Next Article in Journal
Prenatal Exposure to Metabolism-Disrupting Chemicals, Cord Blood Transcriptome Perturbations, and Birth Weight in a Belgian Birth Cohort
Previous Article in Journal
Fully Aqueous Self-Assembly of a Gold-Nanoparticle-Based Pathogen Sensor
Previous Article in Special Issue
Immune-Mediated Neuropathies: Pathophysiology and Management
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

New Roles for Old Friends: Involvement of the Innate Immune System in Tumor Progression

by
María Castaño
1,†,
Eva González-Cantó
2,†,
Cristina Aghababyan
2,3,
Sarai Tomás-Pérez
2,
Julia Oto
1,4,
Raquel Herranz
1,
Pilar Medina
1,
Martin Götte
4,
Bárbara Andrea Mc Cormack
1,2,*,
Josep Marí-Alexandre
2,5,* and
Juan Gilabert-Estellés
2,3,6
1
Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, 46026 Valencia, Spain
2
Research Laboratory in Biomarkers in Reproduction, Gynaecology, and Obstetrics, Research Foundation of the General University Hospital of Valencia, 46014 Valencia, Spain
3
Department of Obstetrics and Gynecology, General University Hospital of Valencia Consortium, 46014 Valencia, Spain
4
Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Muenster, Germany
5
Department of Pathology, General University Hospital of Valencia Consortium, 46014 Valencia, Spain
6
Department of Pediatrics, Obstetrics, and Gynecology, University of Valencia, 46014 Valencia, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(8), 7604; https://doi.org/10.3390/ijms24087604
Submission received: 3 March 2023 / Accepted: 17 April 2023 / Published: 20 April 2023
The association between the immune system and tumor progression has attracted much interest in the research community in recent years. The immune system, constituted by varied cell types that perform a coordinated response, is classified into innate and adaptive immunity. Effectors of innate immunity include neutrophils, macrophages, monocytes, eosinophils, basophils, and natural killer cells (NK). On the other hand, adaptive immunity involves T and B lymphocytes, which present a specific and structurally unique receptor [1].
The adaptive immune system has been the focus of research attention, which led to a better understanding of how lymphocytes interact with tumor cells, the discovery of immune checkpoints, and the development of targeted therapies. Initially, most efforts to identify and select antigens for therapeutic targeting cancer focused on shared tumor antigens, as they can be used to treat a wide range of cancer patients. However, it is becoming increasingly clear that many of these shared antigens are also expressed by self-tissues, either in peripheral cells or the thymus, which can lead to immunologic tolerance for the highest avidity interactions between peptide, major histocompatibility complex I and T-cell antigen receptor. As a result, immune responses against these antigens can be restricted to lower-affinity interactions, which may limit therapeutic efficacy [2].
Moreover, immune checkpoint inhibitors (ICIs) have become the first-line therapy for various solid and liquid tumor types. ICIs act by releasing the inhibitory brakes of T-cells, leading to strong activation of the immune system and effective antitumor immune responses. Currently, the most commonly used immunotherapeutic agents are blocking antibodies that target immune inhibitory receptors, such as CTLA-4 and PD-1 [3]. For instance, three anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) are primarily used to treat urothelial carcinoma, non-small cell lung cancer, Merkel cell carcinoma, and melanoma, among others [3,4]. Nevertheless, while many patients experience a significant reduction in tumor size as a result of ICIs, others with different tumor types do not respond to these therapies, as occurs for metastatic breast cancer [5] and ovarian cancer, in which clinical trials have reported poor outcomes in terms of patient response and survival [6]. Among patients who initially respond, some eventually develop resistance and experience tumor relapse, which is known as acquired resistance [3]. Chimeric antigen receptor (CAR) T-cells are also extensively studied as potential adjuvant therapies for cancer treatment. These cells are designed to promote specific T-cell responses and can be used in combination with surgical resection and chemotherapy to treat various types of cancer [7]. CAR T-cell treatment has provided remarkable results in hematological tumors but not in solid tumors, where T-cells encounter substantial difficulties in penetrating and surviving in the tumor microenvironment (TME) [8].
Undoubtedly, the involvement of the innate immune system in human cancer development and progression is now receiving increasing recognition. Thereby, the identification of various elements of the innate immune system such as tumor-associated macrophages (TAMs) and neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and NK cells in the TME has motivated researchers to investigate its role in creating a favorable environment for tumor growth and metastasis. Here we intend to briefly summarize the involvement and relevance of each of them.
Neutrophils, the most abundant leukocytes in humans, have diverse and critical roles in immunity. Traditionally, they have been considered part of the innate immune system constituting the first line of defense at sites of infection and injury [9]. Surprisingly, high levels of TANs have been linked with an adverse prognosis in different malignancies such as renal cell carcinoma, melanoma, colorectal cancer, hepatocellular carcinoma, glioblastoma, gastric, esophageal, lung, ovarian, and neck cancer [9]. Similarly to the M1/M2 phenotype of macrophages, two polarization states of TANs have been proposed, termed N1 and N2, representing anti-tumor and pro-tumor populations, respectively [10]. Moreover, in response to different stimuli, neutrophils have proven to be capable of releasing NETs composed of DNA, citH3, and cytoplasmic and granular proteins (such as calprotectin, MPO, and elastase) in a process called NETosis [11]. In cancer, those NETs are reportedly able to awaken dormant cancer cells, promoting cancer relapse and metastasis. In addition, they can be involved in inhibiting the immune response through evasion of adaptive immunity and ICIs, resistance to oncologic therapies, and entraping of circulating cancer cells, enhancing metastasis spread [12]. Hence, targeting NETs could represent a promising tool in cancer treatment since their markers in biofluids may serve as predictive biomarkers and their 3D structures as therapeutic targets. Interestingly, NETosis markers have also been shown to be useful indicators of other cancer-related processes, such as thrombotic complications. CitH3 has been presented as a predictor of venous thromboembolic events in cancer patients [13]. Additionally, in biliopancreatic cancer patients, calprotectin measured at diagnosis has been proposed as a biomarker to predict future venous thromboembolic events during follow-up [14]. In glioma, cell-free DNA (cfDNA) and MPO levels in plasma measured before surgery have also been proposed as predictors of incidental post-surgical pulmonary embolism in these patients [15]. Finally, plasma cfDNA and calprotectin have been postulated as potential minimally-invasive biomarkers of high-grade serous ovarian cancer that may improve current diagnostic markers [16].
Macrophages are innate immune cells that differentiate from circulating monocytes after extravasation into tissues. In oncological patients and preclinical experimental models, TAMs correlate with poor prognosis and reduced overall survival [10]. TAMs have two functionally polarized phenotypes in the TME: classically activated M1 and alternatively activated M2 macrophages. In the initial stages of various tumors, TAMs are preferentially polarized toward the M1 phenotype exerting anti-tumor functions. However, during cancer progression, changes in the TME promote the gradual shift towards a polarized M2 phenotype and exhibit distinct pro-tumorigenic functional properties [7,10,17]. Furthermore, monocytes and macrophages can extrude extracellular traps (MoETs and METs, respectively) that are composed of nuclear DNA fibers with myeloperoxidase (MPO), citrullinated histoneH3 (citH3), matrix metalloproteinase 9 and 12, and lysozyme [18]. Recently, a study from Xu et al. revealed that METs participate in tumor progression and identified several sources of NETs and METs in tumor tissues isolated from patients with pancreatic neuroendocrine cancer. The authors concluded that these extracellular traps can be used as biological indicators of patient prognosis [19]. Since the presence of MoETs and METs correlates with a worse prognostic in cancer patients, they could be useful as novel diagnostic and prognostic biomarkers.
MDSC are generated in the bone marrow and rapidly differentiate into macrophages, dendritic cells, neutrophils, eosinophils, basophils, and mast cells in healthy individuals. Thus, MDSCs are a heterogeneous collection of activated immature myeloid cells, comprising a mixture of granulocytic and monocytic subtypes [9]. They can be both polymorphonuclear (PMN-MDSCs) or mononuclear (M-MDSCs). PMN-MDSCs are morphologically and phenotypically similar to neutrophils while M-MDSCs are similar to monocytes [7]. During cancer, MDSCs can migrate to peripheral lymphoid tissues and tumor sites, contributing to TME formation [9]. Consequently, MDSCs have recently emerged as primarily responsible for the immune system blockade observed in cancer [20], since they are present in most cancer patients and have been shown to inhibit cytotoxic T-cell function and block T-cell enrichment in the tumor [9]. Since PMN-MDSCs are generally short-living cells, the differentiation has been studied in more detail for M-MDSCs into TAMs. Accordingly, M-MDSCs represent a potential therapeutic target for cancer therapy, not only because of their ability to suppress immune responses but also because of their high plasticity and differentiation potential. Therefore, it would be interesting that therapeutic targeting includes the blockade of M-MDSC migration to the tumors, the inhibition of M-MDSC differentiation into TAMs, and TAMs polarization [20].
Finally, NK cells are innate immune cells that display rapid and potent cytolytic activity in response to infected or transformed cells and possess a well-documented anti-tumor effect. The presence of NK cell infiltration in some types of cancers, such as colorectal and gastric, has been correlated with a favorable outcome [10]. Therefore, as a significant player in the innate immune response, NK cells present remarkable potential in clinical practice. In recent years, research into NK cell immunotherapy has experienced a boost. The latest developments mainly focused on cytokine supplements, monoclonal antibodies, modification of internal signal pathways, and genetic engineering of NK cells. Moreover, NK-cell-based therapy has achieved favorable results used either alone or in combination with other therapies, suggesting a wide and effective use in malignancies. Some of the therapies designed for NK cells are under clinical trials, such as monoclonal antibodies targeting the NK immunoglobulin-like receptor, anti-KIR (IPH2101, lirilumab), and those targeting ICI of NK, anti-NKG2A (monalizumab) [21].
Even though the adaptive immune system contribution has received considerable attention for several years in cancer, the involvement of the innate immune system in human oncology is now receiving increasing recognition. The identification of various innate immune system effectors in the TME has prompted researchers to investigate their role in tumor progression and metastasis, leading to the development of potential targeted therapies. Undoubtedly, this field is in constant development and much remains to be unraveled. Therefore, we encourage researchers to share their latest findings in this promising field related to the management of oncologic disorders.

Author Contributions

Conceptualization: B.A.M.C. and J.M.-A.; methodology (literature review and selection of manuscripts): M.C., E.G.-C., C.A., S.T.-P., J.O., R.H. and B.A.M.C.; writing—original draft preparation: M.C.; writing—review and editing: M.C., P.M., M.G., B.A.M.C., J.M.-A. and J.G.-E.; supervision: P.M., B.A.M.C., J.M.-A. and J.G.-E.; funding acquisition: P.M., M.G., J.M.-A. and J.G.-E. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kaur, B.P.; Secord, E. Innate Immunity. Immunol. Allergy Clin. N. Am. 2021, 41, 535–541. [Google Scholar] [CrossRef] [PubMed]
  2. Gajewski, T.F.; Schreiber, H.; Fu, Y.-X. Innate and Adaptive Immune Cells in the Tumor Microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
  3. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. Mech. Dis. 2021, 16, 223–249. [Google Scholar] [CrossRef] [PubMed]
  4. Chang, C.-Y.; Park, H.; Malone, D.C.; Wang, C.-Y.; Wilson, D.L.; Yeh, Y.-M.; Van Boemmel-Wegmann, S.; Lo-Ciganic, W.-H. Immune Checkpoint Inhibitors and Immune-Related Adverse Events in Patients with Advanced Melanoma: A Systematic Review and Network Meta-Analysis. JAMA Netw. Open 2020, 3, e201611. [Google Scholar] [CrossRef]
  5. Tommasi, C.; Pellegrino, B.; Diana, A.; Palafox Sancez, M.; Orditura, M.; Scartozzi, M.; Musolino, A.; Solinas, C. The Innate Immune Microenvironment in Metastatic Breast Cancer. J. Clin. Med. 2022, 11, 5986. [Google Scholar] [CrossRef]
  6. Maiorano, B.A.; Maiorano, M.F.P.; Lorusso, D.; Maiello, E. Ovarian Cancer in the Era of Immune Checkpoint Inhibitors: State of the Art and Future Perspectives. Cancers 2021, 13, 4438. [Google Scholar] [CrossRef]
  7. Cui, K.; Hu, S.; Mei, X.; Cheng, M. Innate Immune Cells in the Esophageal Tumor Microenvironment. Front. Immunol. 2021, 12, 654731. [Google Scholar] [CrossRef]
  8. Gianni, C.; Palleschi, M.; Schepisi, G.; Casadei, C.; Bleve, S.; Merloni, F.; Sirico, M.; Sarti, S.; Cecconetto, L.; Di Menna, G.; et al. Circulating Inflammatory Cells in Patients with Metastatic Breast Cancer: Implications for Treatment. Front. Oncol. 2022, 12, 882896. [Google Scholar] [CrossRef]
  9. Rakic, A.; Beaudry, P.; Mahoney, D.J. The Complex Interplay between Neutrophils and Cancer. Cell Tissue Res. 2018, 371, 517–529. [Google Scholar] [CrossRef]
  10. Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the Immune System in Cancer: From Tumor Initiation to Metastatic Progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef]
  11. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil Extracellular Traps Kill Bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  12. Masucci, M.T.; Minopoli, M.; Del Vecchio, S.; Carriero, M.V. The Emerging Role of Neutrophil Extracellular Traps (NETs) in Tumor Progression and Metastasis. Front. Immunol. 2020, 11, 1749. [Google Scholar] [CrossRef] [PubMed]
  13. Mauracher, L.-M.; Posch, F.; Martinod, K.; Grilz, E.; Däullary, T.; Hell, L.; Brostjan, C.; Zielinski, C.; Ay, C.; Wagner, D.D.; et al. Citrullinated Histone H3, a Biomarker of Neutrophil Extracellular Trap Formation, Predicts the Risk of Venous Thromboembolism in Cancer Patients. J. Thromb. Haemost. 2018, 16, 508–518. [Google Scholar] [CrossRef] [PubMed]
  14. Oto, J.; Navarro, S.; Larsen, A.C.; Solmoirago, M.J.; Plana, E.; Hervás, D.; Fernández-Pardo, Á.; España, F.; Kristensen, S.R.; Thorlacius-Ussing, O.; et al. MicroRNAs and Neutrophil Activation Markers Predict Venous Thrombosis in Pancreatic Ductal Adenocarcinoma and Distal Extrahepatic Cholangiocarcinoma. Int. J. Mol. Sci. 2020, 21, 840. [Google Scholar] [CrossRef]
  15. Oto, J.; Plana, E.; Solmoirago, M.J.; Fernández-Pardo, Á.; Hervás, D.; Cana, F.; España, F.; Artoni, A.; Bucciarelli, P.; Carrabba, G.; et al. MicroRNAs and Markers of Neutrophil Activation as Predictors of Early Incidental Post-Surgical Pulmonary Embolism in Patients with Intracranial Tumors. Cancers 2020, 12, 1536. [Google Scholar] [CrossRef]
  16. Tomás-Pérez, S.; Oto, J.; Aghababyan, C.; Herranz, R.; Cuadros-Lozano, A.; González-Cantó, E.; Mc Cormack, B.; Arrés, J.; Castaño, M.; Cana, F.; et al. Increased Levels of NETosis Biomarkers in High-Grade Serous Ovarian Cancer Patients’ Biofluids: Potential Role in Disease Diagnosis and Management. Front. Immunol. 2023, 14, 1111344. [Google Scholar] [CrossRef]
  17. Strakhova, R.; Cadassou, O.; Cros-Perrial, E.; Jordheim, L.P. Regulation of Tumor Infiltrated Innate Immune Cells by Adenosine. Purinergic Signal. 2020, 16, 289–295. [Google Scholar] [CrossRef]
  18. Mamtimin, M.; Pinarci, A.; Han, C.; Braun, A.; Anders, H.-J.; Gudermann, T.; Mammadova-Bach, E. Extracellular DNA Traps: Origin, Function and Implications for Anti-Cancer Therapies. Front. Oncol. 2022, 12, 869706. [Google Scholar] [CrossRef]
  19. Xu, S.-S.; Li, H.; Li, T.-J.; Li, S.; Xia, H.-Y.; Long, J.; Wu, C.-T.; Wang, W.-Q.; Zhang, W.-H.; Gao, H.-L.; et al. Neutrophil Extracellular Traps and Macrophage Extracellular Traps Predict Postoperative Recurrence in Resectable Nonfunctional Pancreatic Neuroendocrine Tumors. Front. Immunol. 2021, 12, 577517. [Google Scholar] [CrossRef]
  20. Tcyganov, E.; Mastio, J.; Chen, E.; Gabrilovich, D.I. Plasticity of Myeloid-Derived Suppressor Cells in Cancer. Curr. Opin. Immunol. 2018, 51, 76–82. [Google Scholar] [CrossRef]
  21. Wu, S.-Y.; Fu, T.; Jiang, Y.-Z.; Shao, Z.-M. Natural Killer Cells in Cancer Biology and Therapy. Mol. Cancer 2020, 19, 120. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Castaño, M.; González-Cantó, E.; Aghababyan, C.; Tomás-Pérez, S.; Oto, J.; Herranz, R.; Medina, P.; Götte, M.; Mc Cormack, B.A.; Marí-Alexandre, J.; et al. New Roles for Old Friends: Involvement of the Innate Immune System in Tumor Progression. Int. J. Mol. Sci. 2023, 24, 7604. https://doi.org/10.3390/ijms24087604

AMA Style

Castaño M, González-Cantó E, Aghababyan C, Tomás-Pérez S, Oto J, Herranz R, Medina P, Götte M, Mc Cormack BA, Marí-Alexandre J, et al. New Roles for Old Friends: Involvement of the Innate Immune System in Tumor Progression. International Journal of Molecular Sciences. 2023; 24(8):7604. https://doi.org/10.3390/ijms24087604

Chicago/Turabian Style

Castaño, María, Eva González-Cantó, Cristina Aghababyan, Sarai Tomás-Pérez, Julia Oto, Raquel Herranz, Pilar Medina, Martin Götte, Bárbara Andrea Mc Cormack, Josep Marí-Alexandre, and et al. 2023. "New Roles for Old Friends: Involvement of the Innate Immune System in Tumor Progression" International Journal of Molecular Sciences 24, no. 8: 7604. https://doi.org/10.3390/ijms24087604

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop