Skip to main content

Advertisement

Log in

Characterizing the Immune Environment in Peritoneal Carcinomatosis: Insights for Novel Immunotherapy Strategies

  • Translational Research
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background or Purpose

Carcinomatosis, a distinct pattern of metastatic cancer in the peritoneal cavity, poses challenges for treatment and has limited therapeutic options. Understanding the immune environment of peritoneal surface malignancies is crucial for developing effective immunotherapeutic approaches. This study characterizes soluble immune mediators in the peritoneal fluid of patients with and without carcinomatosis to identify targets for novel treatment strategies.

Patients and Methods

Serum and peritoneal fluid samples were collected from surgical patients, and a multianalyte analysis was performed using the Luminex platform. Patient characteristics, tumor sites, and sample collection details were recorded. Soluble immune mediator levels were measured and compared between peritoneal fluid and serum samples and among clinical subgroups. Statistical analysis was conducted to assess differences in analyte concentrations and correlations between samples.

Results

There were 39 patients included in the study, with varying surgical indications. Significant differences were observed in soluble immune mediator levels between peritoneal fluid and serum, with peritoneal fluid exhibiting lower concentrations. Carcinomatosis was associated with elevated levels of proinflammatory mediators, including IL-6 and IL-8, while adaptive immune response markers were low in peritoneal fluid.

Conclusions

The peritoneal immune microenvironment in carcinomatosis favors innate immunity, presenting a challenging environment for effective antitumor response. High levels of proinflammatory mediators suggest potential targets for intervention, such as the IL-6 axis, FGF2, IL-8, and CCL2; these could be explored as potential mitigators of malignant ascites and enhance anti-tumor immune responses. These findings provide valuable insights for developing immunotherapy strategies and improving outcomes in patients with peritoneal carcinomatosis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Bootsma S, Bijlsma MF, Vermeulen L. The molecular biology of peritoneal metastatic disease. EMBO Mol Med. 2023;15(3):e15914.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Fanotto V, Salani F, Vivaldi C, et al. Primary tumor resection for metastatic colorectal, gastric and pancreatic cancer patients: In search of scientific evidence to inform clinical practice. Cancers. 2023;15(3):900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ng D, Ali A, Lee K, et al. Investigating the mechanisms of peritoneal metastasis in gastric adenocarcinoma using a novel ex vivo peritoneal explant model. Sci Rep. 2022;12(1):11499.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wall JA, Klempner SJ, Arend RC. The anti-DKK1 antibody DKN-01 as an immunomodulatory combination partner for the treatment of cancer. Expert Opin Investig Drugs. 2020;29(7):639–44.

    Article  CAS  PubMed  Google Scholar 

  5. Kim C, Kim W, Han Y, Kim J, Chon H. Cancer immunotherapy with STING agonist and PD-1 immune checkpoint inhibitor effectively suppresses peritoneal carcinomatosis of colon cancer. Ann Oncol. 2019;30:iv35.

  6. González-Martín A, Sánchez-Lorenzo L. Immunotherapy with checkpoint inhibitors in patients with ovarian cancer: Still promising? Cancer. 2019;125:4616–22.

    Article  PubMed  Google Scholar 

  7. Morisaki T, Hikichi T, Onishi H, et al. Intranodal administration of neoantigen peptide-loaded dendritic cell vaccine elicits epitope-specific T cell responses and clinical effects in a patient with chemorefractory ovarian cancer with malignant ascites. Immunol Invest. 2021;50(5):562–79.

    Article  CAS  PubMed  Google Scholar 

  8. Ornella MSC, Badrinath N, Kim K-A, et al. Immunotherapy for peritoneal carcinomatosis: Challenges and prospective outcomes. Cancer. 2023;15(8)https://doi.org/10.3390/cancers15082383

  9. Lee YS, Lee WS, Kim CW, et al. Oncolytic vaccinia virus reinvigorates peritoneal immunity and cooperates with immune checkpoint inhibitor to suppress peritoneal carcinomatosis in colon cancer. J Immunother Cancer. 2020;8(2)

  10. Oh S, Lee J-H, Kwack K, Choi S-W. Natural killer cell therapy: A new treatment paradigm for solid tumors. Cancer. 2019;11(10):1534.

    Article  CAS  Google Scholar 

  11. Feist M, Zhu Z, Dai E, et al. Oncolytic virus promotes tumor-reactive infiltrating lymphocytes for adoptive cell therapy. Cancer Gene Ther. 2021;28(1–2):98–111.

    Article  CAS  PubMed  Google Scholar 

  12. Sur D, Havasi A, Cainap C, et al. Chimeric antigen receptor T-cell therapy for colorectal cancer. J Clin Med. 2020;9(1):182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Parayath N, Padmakumar S, Nair SV, Menon D, Amiji MM. Strategies for targeting cancer immunotherapy through modulation of the tumor microenvironment. Regenerative Engineering and Translational Medicine. 2020;6:29–49.

    Article  Google Scholar 

  14. Ganesh K, Massagué J. Targeting metastatic cancer. Nature Med. 2021;27(1):34–44.

    Article  CAS  PubMed  Google Scholar 

  15. Schoutrop E, Moyano-Galceran L, Lheureux S, et al. Molecular, cellular and systemic aspects of epithelial ovarian cancer and its tumor microenvironment. Elsevier; 2022:

  16. Morano WF, Aggarwal A, Love P, Richard SD, Esquivel J, Bowne WB. Intraperitoneal immunotherapy: Historical perspectives and modern therapy. Cancer Gene Ther. 2016;23(11):373–81. https://doi.org/10.1038/cgt.2016.49.

    Article  CAS  PubMed  Google Scholar 

  17. Thadi A, Khalili M, Morano WF, Richard SD, Katz SC, Bowne WB. Early investigations and recent advances in intraperitoneal immunotherapy for peritoneal metastasis. Vaccines (Basel). Aug 10 2018;6(3)https://doi.org/10.3390/vaccines6030054

  18. Zeltsman M, Mayor M, Jones DR, Adusumilli PS. Surgical immune interventions for solid malignancies. Am J Surg. Oct 2016;212(4):682-690 e5. https://doi.org/10.1016/j.amjsurg.2016.06.008

  19. Katz SC, Point GR, Cunetta M, et al. Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther. 2016;23(5):142–8. https://doi.org/10.1038/cgt.2016.14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ornella MSC, Badrinath N, Kim KA, et al. Immunotherapy for peritoneal carcinomatosis: Challenges and prospective outcomes. Cancers (Basel). Apr 20 2023;15(8)https://doi.org/10.3390/cancers15082383

  21. Kang D, Kim IH. Molecular mechanisms and potential rationale of immunotherapy in peritoneal metastasis of advanced gastric cancer. Biomedicines. Jun 10 2022;10(6)https://doi.org/10.3390/biomedicines10061376

  22. Kubicka U, Olszewski WL, Tarnowski W, Bielecki K, Ziolkowska A, Wierzbicki Z. Normal human immune peritoneal cells: subpopulations and functional characteristics. Scand J Immunol. 1996;44(2):157–63. https://doi.org/10.1046/j.1365-3083.1996.d01-297.x.

    Article  CAS  PubMed  Google Scholar 

  23. Eum HH, Kwon M, Ryu D, et al. Tumor-promoting macrophages prevail in malignant ascites of advanced gastric cancer. Exp Mol Med. 2020;52(12):1976–88. https://doi.org/10.1038/s12276-020-00538-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Vlaeminck-Guillem V, Bienvenu J, Isaac S, et al. Intraperitoneal cytokine level in patients with peritoneal surface malignancies. A study of the RENAPE (French Network for Rare Peritoneal Malignancies). Ann Surg Oncol. Aug 2013;20(8):2655-62. https://doi.org/10.1245/s10434-013-2933-6

  25. Donnenberg VS, Luketich JD, Sultan I, et al. A maladaptive pleural environment suppresses preexisting anti-tumor activity of pleural infiltrating T cells. Front Immunol. 2023;1410.3389/fimmu.2023.1157697

  26. Langmuir T, Chu A, Sehabi G, et al. A new landscape in illness uncertainty: A systematic review and thematic synthesis of the experience of uncertainty in patients with advanced cancer receiving immunotherapy or targeted therapy. Psychooncology. 2023;

  27. Marei HE, Hasan A, Pozzoli G, Cenciarelli C. Cancer immunotherapy with immune checkpoint inhibitors (ICIs): potential, mechanisms of resistance, and strategies for reinvigorating T cell responsiveness when resistance is acquired. Cancer Cell Int. 2023;23(1):64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Salmon H, Remark R, Gnjatic S, Merad M. Host tissue determinants of tumour immunity. Nature Rev Cancer. 2019;19(4):215–27.

    CAS  Google Scholar 

  29. Chalaris A, Garbers C, Rabe B, Rose-John S, Scheller J. The soluble Interleukin 6 receptor: generation and role in inflammation and cancer. Eur J Cell Biol. 2011;90(6–7):484–94.

    Article  CAS  PubMed  Google Scholar 

  30. Onogawa T, Saito-Taki T, Yamamoto H, Wada T. IL6 trans-signaling promotes functional recovery of hypofunctional phagocytes through STAT3 activation during peritonitis. Inflamm Res. 2013;62:797–810.

    Article  CAS  PubMed  Google Scholar 

  31. Tahmasebi S, Alimohammadi M, Khorasani S, Rezaei N. Pro-tumorigenic and anti-tumorigenic roles of pro-inflammatory cytokines in cancer. Handbook of Cancer and Immunology. Springer; 2022:1-25.

  32. Kumari S, Advani D, Sharma S, Ambasta RK, Kumar P. Combinatorial therapy in tumor microenvironment: where do we stand? Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2021;1876(2):188585.

  33. Furuya M. Ovarian cancer stroma: pathophysiology and the roles in cancer development. Cancer. 2012;4(3):701–24.

    Article  Google Scholar 

  34. Rodrigues M, Griffith LG, Wells A. Growth factor regulation of proliferation and survival of multipotential stromal cells. Stem Cell Res Therapy. 2010;1(4):1–12.

    Article  Google Scholar 

  35. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trend Immunol. 2016;37(3):208–20.

    Article  CAS  Google Scholar 

  36. Witte MH, Daley SK. Lymphatic system biology, pathobiology, and relation to cancer metastasis. Cancer Metastasis Through the Lymphovascular System. 2022:187-198.

  37. Kusamura S, Baratti D, Zaffaroni N, et al. Pathophysiology and biology of peritoneal carcinomatosis. World Jl Gastroint Oncol. 2010;2(1):12.

    Article  Google Scholar 

  38. Donnenberg VS, Wagner PL, Luketich JD, Bartlett DL, Donnenberg AD. Localized intra-cavitary therapy to drive systemic anti-tumor immunity. Front Immunol. 2022;13:363.

    Article  Google Scholar 

  39. Cui Q, Li L, Hu Y, Wang F, Zhang C, Li C. Effects of stress response induced by laparoscopic colectomy and laparotomy on TLR-mediated innate immune responses in colon cancer patients. Oncologie. 2023;(0)

  40. Naseri S, Rosenberg‐Hasson Y, Maecker HT, Avrutsky MI, Blumenthal PD. A cross‐sectional study comparing the inflammatory profile of menstrual effluent vs. peripheral blood. Health Sci Rep. 2023;6(1):e1038.

  41. Lohani K, Shetty S, Sharma P, Govindarajan V, Thomas P, Loggie B. Pseudomyxoma peritonei: inflammatory responses in the peritoneal microenvironment. Ann Surgical Oncol. 2014;21:1441–7.

    Article  Google Scholar 

  42. Hirano T. IL-6 in inflammation, autoimmunity and cancer. Int immunol. 2021;33(3):127–48.

    Article  CAS  PubMed  Google Scholar 

  43. Johnson DE, O’Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nature reviews Clinical ncology. 2018;15(4):234–48.

    Article  CAS  Google Scholar 

  44. Chung AS, Lee J, Ferrara N. Targeting the tumour vasculature: insights from physiological angiogenesis. Nature Rev Cancer. 2010;10(7):505–14.

    Article  CAS  Google Scholar 

  45. Fousek K, Horn LA, Palena C. Interleukin-8: A chemokine at the intersection of cancer plasticity, angiogenesis, and immune suppression. Pharmacol Ther. 2021;219:107692.

    Article  CAS  PubMed  Google Scholar 

  46. Saleh R. Elkord E. Acquired resistance to cancer immunotherapy: Role of tumor-mediated immunosuppression. Elsevier; 2020. p. 13–27.

    Google Scholar 

Download references

Acknowledgments

This work was supported by W81XWH2210009_BC210533 and W81XWH2211069_ BC211396 from the Department of Defense, MetaVivor FP00002718, The Pennsylvania Breast Cancer Coalition, the Glimmer of Hope Foundation, and the David Downing Fund. The Hillman Cancer Center Luminex Facility is supported by Cancer Center Support Grant P30CA047904.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrick L. Wagner MD, MPH.

Ethics declarations

Disclosure

None.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wagner, P.L., Knotts, C.M., Donneberg, V.S. et al. Characterizing the Immune Environment in Peritoneal Carcinomatosis: Insights for Novel Immunotherapy Strategies. Ann Surg Oncol 31, 2069–2077 (2024). https://doi.org/10.1245/s10434-023-14553-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-023-14553-6

Navigation