1
|
Park SM, Chen CJJ, Verdon DJ, Ooi MPY, Brooks AES, Martin RCW, Mathy JA, Emanuel PO, Dunbar PR. Proliferating macrophages in human tumours show characteristics of monocytes responding to myelopoietic growth factors. Front Immunol 2024; 15:1412076. [PMID: 38903497 PMCID: PMC11188303 DOI: 10.3389/fimmu.2024.1412076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Macrophages play essential roles in maintaining tissue homeostasis and immune defence. However, their extensive infiltration into tumours has been linked to adverse outcomes in multiple human cancers. Within the tumour microenvironment (TME), tumour-associated macrophages (TAMs) promote tumour growth and metastasis, making them prime targets for cancer immunotherapy. Recent single-cell analysis suggest that proliferating TAMs accumulate in human cancers, yet their origins and differentiation pathways remain uncertain. Here, we show that a subpopulation of CD163+ TAMs proliferates in situ within the TME of melanoma, lung cancer, and breast cancer. Consistent with their potential role in suppressing anti-tumour activities of T cells, CD163+ TAMs express a range of potent immunosuppressive molecules, including PD-L1, PD-L2, IL-10, and TGF-β. Other phenotypic markers strongly suggested that these cells originate from CD14+ CCR2+ monocytes, a cell population believed to have minimal capacity for proliferation. However, we demonstrate in vitro that certain myelopoietic cytokines commonly available within the TME induce robust proliferation of human monocytes, especially the combination of interleukin 3 (IL-3) and Macrophage Colony-Stimulating Factor 1 (M-CSF). Monocytic cells cultured with these cytokines efficiently modulate T cell proliferation, and their molecular phenotype recapitulates that of CD163+ TAMs. IL-3-driven proliferation of monocytic cells can be completely blocked by IL-4, associated with the induction of CDKN1A, alongside the upregulation of transcription factors linked to dendritic cell function, such as BATF3 and IRF4. Taken together, our work suggests several novel therapeutic routes to reducing immunosuppressive TAMs in human tumours, from blocking chemokine-mediated recruitment of monocytes to blocking their proliferation.
Collapse
Affiliation(s)
- Saem Mul Park
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Chun-Jen J. Chen
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Daniel J. Verdon
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Marcus P. Y. Ooi
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Anna E. S. Brooks
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | | | - Jon A. Mathy
- Department of Surgery, Faculty of Medical Health Sciences, The University of Auckland, Auckland, New Zealand
- Auckland Regional Plastic, Reconstructive and Hand Surgery Unit, Auckland, New Zealand
| | - Patrick O. Emanuel
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - P. Rod Dunbar
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| |
Collapse
|
2
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
3
|
Blanc F, Bertho N, Piton G, Leplat JJ, Egidy G, Bourneuf E, Vincent-Naulleau S, Prévost-Blondel A. Deciphering the immune reaction leading to spontaneous melanoma regression: initial role of MHCII + CD163 - macrophages. Cancer Immunol Immunother 2023; 72:3507-3521. [PMID: 37526660 PMCID: PMC10576715 DOI: 10.1007/s00262-023-03503-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023]
Abstract
The human cutaneous metastatic melanoma is the deadliest skin cancer. Partial, or less frequently complete spontaneous regressions could be observed, mainly mediated by T cells. Nevertheless, the underlying mechanisms are not fully unraveled. We investigated the first events of the immune response related to cancer regression in Melanoma-bearing Libechov Minipigs (MeLiM), a unique swine model of cutaneous melanoma that regresses spontaneously. Using a multiparameter flow cytometry strategy and integrating new clinical and histological criteria of the regression, we show that T cells and B cells are present only in the late stages, arguing against their role in the initial destruction of malignant cells. NK cells infiltrate the tumors before T cells and therefore might be involved in the induction of the regression process. Myeloid cells represent the main immune population within the tumor microenvironment regardless of the regression stage. Among those, MHCII+ CD163- macrophages that differ phenotypically and functionally compared to other tumor-associated macrophages, increase in number together with the first signs of regression suggesting their crucial contribution to initiating the regression process. Our study supports the importance of macrophage reprogramming in humans to improve current immunotherapy for metastatic melanoma.
Collapse
Affiliation(s)
- Fany Blanc
- INSERM, U1016, Institut Cochin, 75014, Paris, France.
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
- CEA, DSV/iRCM/SREIT/LREG, 78350, Jouy-en-Josas, France.
| | - Nicolas Bertho
- Université Paris-Saclay, INRAE, VIM, 78350, Jouy-en-Josas, France
- INRAE, Oniris, BIOEPAR, 44300, Nantes, France
| | - Guillaume Piton
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- CEA, DSV/iRCM/SREIT/LREG, 78350, Jouy-en-Josas, France
| | - Jean-Jacques Leplat
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- CEA, DSV/iRCM/SREIT/LREG, 78350, Jouy-en-Josas, France
| | - Giorgia Egidy
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Emmanuelle Bourneuf
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- CEA, DSV/iRCM/SREIT/LREG, 78350, Jouy-en-Josas, France
- Laboratoire de Cancérologie Expérimentale, CEA/DRF/IBFJ/IRCM, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, Paris, France
| | - Silvia Vincent-Naulleau
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- CEA, DSV/iRCM/SREIT/LREG, 78350, Jouy-en-Josas, France
- Plateforme animalerie, CEA/DRF/IBFJ/IRCM, 92265, Fontenay-Aux-Roses, France
- Bureau des Etudes Biomédicales chez l'Animal, CEA/DRF/BEBA, 92265, Fontenay-Aux-Roses, France
| | - Armelle Prévost-Blondel
- INSERM, U1016, Institut Cochin, 75014, Paris, France.
- Université Paris Cité, Paris, France.
- CNRS, UMR8104, Paris, France.
| |
Collapse
|
4
|
The Dog as a Model to Study the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:123-152. [PMID: 34664237 DOI: 10.1007/978-3-030-73119-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Cancer is a complex and dynamic disease with an outcome that depends on a strict crosstalk between tumor cells and other components in tumor microenvironment, namely, tumor-infiltrating immune cells, fibroblasts, cancer stem cells, adipocytes, and endothelial cells. Within the tumor microenvironment, macrophages and T-lymphocytes appear to be key effectors during the several steps of tumor initiation and progression. Tumor cells, through the release of a plethora of signaling molecules, can induce immune tolerance, by avoiding immune surveillance, and inhibit immune cells cytotoxic functions. Furthermore, as the tumor grows, tumor microenvironment reveals a series of dysfunctional conditions that potentiate a polarization of harmful humoral Th2 and Th17, an upregulation of Treg cells, and a differentiation of macrophages into the M2 subtype, which contribute to the activation of several signaling pathways involving important tissue biomarkers (COX-2, EGFR, VEGF) implicated in cancer aggressiveness and poor clinical outcomes. In order to maintain the tumor growth, cancer cells acquire several adaptations such as neovascularization and metabolic reprogramming. An extensive intracellular production of lactate and protons is observed in tumor cells as a result of their high glycolytic metabolism. This contributes not only for the microenvironment pH alteration but also to shape the immune response that ultimately impairs immune cells capabilities and effector functions.In this chapter, the complexity of tumor microenvironment, with special focus on macrophages, T-lymphocytes, and the impact of lactate efflux, was reviewed, always trying to demonstrate the strong similarities between data from studies of humans and dogs, a widely proposed model for comparative oncology studies.
Collapse
|
5
|
Kuraitis D, Rosenthal N, Boh E, McBurney E. Macrophages in dermatology: pathogenic roles and targeted therapeutics. Arch Dermatol Res 2021; 314:133-140. [PMID: 33641015 DOI: 10.1007/s00403-021-02207-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/10/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022]
Abstract
The field of macrophage biology is rapidly growing. Recent studies have shifted focus from classic wound healing roles to newly identified roles in dermatologic pathology. These studies have identified pathogenic roles of macrophages in relatively common conditions, such as psoriasis, skin cancer, and cutaneous T-cell lymphoma. Selective depletion of these cells or their associated cytokines leads to improved clinical outcome. Herein, we review recent animal and human studies that have elucidated novel pathogenic roles of macrophages in conditions frequently encountered by dermatologists and discuss clinically relevant macrophage-targeted therapies.
Collapse
Affiliation(s)
- Drew Kuraitis
- Department of Dermatology, Tulane University, New Orleans, USA.
| | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, USA.,National Heart and Lung Institute and BHF Centre for Research Excellence, Imperial College London, London, UK
| | - Erin Boh
- Department of Dermatology, Tulane University, New Orleans, USA
| | | |
Collapse
|
6
|
Cui C, Xu C, Yang W, Chi Z, Sheng X, Si L, Xie Y, Yu J, Wang S, Yu R, Guo J, Kong Y. Ratio of the interferon-γ signature to the immunosuppression signature predicts anti-PD-1 therapy response in melanoma. NPJ Genom Med 2021; 6:7. [PMID: 33542239 PMCID: PMC7862369 DOI: 10.1038/s41525-021-00169-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/06/2021] [Indexed: 01/30/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) treatments produce clinical benefit in many patients. However, better pretreatment predictive biomarkers for ICI are still needed to help match individual patients to the treatment most likely to be of benefit. Existing gene expression profiling (GEP)-based biomarkers for ICI are primarily focused on measuring a T cell-inflamed tumor microenvironment that contributes positively to the response to ICI. Here, we identified an immunosuppression signature (IMS) through analyzing RNA sequencing data from a combined discovery cohort (n = 120) consisting of three publicly available melanoma datasets. Using the ratio of an established IFN-γ signature and IMS led to consistently better prediction of the ICI therapy outcome compared to a collection of nine published GEP signatures from the literature on a newly generated internal validation cohort (n = 55) and three published datasets of metastatic melanoma treated with anti-PD-1 (n = 54) and anti-CTLA-4 (n = 42), as well as in patients with gastric cancer treated with anti-PD-1 (n = 45), demonstrating the potential utility of IMS as a predictive biomarker that complements existing GEP signatures for immunotherapy.
Collapse
Affiliation(s)
- Chuanliang Cui
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Canqiang Xu
- Aginome-XMU Joint lab, Xiamen University, Xiamen, China
| | - Wenxian Yang
- Aginome-XMU Joint lab, Xiamen University, Xiamen, China
| | - Zhihong Chi
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Xinan Sheng
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Lu Si
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Yihong Xie
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Jinyu Yu
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Shun Wang
- Department of Computer Science, School of Informatics, Xiamen University, Xiamen, China
| | - Rongshan Yu
- Aginome-XMU Joint lab, Xiamen University, Xiamen, China.
- Department of Computer Science, School of Informatics, Xiamen University, Xiamen, China.
| | - Jun Guo
- Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yan Kong
- Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
7
|
Ceci C, Atzori MG, Lacal PM, Graziani G. Targeting Tumor-Associated Macrophages to Increase the Efficacy of Immune Checkpoint Inhibitors: A Glimpse into Novel Therapeutic Approaches for Metastatic Melanoma. Cancers (Basel) 2020; 12:cancers12113401. [PMID: 33212945 PMCID: PMC7698460 DOI: 10.3390/cancers12113401] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/03/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent a promising therapeutic intervention for a variety of advanced/metastatic solid tumors, including melanoma, but in a large number of cases, patients fail to establish a sustained anti-tumor immunity and to achieve a long-lasting clinical benefit. Cells of the tumor micro-environment such as tumor-associated M2 macrophages (M2-TAMs) have been reported to limit the efficacy of immunotherapy, promoting tumor immune evasion and progression. Thus, strategies targeting M2-TAMs have been suggested to synergize with immune checkpoint blockade. This review recapitulates the molecular mechanisms by which M2-TAMs promote cancer immune evasion, with focus on the potential cross-talk between pharmacological interventions targeting M2-TAMs and ICIs for melanoma treatment.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.G.A.)
| | - Maria Grazia Atzori
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.G.A.)
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (C.C.); (M.G.A.)
- Correspondence: ; Tel.: +39-06-7259-6338
| |
Collapse
|
8
|
Differential association of CD68 + and CD163 + macrophages with macrophage enzymes, whole tumour gene expression and overall survival in advanced melanoma. Br J Cancer 2020; 123:1553-1561. [PMID: 32843682 PMCID: PMC7653046 DOI: 10.1038/s41416-020-01037-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 07/20/2020] [Accepted: 08/06/2020] [Indexed: 12/28/2022] Open
Abstract
Background The density and phenotype of tumour-associated macrophages have been linked with prognosis in a range of solid tumours. While there is strong preclinical evidence that tumour-associated macrophages promote aspects of tumour progression, it can be challenging to infer clinical activity from surface markers and ex vivo behaviour. We investigated the association of macrophage infiltration with prognosis and functional changes in the tumour microenvironment in primary human melanoma. Methods Fifty-seven formalin-fixed, paraffin-embedded primary melanomas were analysed by immunohistochemical analysis of CD68, CD163, inducible nitric oxide synthase (iNOS) and arginase expression. RNA sequencing was performed on serial sections of 20 of the stained tumours to determine the influence of macrophage infiltration on gene expression. Results CD68+ cells are a functionally active subset of macrophages that are associated with increased iNOS and arginase staining and altered gene expression. In comparison, while there is a greater accumulation of CD163+ macrophages in larger tumours, these cells are comparatively inactive, with no association with the level of iNOS or arginase staining, and no effect on gene expression within the tumour. The infiltration of either subset of macrophages did not correlate to overall survival. Conclusions Thus, melanomas contain distinct macrophage populations with diverse phenotypes, but with no observable prognostic role.
Collapse
|
9
|
Kim YJ, Won CH, Lee MW, Choi JH, Chang SE, Lee WJ. Correlation Between Tumor-Associated Macrophage and Immune Checkpoint Molecule Expression and Its Prognostic Significance in Cutaneous Melanoma. J Clin Med 2020; 9:jcm9082500. [PMID: 32756500 PMCID: PMC7465191 DOI: 10.3390/jcm9082500] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022] Open
Abstract
The association between tumor-associated macrophages (TAMs) and the expression of immune checkpoint molecules has not been well described in cutaneous melanoma. We evaluated the correlations between the expression of markers of TAMs, cluster of differentiation 163 (CD163), and immune checkpoint molecules, programmed cell death protein-1 (PD-1), and lymphocyte activating gene-3 (LAG-3). We also determined their relationships with the clinicopathological features and disease outcomes in melanoma. Diagnostic tissues collected from melanoma patients were evaluated using immunohistochemistry for CD163, PD-1, and LAG-3 expression. CD163 expression positively correlated with PD-1 and LAG-3 expression. High expression of both CD163 and PD-1 expressions was significantly associated with negative prognostic factors and worse prognosis than high expression of the single markers. High co-expression of CD163 and LAG-3 was associated with poor clinicopathological indexes of melanoma and worse survival compared to the high expression of the single markers. The expression of immune checkpoint molecules PD-1 and LAG-3 positively correlated with the M2-TAM density in melanoma tissue. Simultaneous high M2-TAM density and immune checkpoint molecules expression acted as independent poor prognostic factors in cutaneous melanoma.
Collapse
Affiliation(s)
| | | | | | | | - Sung Eun Chang
- Correspondence: (S.E.C.); (W.J.L.); Tel.: +82-2-3010-3460 (S.E.C.); +82-2-3010-3467 (W.J.L.)
| | - Woo Jin Lee
- Correspondence: (S.E.C.); (W.J.L.); Tel.: +82-2-3010-3460 (S.E.C.); +82-2-3010-3467 (W.J.L.)
| |
Collapse
|
10
|
Skytthe MK, Graversen JH, Moestrup SK. Targeting of CD163 + Macrophages in Inflammatory and Malignant Diseases. Int J Mol Sci 2020; 21:E5497. [PMID: 32752088 PMCID: PMC7432735 DOI: 10.3390/ijms21155497] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
The macrophage is a key cell in the pro- and anti-inflammatory response including that of the inflammatory microenvironment of malignant tumors. Much current drug development in chronic inflammatory diseases and cancer therefore focuses on the macrophage as a target for immunotherapy. However, this strategy is complicated by the pleiotropic phenotype of the macrophage that is highly responsive to its microenvironment. The plasticity leads to numerous types of macrophages with rather different and, to some extent, opposing functionalities, as evident by the existence of macrophages with either stimulating or down-regulating effect on inflammation and tumor growth. The phenotypes are characterized by different surface markers and the present review describes recent progress in drug-targeting of the surface marker CD163 expressed in a subpopulation of macrophages. CD163 is an abundant endocytic receptor for multiple ligands, quantitatively important being the haptoglobin-hemoglobin complex. The microenvironment of inflammation and tumorigenesis is particular rich in CD163+ macrophages. The use of antibodies for directing anti-inflammatory (e.g., glucocorticoids) or tumoricidal (e.g., doxorubicin) drugs to CD163+ macrophages in animal models of inflammation and cancer has demonstrated a high efficacy of the conjugate drugs. This macrophage-targeting approach has a low toxicity profile that may highly improve the therapeutic window of many current drugs and drug candidates.
Collapse
Affiliation(s)
- Maria K. Skytthe
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.K.S.); (S.K.M.)
| | - Jonas Heilskov Graversen
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.K.S.); (S.K.M.)
| | - Søren K. Moestrup
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.K.S.); (S.K.M.)
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| |
Collapse
|
11
|
Tudor DV, Bâldea I, Lupu M, Kacso T, Kutasi E, Hopârtean A, Stretea R, Gabriela Filip A. COX-2 as a potential biomarker and therapeutic target in melanoma. Cancer Biol Med 2020; 17:20-31. [PMID: 32296574 PMCID: PMC7142851 DOI: 10.20892/j.issn.2095-3941.2019.0339] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
With a constantly increasing incidence, cutaneous melanoma has raised the need for a better understanding of its complex microenvironment that may further guide therapeutic options. Melanoma is a model tumor in immuno-oncology. Inflammation represents an important hallmark of cancer capable of inducing and sustaining tumor development. The inflammatory process also orchestrates the adaptative immunosuppression of tumor cells that helps them to evade immune destruction. Besides its role in proliferation, angiogenesis, and apoptosis, cyclooxygenase-2 (COX-2) is a well-known promoter of immune suppression in melanoma. COX-2 inhibitors are closely involved in this condition. This review attempts to answer two controversial questions: is COX-2 a valuable prognostic factor? Among all COX-2 inhibitors, is celecoxib a suitable adjuvant in melanoma therapy?
Collapse
Affiliation(s)
- Diana Valentina Tudor
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Ioana Bâldea
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Mihai Lupu
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Teodor Kacso
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Eniko Kutasi
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Andreea Hopârtean
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Roland Stretea
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| | - Adriana Gabriela Filip
- Department of Physiology, University of Medicine and Pharmacy “Iuliu Hațieganu”, Cluj-Napoca 400000, Romania
| |
Collapse
|
12
|
Etzerodt A, Tsalkitzi K, Maniecki M, Damsky W, Delfini M, Baudoin E, Moulin M, Bosenberg M, Graversen JH, Auphan-Anezin N, Moestrup SK, Lawrence T. Specific targeting of CD163 + TAMs mobilizes inflammatory monocytes and promotes T cell-mediated tumor regression. J Exp Med 2019; 216:2394-2411. [PMID: 31375534 PMCID: PMC6781002 DOI: 10.1084/jem.20182124] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/14/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022] Open
Abstract
Etzerodt et al. show that the specific targeting of CD163+ macrophages in melanoma drives inflammatory monocyte influx and promotes antitumor immunity, illustrating the importance of selective targeting of tumor-associated myeloid cells for achieving optimal therapeutic responses. Tumor-associated macrophages (TAMs) play critical roles in tumor progression but are also capable of contributing to antitumor immunity. Recent studies have revealed an unprecedented heterogeneity among TAMs in both human cancer and experimental models. Nevertheless, we still understand little about the contribution of different TAM subsets to tumor progression. Here, we demonstrate that CD163-expressing TAMs specifically maintain immune suppression in an experimental model of melanoma that is resistant to anti–PD-1 checkpoint therapy. Specific depletion of the CD163+ macrophages results in a massive infiltration of activated T cells and tumor regression. Importantly, the infiltration of cytotoxic T cells was accompanied by the mobilization of inflammatory monocytes that significantly contributed to tumor regression. Thus, the specific targeting of CD163+ TAMs reeducates the tumor immune microenvironment and promotes both myeloid and T cell–mediated antitumor immunity, illustrating the importance of selective targeting of tumor-associated myeloid cells in a therapeutic context.
Collapse
Affiliation(s)
- Anders Etzerodt
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France .,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Maciej Maniecki
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Dermatology, Yale University School of Medicine, New Haven, CT
| | - William Damsky
- Department of Dermatology, Yale University School of Medicine, New Haven, CT
| | | | - Elodie Baudoin
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Morgane Moulin
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France.,Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Marcus Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, CT
| | | | | | - Søren Kragh Moestrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Toby Lawrence
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France .,Centre for Inflammation Biology and Cancer Immunology, School of Immunology and Microbial Sciences, King's College London, London, UK.,Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|