1.33-6.28)

2.46 (1.21-5.12)

0.006

0.007

0.01

P value

TLS

High levels of serum ANC

Univariate

Multivariate

Multivariate

OR (95%CI)

ORa (95%CI)

ORb (95%CI)

No. of

cases

162

No. of cases with

high levels of ANC

80 (49.4%)

Absence

50

24 (48.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

56 (50.0%)

1.08 (0.56-2.12)

1.33 (0.64-2.75)

1.11 (0.54-2.30)

0.81

0.44

0.78

Multivariate

ORb (95%CI)

P value

TLS

High levels of serum ALC

Multivariate

Multivariate

ORa (95%CI)

ORb (95%CI)

No. of

cases

162

No. of

cases

162

No. of cases with

low levels of NLR

80 (49.4 %)

Univariate

OR (95%CI)

Low levels of NLR

Multivariate

ORa (95%CI)

Absence

50

15 (30.0%)

1 (reference)

1 (reference)

1 (reference)

Presence

112

65 (58.0%)

3.23 (1.61-6.72)

3.21 (1.51-7.10)

3.15 (1.53-6.72)

0.0009

0.002

0.002

P value

ALC and ANC counts and NLR were divided into high and low categories by the median split.

The median of ALC and ANC were 1522 and 3251 count/mm2, respectively. The median of NLR

was 2.1.

a The odds ratio was initially adjusted for age, sex, BMI, amount of alcohol consumption, family

history of pancreatic cancer, history of smoking, diabetes mellitus, serum CA19-9, serum CEA.

b The odds ratio was initially adjusted for stage, histology, adjuvant, neo-adjuvant, residual

tumor status, gene alterations (KRAS, TP53, CDKN2A, SMAD4).

ALC, absolute lymphocyte counts; ANC, absolute neutrophil counts; NLR, neutrophil-to

lymphocyte ratio; OR, odds ratio; PDAC, pancreatic ductal adenocarcinoma; TLS, tertiary

lymphoid structure.

Table 4. Presence of TLS and cancer specific mortality in PDAC

Pancreatic cancer specific mortality

Univariate

Multivariate

Multivariate

HR (95%CI)

HRa (95%CI)

HRb (95%CI)

All cases

No. of

cases

162

No. of

events

107

TLS absence

50

43

1 (reference)

1 (reference)

TLS presence

112

64

0.40 (0.27-0.59)

0.40 (0.26-0.61)

<0.0001

<0.0001

<0.0001

1 (reference)

1 (reference)

P value

Pathological

stage IIb-IIIc

119

88

TLS absence

40

37

1 (reference)

TLS presence

79

51

0.34 (0.22-0.53)

P value

<0.0001

0.35 (0.22-0.56)

<0.0001

1 (reference)

0.37 (0.25-0.56)

0.33 (0.21-0.52)

<0.0001

The hazard ratio was initially adjusted for age, sex, BMI, amount of alcohol consumption, family

history of pancreatic cancer, history of smoking, diabetes mellitus, serum CA19-9, serum CEA.

b The hazard ratio was initially adjusted for stage, histology, adjuvant, neo-adjuvant, residual tumor

status, gene alterations (KRAS, TP53, CDKN2A, SMAD4).

c Pathological stage was diagnosed based on TNM Classification of Malignant Tumors, 8th Edition.

HR, hazard ratio; PDAC, pancreatic ductal adenocarcinoma; TLS, tertiary lymphoid structure.

Table 5. Adjuvant chemotherapy and patient mortality in PDAC

TLS

Presence

TLS

Absence

Pancreatic cancer specific mortality

Univariate

Multivariate

P value

HR (95%CI)

HR (95%CI)

1 (reference)

1 (reference)

Adjuvant

chemotherapy

Absent

No. of

cases

29

No. of

events

19

S-1

60

27

0.43 (0.24-0.79)

0.007

0.51 (0.26-0.97)

0.04

GEM

Absent

23

16

18

15

0.66 (0.34-1.28)

1 (reference)

0.21

0.60 (0.29-1.26)

1 (reference)

0.18

S-1

14

12

0.66 (0.30-1.42)

0.28

0.61 (0.27-1.37)

0.23

GEM

20

16

0.71 (0.35-1.46)

0.35

0.67 (0.27-1.37)

0.30

P value

The hazard ratio was initially adjusted for age, BMI, history of smoking, residual tumor status. BMI,

body mass index; GEM, gemcitabine; HR, hazard ratio; PDAC, pancreatic ductal adenocarcinoma;

TLS, tertiary lymphoid structure.

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る