Subscribe to RSS
DOI: 10.1055/a-2739-5187
Prognostic Significance of CD68+ Macrophages and FoxP3+ Regulatory T Cells in Neuroendocrine Tumors
Prognostische Bedeutung von CD68-positiven Macrophagen und FoxP3-positiven regulatorischen T-Zellen in neuroendokrinen TumorenAuthors
Supported by: Medical Faculty of the University of Bonn, “Förderinstrument Klinische Studien” (FKS) 2202-FKS-01
Abstract
Aim
Neuroendocrine tumors (NETs) are a heterogeneous group of malignancies characterized by variable immune microenvironment. This study aimed to analyze the infiltration of NETs by CD68+ tumor-associated macrophages (TAMs) and FoxP3+ regulatory T cells (Tregs) using Qupath software for semi-automated histopathological quantification
Methods
Thirty-two patients with resected NETs were included. CD68 and FoxP3 expression were assessed by immunohistochemistry followed by automated cell detection and spatial analysis.
Results
The mean percentage of CD68+ cells was 0.35%, with no significant differences between pancreatic and gastrointestinal NETs. FoxP3+ cells were less frequent (mean: 0.06%), with higher levels in gastrointestinal NETs compared to pancreatic NETs (p < 0.05). Kaplan-Meier analysis revealed that CD68+ cells showed a trend to positive correlation with progression-free survival (PFS) and overall survival (OS). Thereby, patients exceeding a 0.1% CD68+ cell threshold exhibited longer survival (PFS: not reached vs. 106 months). In contrast, FoxP3+ cells showed a trend to inverse correlation with survival; patients with >0.05% FoxP3+ cells had shorter PFS (36 months vs. 147 months) and OS (180 months vs. not reached). Multivariate Cox regression identified N-stage as the sole predictor of OS. CD68+ TAM density was not associated with other clinical parameters, while FoxP3+ cell infiltration correlated significantly with venous invasion and advanced N- and M-stages.
Conclusion
These findings highlight the potential prognostic relevance of immune cell infiltration in NETs and underscore the utility of Qupath for quantifying immune markers in histopathological analyses. The contrasting roles of CD68+ TAMs and FoxP3+ Tregs in the NET microenvironment warrant further exploration to inform immunotherapeutic strategies.
Publication History
Received: 26 September 2025
Accepted: 05 November 2025
Article published online:
01 December 2025
© 2025. Thieme. All rights reserved.
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
References
- 1 Modlin IM, Oberg K, Chung DC. et al. Gastroenteropancreatic neuroendocrine tumours. Lancet Oncol 2008; 9 (01) 61-72
- 2 Dasari A, Shen C, Halperin D. et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol 2017; 3 (10) 1335-1342
- 3 Strosberg J, El-Haddad G, Wolin E. et al. Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med 2017; 376 (02) 125-135
- 4 Caplin ME, Pavel M, Ćwikła JB. et al. Lanreotide in Metastatic Enteropancreatic Neuroendocrine Tumors. N Engl J Med 2014; 371 (03) 224-233
- 5 Dasari A, Shen C, Halperin D. et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol 2017; 3 (10) 1335-1342
- 6 Yao JC, Shah MH, Ito T. et al. Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N Engl J Med 2011; 364 (06) 514-523
- 7 Raymond E, Dahan L, Raoul JL. et al. Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N Engl J Med 2011; 364 (06) 501-513
- 8 Pavel ME, Hainsworth JD, Baudin E. et al. Everolimus plus Octreotide Long-Acting Repeatable for the Treatment of Advanced Neuroendocrine Tumors Associated with Carcinoid Syndrome (RADIANT-2): A Randomised, Placebo-Controlled, Phase 3 Study. Lancet 2011; 378 (9808) 2005-2012
- 9 Strosberg JR, Caplin ME, Kunz PL. et al. 177Lu-Dotatate Plus Long-Acting Octreotide Versus High-Dose Long-Acting Octreotide in Patients with Midgut Neuroendocrine Tumours (NETTER-1): Final Overall Survival and Long-Term Safety Results from an Open-Label, Randomised, Controlled, Phase 3 Trial. Lancet Oncol 2021; 22 (12) 1752-1763
- 10 Caplin ME, Pavel M, Ćwikła JB. et al. CLARINET Investigators. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med 2014; 371 (03) 224-233
- 11 Kulke MH, Siu LL, Tepper JE. et al. Future Directions in the Treatment of Neuroendocrine Tumors: Consensus Report of the National Cancer Institute Neuroendocrine Tumor Clinical Trials Planning Meeting. J Clin Oncol 2011; 29 (07) 934-943
- 12 Rinke A, Müller HH, Schade-Brittinger C. et al. Placebo-Controlled, Double-Blind, Prospective, Randomized Study on the Effect of Octreotide LAR in the Control of Tumor Growth in Patients with Metastatic Neuroendocrine Midgut Tumors: A Report from the PROMID Study Group. J Clin Oncol 2009; 27 (28) 4656-4663
- 13 Singh S. et al. [177Lu]Lu-DOTA-TATE in newly diagnosed patients with advanced grade 2 and grade 3, well-differentiated gastroenteropancreatic neuroendocrine tumors: Primary analysis of the phase 3 randomized NETTER-2 study. J Clin Oncol 2024; 42 (Suppl. 3) LBA588
- 14 Baudin E. et al. First multicentric randomized phase II trial investigating the antitumor efficacy of peptide receptor radionuclide therapy with 177Lutetium-Octreotate (OCLU) in unresectable progressive neuroendocrine pancreatic tumor. Results of the Ann Oncol 2022; 33 (Suppl. 07) S808-S869
- 15 Bankhead P, Loughrey MB, Fernández JA. et al. QuPath: Open source software for digital pathology image analysis. Sci Rep 2017; 7: 16878
- 16 Cavalcanti E, Armentano R, Valentini A. et al. Role of PD-L1 expression as a biomarker for GEP neuroendocrine neoplasm grading. Cell Death Dis 2017; 8: e3004
- 17 Takahashi D, Kojima M, Suzuki T. et al. Profiling the tumour immune microenvironment in pancreatic neuroendocrine neoplasms with multispectral imaging indicates distinct subpopulation characteristics concordant with WHO 2017 classification. Sci Rep 2018; 8: 13166
- 18 Cives M, Strosberg J, Al Diffalha S. et al. Analysis of the immune landscape of small bowel neuroendocrine tumors. Endocr Relat Cancer 2019; 26: 119-130
- 19 Gong J, Chehrazi-Raffle A, Reddi S. et al. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer 2018; 6 (01) 8
- 20 Kim ST, Ha SY, Lee S. et al. The impact of PD-L1 expression in patients with metastatic GEP-NETs. J Cancer 2016; 7 (05) 484-489
- 21 da Silva A, Bowden M, Zhang S. et al. Characterization of the neuroendocrine tumor immune microenvironment. Pancreas 2018; 47 (09) 1123-1129
- 22 Coppola D, Nebozhyn M, Khalil F. et al. Unique ectopic lymph node-like structures present in human primary colorectal carcinoma are identified by immune gene array profiling. Am J Pathol 2011; 179 (01) 37-45
- 23 Mehnert JM, Bergsland E, O’Neil BH. et al. Pembrolizumab for patients with PD-L1-positive advanced carcinoid or pancreatic neuroendocrine tumors: results from the KEYNOTE-028 study. Ann Oncol 2017; 28 (04) 1033-1039
- 24 Cives M, Pelle’ E, Quaresmini D. et al. The tumor microenvironment in neuroendocrine tumors: biology and therapeutic implications. Neuroendocrinology 2019; 109 (02) 83-99
- 25 Kzhyshkowska J, Shen J, Larionova I. Targeting of TAMs: can we be more clever than cancer cells?. Cell Mol Immunol 2024; 21 (12) 1376-1409
