Next Article in Journal
The 2EF Antibody Targets a Unique N-Terminal Epitope of Trop-2 and Enhances the In Vivo Activity of the Cancer-Selective 2G10 Antibody
Next Article in Special Issue
Metabolites and Immune Response in Tumor Microenvironments
Previous Article in Journal
Predictive Risk Score for Acute Kidney Injury in Hematopoietic Stem Cell Transplant
Previous Article in Special Issue
Combination of High-Resolution Structures for the B Cell Receptor and Co-Receptors Provides an Understanding of Their Interactions with Therapeutic Antibodies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Macrophage-Based Therapeutic Strategies in Hematologic Malignancies

1
Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran
2
Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
3
Universal Scientific Education and Research Network (USERN), Tehran 4188783417, Iran
4
Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
5
Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol 4717641367, Iran
6
Faculty of Medicine, Mazandaran University of Medical Sciences, Sari 4815733971, Iran
7
School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran
8
Department of Veterinary Medicine, Shabestar Branch, Islamic Azad University, Shabestar 5381637181, Iran
9
Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
10
Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 1416634793, Iran
11
Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE 106 91 Stockholm, Sweden
*
Authors to whom correspondence should be addressed.
Cancers 2023, 15(14), 3722; https://doi.org/10.3390/cancers15143722
Submission received: 8 June 2023 / Revised: 15 July 2023 / Accepted: 19 July 2023 / Published: 22 July 2023
(This article belongs to the Special Issue Tumor Microenvironment and Treatment Resistance)

Abstract

:

Simple Summary

Tumor-associated macrophages (TAMs) are the most prevalent immunosuppressive myeloid cells in the tumor microenvironment, playing significant functions in the regulation of tumor progression, invasion, and metastatic processes. The M1 and M2-polarized phenotypes of TAMs (immunostimulatory and immunosuppressive myeloid cells, respectively) have been potentially implicated in various cancers and autoimmune diseases. Understanding the precise function of TAMs could improve the assessment of the cancer response to T cell-based treatments and reverse tumor resistance to conventional therapies. Here, the involvement of TAMs in the development of various cancers, mainly hematologic tumors, and their pleiotropic activities are comprehensively discussed.

Abstract

Macrophages are types of immune cells, with ambivalent functions in tumor growth, which depend on the specific environment in which they reside. Tumor-associated macrophages (TAMs) are a diverse population of immunosuppressive myeloid cells that play significant roles in several malignancies. TAM infiltration in malignancies has been linked to a poor prognosis and limited response to treatments, including those using checkpoint inhibitors. Understanding the precise mechanisms through which macrophages contribute to tumor growth is an active area of research as targeting these cells may offer potential therapeutic approaches for cancer treatment. Numerous investigations have focused on anti-TAM-based methods that try to eliminate, rewire, or target the functional mediators released by these cells. Considering the importance of these strategies in the reversion of tumor resistance to conventional therapies and immune modulatory vaccination could be an appealing approach for the immunosuppressive targeting of myeloid cells in the tumor microenvironment (TME). The combination of reprogramming and TAM depletion is a special feature of this approach compared to other clinical strategies. Thus, the present review aims to comprehensively overview the pleiotropic activities of TAMs and their involvement in various stages of cancer development as a potent drug target, with a focus on hematologic tumors.

1. Introduction

Macrophages are among the major cellular components which are involved in numerous tumors. They have remarkable functions in the promotion of tumorigenesis in the tumor microenvironment (TME) through the facilitation of angiogenesis, invasion, metastasis, and immunosuppression [1]. Tumor-associated macrophages (TAMs) are the most prevalent immune-related cells in the TME. They play a substantial role in tumor progression and metastatic processes through various mechanisms [2,3]. Poor survival and a high rate of infiltration are the remarkable properties of macrophages that indicate them as promising targets of anticancer therapies. The efficacy of TAM targeting has already been confirmed in numerous clinical trials [4,5]. Furthermore, the combination of macrophage-directed therapies with other therapies (such as chemotherapies and immunotherapies) has shown complementary effects. Thus, devising novel combined therapeutic strategies requires a good grasp of TAM biology and its intricate interplay with the TME [6].
In this review, we aimed to discuss the diverse roles of macrophages in different cancer development pathways, including cancer initiation, promotion, invasion, metastasis, and angiogenesis. The role of TAMs as diagnostic and prognostic biomarkers is also outlined. The promising application of TAM-based approaches in the treatment of malignancies is also discussed. Moreover, the role of macrophages in hematologic cancers, like acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic lymphocytic leukemia (CLL), is comprehensively discussed. These cancers are characterized by the presence of leukemia-associated macrophages (LAMs).

2. Diversity, Polarization, and Function of TAMs

Macrophages are key immune cells that are derived from monocytic progenitors in the bone marrow and are known as tumor-associated macrophages (TAMs) [5]. Aside from their role in the initiation of inflammatory responses against a stimulus, homeostasis, and the elimination of unnecessary cells, macrophages play various roles in cancer development. The highly plastic macrophages can undergo remarkable changes in their function in response to cues in the TME [7]. In established malignancies, poor prognosis or tumor progression is often strongly associated with a high macrophage infiltration in different tumors, including glioma [8], melanoma [9], breast [10], bladder [11], and prostate cancer [12]. Conversely, high macrophage infiltration is related to better prognosis in colorectal and gastric cancers [13]. This certain discrepancy could be rooted in the functional and phenotypical heterogeneity of macrophages in different types of tumors. TAMs can be broadly classified into two subsets based on their functions. M1-like TAMs (pro-inflammatory and anti-tumor) and M2-like TAMs (anti-inflammatory and pro-tumor) are the major subsets [14]. Lipopolysaccharides, interleukin-1, tumor necrosis factor, and/or granulocyte-macrophage colony-stimulating factor (M-CSF) can actuate the M1-like TAMs. These TAMs can detect and eliminate cancer cells through phagocytosis and cytotoxicity, and initiation of anti-tumor immunity via pro-inflammatory cytokines [15]. On the other hand, M2-like TAMs, which are alternatively activated and induced by various factors within the local microenvironment, can promote tumor growth and TME remodeling through the production of immunosuppressive factors, growth factors, proteases, and pro-angiogenic molecules [16]. The expression of inducible nitric oxide synthase (iNOS) and arginase 1 (ARG1) is suggested to be involved in the intrinsic regulation of macrophage polarization, which could lead to the activation of M1 and M2 macrophages. Tricarboxylic acid (TCA), glutamine (due to its ability to refill TCA cycle metabolites), and serine (that feeds into the one-carbon metabolism) are also involved in macrophage polarization. The amino acids corresponding metabolic pathways, and probable mechanisms of TAMs polarization, are comprehensively discussed by Kieler et al. [17]. Overall, phenotypic switches in TAMs depend on combinational microenvironmental factors, including the action of hypoxia and the availability of cytokines. In addition, the lack of an approved strategy for intratumoral hypoxia measurement remains the main obstacle to a better characterization of hypoxic TAMs. For example, the M1 phenotype is generally generated in LPS-mediated hypoxic responses, while hypoxic TAMs are more strongly associated with an M2-like response [18]. These properties emphasize the necessity of considering hypoxic stress in the context of tumor immunotherapy.

3. Pleiotropic Activities of TAMs in Tumors

Macrophages are specialized to function in specific microenvironments, which contributes to their location in tumor tissues [19]. The tumor stroma, the tumor center, and the boundary between the tumor cells and the stroma, which is called the invasive front, are the three main locations where macrophages can be found [20,21]. It has been indicated that the distribution pattern and distinct functions of macrophages may be related to different cancer progression mechanisms and the location-related signals they receive [22]. For example, in colorectal cancers, M2 phenotype macrophages may be preferentially involved in promoting the movement of cancerous cells in the invasion zone, facilitating metastasis in stromal and perivascular areas, and stimulating angiogenesis in avascular and peri-necrotic hypoxic areas via the induction of S100A8 and S100A9 (calcium-binding proteins correlated with differentiation and metastasis) [23]. Otherwise stated, the distribution pattern of macrophages may be linked to different mechanisms of cancer development. Notably, some gastric cancer cases have been characterized by a stroma-dominant pattern, leading to greater malignancy. This property could be engrained into the aggregation of macrophages in the tumor stroma. It may also contribute to the remodeling of the extracellular matrix (ECM) and stroma activation in conjunction with other elements of the stromal compartment such as matrix metalloproteinase 9, lysyl oxidase, and type IV collagen [24]. Moreover, the ratio of CD163+ to CD68+ macrophages on the invasive front of colorectal cancer has been suggested as a potential prognostic marker. Future studies could focus on exploring the relationship between the varied morphologies of macrophages, tumor positions, and their role in different distribution patterns [25].

4. Macrophages and Cancer Development

The frequent presence of TAMs is often associated with insignificant clinical outcomes in most tumors [5,26], influencing the relapse of tumors after conventional cancer treatments. TAM targeting has garnered a lot of interest as a potential therapeutic strategy and several therapeutic agents that specifically target these cells have been tested in clinical trials. In a recently published review that investigated 300 studies, an obvious relation was identified between the infiltration of macrophages (M1 or M2 subtypes) and the prognosis of various solid cancer types. Specifically, the attending of M2-subtype macrophages was related to a poor outcome, while the presence of M1-subtype macrophages was associated with a favorable prognosis [27]. Hitherto, immune suppression, angiogenesis, chronic inflammation, and invasion/metastasis are the well-characterized tumor-promoting mechanisms of TAMs [5].

4.1. Tumor-Progressing Inflammation and Macrophages

In a healthy state, inflammation is a response to external factors that helps to restore homeostasis [28]. However, chronic inflammation can increase the risk of carcinogenesis. Before the formation of tumors, tumor-promoting inflammation can occur and support tumor growth through suppression of the immune system, promotion of neoangiogenesis (the generation of new vascular networks to supply cancer cells), and oncogenic mutations [29]. Cell death within the tumors could lead to the liberation of damage-associated molecular patterns (DAMPs), like high mobility group box 1 (HMGB1), heat shock proteins (HSPs), or ATP [5,30]. The released DAMPS can activate macrophages and dendritic cells, causing anti-tumor immunity stimulation. Chronic stimulation can result in immunosuppression via the elevated production of interleukin-10 (IL-10), which suppresses the expression of pro-inflammatory cytokines and promotes the formation of regulatory T cells (Tregs) (Figure 1) [31]. In addition, macrophages can contribute to tumor-promoting inflammation through the secretion of immunostimulatory cytokines, such as interleukin-6 (IL-6) [32], and tumor necrosis factor-alpha (TNFα) [33]. These cytokines can stimulate the immune response, backing the tumor growth and survival of cancerous cells [34]. TNFα activates the nuclear factor kappa-B (NF-κB) pathway upon binding to TNFR1/2 receptors. TNFRs (tumor necrosis factor receptors) are membrane proteins that activate cell death. Activation of this pathway could lead to the control of target gene expression (e.g., vascular endothelial growth factor (VEGF) and IL-6) and the stimulation of neo-angiogenesis. IL-6 could then promote cell proliferation and differentiation [35] via the JAK/STAT3 pathway [36].
Another tumoricidal function of TAMs is the blockade of macrophage recruitment by the macrophage migration inhibitor factor (MIF), which is induced via phagocytosis [37] and the propagation of TNFα and interleukin-1 beta (IL1β) [38]. Moreover, the TAM-secreted interleukin-18 and 22 can increase the production of IFNγ and IL-2, which could lead to the increased cytotoxic activity of natural killer cells [39]. The significance of TAMs in tumor development has been highlighted in numerous studies [40]. For instance, the genetic ablation of the Csf1 gene (which encodes M-CSF and is required in macrophage maturation) could lead to the delayed metastasis of mammary carcinoma, whereas the transgenic expression of the M-CSF could speed up pulmonary metastasis [41]. Analogous results have been recognized in thyroid and osteosarcoma cancer cells [42]. These outcomes indicate the existence of an intricate balance between the tumor-promoting or killing functions of TAMs.

4.2. Angiogenesis

Macrophages are essential in the development of cancer due to their ability to promote angiogenesis. Their presence is often correlated with increased blood vessel density in the tumor microenvironment [43]. To support the swift proliferation of malignant cells, the tumor requires a high supply of nutrients and oxygen which are delivered through a capillary network formed during angiogenesis [44]. The released growth factors in the tumor microenvironment are responsible for the regulation of this process. However, the obligation and structure of the newly made vascular tissues are often abnormal due to poor regulation. This property leads to the elevated permeability of vessels and connections to disease development. Considering the elevated rate of cell death in tumor tissues, TAMs are attracted to hypoxic areas to stimulate the formation of new blood vessels [45]. The transcription factor HIF1α (hypoxia-inducible factor α) is an oxygen-dependent transcriptional activator consistently observed in macrophages. It plays critical roles in the angiogenesis of tumors, regulates the response to hypoxic stress (by switching from aerobic to anaerobic metabolism), and induces CCL2, CXCR4, and endothelin expression as HIF1(hypoxia inducible factor-1) target genes [46], which could lead to macrophage recruitment into tumors [47]. In addition, the process of neo-angiogenesis is adjusted by various elements which are produced by TAMs, including platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), and angiopoietin-1 [48]. These factors play a role in the proliferation and maturation of endothelial cells, the chemotaxis of macrophages and ECs [48], and the breakdown of the extracellular matrix (to allow for the formation of novel vascular sprouts) [49]. PDGF released by TAMs and platelets could promote the infiltration of pericytes, which are important for vessel maturation and remodeling [44]. They can also release Angiopoietin-1, which helps to stabilize newly formed vessels by binding to the Tie-2 receptor on endothelial cells. Tie 2 is a tyrosine kinase receptor with a critical function in vascular stability. Tie-2 receptor-expressing monocytes (TEMs) are responsible for enhancing the blood vessel formation of macrophages in tumors and may act as precursors of proangiogenic TAMs [50]. Therapeutic targeting of these pathways may be a potential approach for cancer treatment (Figure 2) [5].

4.3. Role of Macrophages in Tumor Cell Invasion and Metastasis

Tumor cell invasion and metastasis are responsible for failure in cancer treatment and the great number of cancer-related deaths [51,52]. Based on previous pieces of evidence, M2 TAMs can increase the growth, invasion, and metastasis of tumor cells and stimulate angiogenesis; on the other hand, M1 TAMs can provoke anti-tumor effects by producing and secreting pro-inflammatory cytokines and exerting macrophage-mediated cytotoxicity [2,3,53,54,55,56]. Overall, TAM inhibition is considered a promising cancer treatment strategy [54,57]. Yang et al. [54] found an increased infiltration of TAMs, especially M2 macrophages, in both the peritumoral and intertumoral sites of the solid pseudopapillary pancreatic tumor with metastatic features compared to the patients with capsular features [54]. In contrast, Konstantinov et al. [58] found no direct associations between M2 macrophages with metastatic behavior in colorectal cancer. Their results suggest that M2 macrophages could restrict the metastatic processes [58]. TAMs play a substantial role in the metastatic process by contributing to invasion, angiogenesis, intravasation, extravasation, colonization, survival of tumor cells, induction of hypoxia, and pre-metastatic niche formation [2,56,59]. TAMs could suppress CD8⁺ T cell responses by the secretion of interleukin-10 (IL-10) and promote the differentiation of naive CD4⁺ T cells into Treg [57,60]. Activated TAMs produce and secrete various soluble factors, such as tumor-transforming growth factor-β (TGF-β), necrosis factor-α (TNF-α), IL-1β, and IL-8. These factors could ultimately damage the basement membrane of tumor endothelial cells and facilitate epithelial-mesenchymal transition (EMT) processes, which could promote invasion [3,55,61,62]. Furthermore, TAMs, especially M2 macrophages, are capable of extracellular matrix degradation and help tumor cell migration by secreting proteolytic enzymes, including matrix metalloproteinases (MMPs, such as MMP9, MMP7, and MMP2), cathepsins, and serine proteases [3,61,63]. Furthermore, TAMs can enhance invasion, migration, and the circulating tumor cell-mediated metastasis of colorectal cancer by regulating the JAK2/STAT3/miR-506-3p/FoxQ1 axis, which leads to CCL2 production [64]. M2 macrophages also secrete chitinase-3-like protein 1 (CHI3L1), which could promote gastric and breast cancer metastasis by the initiation of the mitogen-activated protein kinase (MAPK) signaling pathway [65]. In addition, TAMs promote tumor angiogenesis through secreting pro-angiogenic factors such as fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF), which facilitate metastasis [2,60]. Intravasation and extravasation of tumor cells are critical steps in metastasis, which are both promoted by TAMs [3]. Triggering the PI3K/Akt survival pathway through engaging vascular cell adhesion molecule-1 (VCAM1) and the secretion of cytokines and chemokines can increase the survival of cancer cells [3,66,67,68,69,70]. In addition, cat eye syndrome chromosome region candidate 2 (CECR2) is an epigenetic regulator which is necessary for breast cancer metastasis [71]. Zhang et al. [71] found an association between CECR2 expression and increased M2 TAMs in the TME, which promotes breast tumor metastasis. They found that CECR2 promotes breast cancer metastasis by regulating M2 TAMs [71].
The pre-metastatic niche (a well-prepared environment in a secondary organ for the colonization of tumor cells) is a specialized environment, including infiltrating immune cells, tumor cells, activated stromal cells, and extracellular matrix regulating tumor progression, while the pre-metastatic niche is an essential requirement for the colonization of circulating pro-tumor cells in a particular organ and metastasis of primary tumors. Macrophages are one of the most remarkable immune cells within pre-metastatic niches [72]. Zhao et al. [73] found that polarized M2 TAMs could induce the formation of pre-metastatic niches. Macrophage depletion could significantly decrease the number of metastatic nodules, which portrays the substantial role of macrophages in tumor metastasis [72,74,75]. Recent studies have evaluated TAM-based cancer treatment; chimeric antigen receptor-macrophage (CAR-M) therapy is a promising cancer treatment with significant outcomes [76,77]. CAR-M is a cancer therapy option in which M2 macrophages are manipulated into the M1 phenotype; as previously pointed out, M1 macrophages have anti-tumor and pro-inflammatory effects [76,78]. According to recent studies, CAR-M therapy could increase overall survival and suppress tumor growth by converting M2 into M1 macrophages, which express pro-inflammatory cytokines and chemokines, resist the effect of immunosuppressive chemokines, present antigens to T cells, upregulate antigen presentation machinery, and prevent metastasis [76,79,80].
The stimulation of cancer cell motility by Wnt5a [81], expression of MMPs via SPARC/Osteonectin, and adjustment of collagen fibers are the other agents causing macrophage-mediated tumor invasion [82,83]. It has been mentioned that activation of the CCL2/CCR2 axis could facilitate the leakage of cancer cells to other areas [83], enhance the secretion of MAM (metastasis-associated macrophage)-derived CCL3, and promote the bone metastasis of prostate cancer [84]. Bone destruction through the activation of osteoclasts could trigger the release of tumor growth factors [85], while the blockade of CCL2 with specific shRNA or neutralizing antibodies could significantly impair bone resorption and prostate cancer-induced formation of osteoclasts [86]. Moreover, the interaction between α4-integrin (expressed by MAMs) and vascular cell adhesion protein 1 in cancer cells could increase lung tumor development [67]. Given these circumstances, the multidimensional function of macrophages could be determined in the metastatic process via clear and explicit evidence.

4.4. TAMs as Diagnostic and Prognostic Target

TAMs have been demonstrated to act as potential biomarkers of cancer stage and progression. Research has demonstrated that the concentration of TAMs in the tumor stroma can predict the size, stage, and metastasis of various tumors. This property could lead to a more accurate prognosis and personalized treatment planning for cancer patients. Patients with higher levels of TAMs have a lower overall survival rate compared to those with lower levels of TAMs. As such, TAMs may be useful for risk assessment, early diagnosis, and prognosis in cancer patients [87]. The quantification of infiltrated TAMs as an essential diagnostic target in various tumors can be accomplished through different morphological methods, cell-surface marker profiling, and gene expression analysis [88]. Although TAMs are mainly recognized as CD68 positive, the AAM (alternatively activated macrophages or alternatively activated M2 macrophages) endotype is distinguished with CD163, CD206, and CD204, while the CAM endotype is distinguished with CD40 [89] and HLA-DR expression [90]. An indication of advanced cancer stages [88], high macrophage density is known as a prognostic marker to estimate chemotherapy results and survival [91]. For instance, failure in Hodgkin lymphoma treatments is correlated with overexpressed macrophages in lymph nodes [92]. Moreover, the prevention of tumor progression through TAM re-education has been considered a clear determinant of the efficiency of postsurgical chemotherapy in pancreatic cancer [88]. Therefore, TAM quantification could be considered a useful approach in patients who are more responsive to chemotherapy.

5. TAMs as a Therapeutic Target

Considering the dual function of TAMs in tumor microenvironments, they have appeared as promising therapeutic targets. Various therapeutic strategies have focused on TAM targeting, aiming to reprogram, deplete, or adjust any TAM-secreted mediators. Early clinical trials have suggested that targeting the checkpoints of myeloid cell function as negative regulators could bear antitumor potential. Macrophages are proper candidates for cell therapy due to the continuous recruitment of myelomonocytic cells into tumor tissues [93]. Overall, ongoing available implements in the oncology armamentarium could be complemented and synergized with macrophage-centered therapeutic strategies (Table 1). The widely applied TAM-based strategies include conventional anticancer therapies (recruitment, repolarization, and depletion), immune checkpoint blockade (ICB), vaccination, cell therapy, and the administration of apoptotic peptides and nanoparticles targeting TAMs.

5.1. TAMs in Conventional Cancer Therapies

Some chemotherapy drugs can stimulate the dissemination of cancer-related molecules through a process called immunogenic cell death. Macrophages are engaged through this process in a beneficial immune response against cancer [176]. Other cancer treatment strategies target macrophages by a reduction in their numbers, like the treatment of ovarian cancer and certain types of sarcomas with an approved marine-derived compound named Trabectedin. The depletion of TAMs is required for the full anti-tumor effect of trabectedin [102]. Some anti-cancer drugs can also change the polarization of TAMs. This change could lead to an increased responsiveness to treatment, such as 5-fluorouracil in colorectal cancer, gemcitabine in pancreatic cancer [88], and high-grade ovarian cancer treated with platinum-based neoadjuvant [177]. Platinum-based neoadjuvant can lead to DNA damage through the production of reactive oxygen species (ROS). In addition, the gut microbiota can stimulate the production of ROS with intratumoral phagocytes, therefore, the effectiveness of these compounds can be enhanced [178].
Since total macrophage depletion is not clinically bearable for a long time [16], strategies have been developed to suppress macrophages via antibodies [126] or small molecule compounds [179]. These strategies involve macrophage targeting with antibodies (such as anti-CSF1R) or small compounds (such as bisphosphonates). These targeting antibodies and molecules inhibit the recruitment of macrophages, deplete their number, and re-educate them [180]. Tumor-infiltrating leukocytes, including TAMs, are fundamental players in the antitumor activity of certain monoclonal antibodies (mAbs). These mAbs trigger the FcγR expressing immune cells to kill tumor cells and perform phagocytosis. The currently prescribed antibodies include rituximab [181], cetuximab [182,183], trastuzumab [184], and daratumumab. Frequently, there is a relation between the density of TAMs and vessels in tumor tissues, which is due to the active responsiveness of TAMs to angiogenic growth factors. VEGF is primarily among these growth factors [185]. Therefore, TAMs modulate the efficiency of antiangiogenic treatments, and VEGF antagonists could remodel the TAM phenotype and induce vascular normalization. TAMs also increase the expression of cysteine cathepsins [186] which aid in the recruitment of monocytes to the TME and support cancer cells from various chemotherapeutic drugs [187]. Chemokine conjugates have been designed which could be activated by enzymes that target TAMs in mouse cancer models. These agents were produced by the conjugation of mCCL2-thiol to cathepsin-activatable fluorophores or caged prodrugs [188]. These probes interact with intracellular cysteine cathepsins in macrophages through CCR2-mediated endocytosis. They also release the cytotoxic chemical doxorubicin for the ablation of macrophages or fluorescently marked active cathepsins in macrophages [189]. Angiogenesis is essential for the development and spread of tumors. Anti-angiogenic methods, particularly anti-VEGF therapy, have been FDA approved for the treatment of various malignancies [190]. A high M2-like/M1-like macrophage ratio has been linked to resistance to anti-VEGF antibody (AVA) therapy, and macrophage M2 polarization may be involved in AVA resistance. Therefore, targeting macrophages may be a potentially innovative approach to overcome AVA treatment resistance in ovarian cancer [191]. The interaction between microseminoprotein (MSMP) and CCR2 could promote adaptive resistance to AVA in ovarian cancer models. BET inhibitor (BETi) is a compound that reduces macrophage recruitment, CCR2 [192], and MSMP expression. This compound could also enhance the efficacy of AVA therapy in ovarian cancer by inducing apoptosis in M2-like macrophages and reprogramming them to have an M1-like phenotype. BETi has been shown to overcome resistance to AVA treatment and increase survival in an adaptive resistance model of ovarian cancer (Figure 3) [193].

5.2. TAMs and the Immune Checkpoint Blockade (ICB)

Immune checkpoint blockade (ICB) immunotherapy, which activates T cell-mediated type 1 immune response, has become a key treatment approach for cancer [194]. However, myelomonocytic cells, which include macrophages, can contribute to primary (before) and adaptive (after) resistance to ICB through the expression of immunosuppressive molecules, such as checkpoint ligands (CD80, CD86, PDL1, PDL2, etc.) and the poliovirus receptor [195,196]. In vivo studies have demonstrated the association of the expressed PDL1 on immune cells (in the TME), with a response to antibodies against PD1 or PDL1 [197]. Interestingly, the expressed PD1 on TAMs has a contrariwise correlation with their ability to phagocytose tumor cells [198,199]. Additionally, other counter-receptors have been expressed on myelomonocytic cells capable of interacting with regulators expressed by natural killer cells and T cells (negative regulators like VISTA) [200]. This molecule could interact with P-selectin glycoprotein ligand 1 (PSGL1) [201] and function as a T cell checkpoint antagonist. The composition of the microbiome can also influence the response to immunotherapy. For anti-PD1 and anti-CTLA4 [202] treatments, the diversity and frequency of gut flora can shape the infiltration of myeloid cells into tumors [203]. The depletion of macrophages has been shown to enhance the effectiveness of different types of immunotherapeutic methods, comprising vaccination [204] and checkpoint inhibitors [205,206]. Multiple clinical trials are currently underway that combine varied TAM-targeted therapeutic approaches (Table 1).

5.3. Targeting of TAMs by Vaccination

Immunomodulatory vaccination is an innovative approach targeting the myeloid cells in TAMs [207]. Anti-regulatory T cells (anti-Tregs) specifically recognize and respond to TAMs by restricting the various immunosuppressive signals they mediate. They could distinguish the HLA-restricted epitopes of arginase, PD-L1, and indoleamine 2,3-dioxygenase (IDO) [208]. Activated anti-Tregs can transform the TME into an immune permissive site. The first IDO vaccinations were conducted in patients with NSCLC (non-small cell lung cancer). A median of 26 months of survival was observed in vaccinated patients, which was significantly longer than the median overall survival of 8 months for the untreated control patients [209]. Currently, there is an industry-sponsored phase II clinical trial underway to test the combination of IDO vaccinations and pembrolizumab as a first-line treatment for non-small cell lung cancer [165]. There is another ongoing phase I trial of PD-L1-based vaccinations in multiple myeloma [210], in which a combination of IDO and PD-L1-specific T cell vaccinations with nivolumab is employed for metastatic melanoma [211]. No observation of toxicity (grade III or IV) indicates good tolerability in vaccinated patients.
Activation of CD8 and CD4 anti-Tregs is essential in the development of therapeutic immune modulatory vaccines. Current cancer vaccine strategies are based on the induction of cancer-specific CD8 cytotoxic T cells, while the immunosuppressive mode is converted to a pro-inflammatory one via activated anti-Tregs. The pro-inflammatory stimulus can convert not terminally differentiated TAMs into M1 macrophages. CD4 cells are particularly effective cytokine-producing cells. Therefore, the activation of CD4 anti-Tregs may be as essential as the activation of CD8 anti-Tregs in a therapeutic process. This is due to the fact that, unlike other strategies of TAM targeting, the activation of anti-Tregs combines both TAM depletion (through direct killing of T cells) and TAM reprogramming (through the provision of pro-inflammatory cytokines in the immune suppressive microenvironment). These processes are crucial in the rebalancing of the microenvironment and enhancing the effectiveness of checkpoint inhibitors such as T cell-enhancing drugs. In many cancer patients, the infiltration of TAMs into the TME majorly contributes to the limited effect of checkpoint inhibitors. Anti-Tregs activated by therapeutic vaccines can result in T cell gathering, Th1 inflammation induction, and elevation of protein expression like IDO and PD-L1 in cancer and immune cells. This creates more targets that could respond to anti-PD1/PD-L1 immunotherapy. Therefore, immune modulatory vaccines that rebalance the microenvironment could increase the effectiveness of T cell-enhancing drugs like checkpoint inhibitors. Combining these vaccines with checkpoint-blocking antibodies could potentially enhance the number of recovered patients [212].

5.4. Macrophage Cell Therapy

The pool of TAMs is continually replenished through the recruitment of circulating monocytes. Macrophage-based cell therapies may have the potential to prevail over this limitation due to the steady influx of mononuclear phagocytes into tumors. These therapies are established based on the modification of mononuclear phagocytes with engineered receptors or the ability of monocytes to deliver nanoparticles or cytokines to the TME. According to an in vivo study, the replenished monocytes with drug-loaded nanoparticles were capable of reaching the tumor cells with higher efficiency compared to free nanoparticles [213]. De Palma et al. explored the possibility of delivering interferon alpha (IFNα) to the tumor cells using macrophages and stimulating an immune-related response [214]. They transduced the Ifna1 gene into hematopoietic progenitors under the control of the Tie2 promoter. These Tie2-expressing monocytes, which had a high affinity for tumors, triumphantly entered tumors, delivered IFNα into the TME, activated immune cells, and inhibited angiogenesis and tumor development [214]. In a similar manner, soft particles called “backpacks” comprising IFNα on their internal side were attached to macrophage surfaces [215]. The study showed that macrophages, which were carrying these backpacks, acquired an M1 phenotype. Moreover, upon intratumoral injection, the phenotype was preserved without being influenced by the immunosuppressive TME. A significant reduction in metastatic tumors was observed in a mouse model treated with macrophages carrying IFNγ backpacks [215]. In a mouse model of sarcoma, pro-metastatic niches were determined by the signature of an immune suppression gene centered on myeloid cells. Genetically engineered myeloid cells expressing interleukin-12 (IL-12) were also seen to trigger a type 1 immune response and reduce primary tumor growth upon adoptive transfer [216].
Tanoto et al. reported an engineered macrophage, named “MacTrigger”, capable of inflammation induction in only tumor tissues. According to evidence, the MacTrigger accelerated the release of TNF-α, natural killer cells, and CD8+T cells, causing efficient effective anti-tumor effects [217]. The major challenge in creating phagocyte-based cellular therapy is the difficulty of transducing human macrophages. This issue was addressed through the development of various technological scaffolds [218,219]. Human CAR-M cells armed with receptors recognizing CD19, CD22, the carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), CD514, and HER2 [220,221], have been developed to target primary and metastatic tumors, mediate phagocytosis, and stably express M1 functions [79]. Clinical trials are ongoing to examine the potency of CAR-M-based therapies in various tumors [93]. In addition to utilizing polarization for cancer therapy, Aalipour et al. reported a new class of cell-based in vivo sensors as highly sensitive cancer diagnostics which was claimed to be more sensitive than both protein and nucleic acid cancer biomarkers. The engineered immune cells as diagnostic sensors can detect tumors as small as 4 mm [217,222].

5.5. Peptides Targeting TAMs

Nanomedicine offers significant advancements in cancer therapy, particularly in terms of improving treatment effectiveness and minimizing side effects. These advantages are achieved through the specific targeting of TAMs. Multiple TAM-specific peptides are currently being examined, including M2pep [168], UNO [169], Melittin [170], RP-182 [171], IL4RPep-1 [223], T4 peptide [173], Pep-20 [174], and CRV [175]. A study by Cieslewicz et al. [224] employed in vitro and in vivo phage peptide display libraries to identify M2pep as a peptide that binds to TAMs. M2pep has demonstrated the ability to reduce TAM levels and enhance the survival of CT26 murine colorectal cancer cells in modeling experiments.
Presently, M2pep is considered a pro-apoptotic peptide and is the primary focus of research on nanocarrier development to deliver CSF-1/CSF-1R inhibitors [168]. Several studies have aimed to improve the stability and targeting capabilities of M2pep [225]. These efforts include modifying M2pep through amino acid substitutions, incorporating decafluorobiphenyl cyclization, and developing a pH-sensitive variant by replacing tyrosine with 3,5-diiodotyrosine [226].

5.6. Nanoparticles Targeting Macrophages

Recent studies have highlighted that nanoparticles (NPs) targeting macrophages offer two main strategies in the battle against cancer. The first strategy focuses on depleting TAMs, aiming to reduce their tumor-promoting effects. The second strategy emphasizes the reprogramming or re-education of TAMs to unleash their inherent anti-tumor potential [227]. Macrophage depletion can be achieved through various approaches, such as targeting the signaling pathway of colony-stimulating factor 1 (CSF1) and its receptor (CSF1R), which prompts apoptosis in a significant proportion of TAMs [143,228].
In addition, blocking the recruitment of circulating inflammatory monocytes to the tumor site is crucial. This recruitment process relies heavily on the signaling pathway of CC-chemokine ligand 2 (CCL2) and its receptor, CC-chemokine receptor 2 (CCR2). By inhibiting the CCL2-CCR2 signaling pathway, the retention of mononuclear cells in the bone marrow occurs, leading to reduced recruitment to both primary and metastatic tumor sites [229]. Reprogramming TAMs is a promising strategy in cancer treatment, reversing their pro-tumor phenotype to an antitumor one. This approach activates M1 macrophages, promoting the activity of cytotoxic T cells and other effector cells. Small molecules and NP formulations, such as TLR agonists, cytokines, antibodies, and RNAs, are also being explored to achieve macrophage repolarization and inhibit cancer growth.
For instance, Xiao et al. discovered that a micellar nano-drug, through the M2-targeting co-delivery of IKKβ siRNA and STAT6 inhibitor AS1517499, effectively repolarized M2-like TAMs into M1-like TAMs. Furthermore, the nano design was tailored to function in the acidic pH of the TME and minimize off-target effects in normal tissues [230]. Furthermore, it was demonstrated that combining nanoparticle-targeted macrophage strategies with other immunotherapies, such as immune checkpoint blockade, yields significant benefits in cancer treatment. For example, in a study by Rodell et al., it was shown that the in vivo delivery of TLR7/8 agonists to TAMs was effectively achieved through the use of R848-loaded β-cyclodextrin nanoparticles, which led to M1 polarization. When combined with the immune checkpoint inhibitor anti-PD-1, the utilization of these nanoparticles resulted in enhanced response rates to immunotherapy, even in a tumor model that exhibited resistance to anti-PD-1 therapy as a standalone treatment [159]. The challenges in NP-based macrophage-targeting therapies involve optimizing timing for NP delivery, addressing the complexity of macrophage subtypes, and understanding NP-cell interactions. While early clinical trials indicate promise, further work is required to ensure NP safety and efficacy, personalize treatments, and bridge the gap between research and clinical applications [231].
In conclusion, the targeting of tumor-associated macrophages (TAMs) represents a promising avenue for improving cancer therapy. Immune checkpoint blockade (ICB) has revolutionized cancer treatment by activating T cell-mediated immune responses, but TAMs can contribute to resistance through immunosuppressive mechanisms. However, strategies targeting TAMs, such as vaccination and macrophage cell therapy, show potential in overcoming this resistance and enhancing treatment outcomes.

6. Macrophage and Hematologic Malignancies

Macrophages residing in the TME of myeloma, lymphoma, or leukemia can provide insights regarding disease progression and the effectiveness of chemotherapy. TAM interactions with other cells in the TME could lead to a pro-tumorigenic environment that includes the promotion of chemo-resistance in cancer cells, stimulation of tumor cell development through the production of growth and matrix remodeling factors, and induction of immunosuppression through influencing the behavior of immune cells [232]. Although the role of TAMs in solid tumors has been under the spotlight in past years, the significance of TAMs in hematologic malignancies has only recently been appreciated, owing to the distinctive and varied microenvironments of these conditions. This review will center on the current preclinical and clinical findings regarding macrophages in hematologic malignancies (Table 2 and Figure 4).

6.1. Macrophage Role in Leukemia

Leukemic stem cells (LSCs) share the survival and functional properties of the hematopoietic stem cell (HSC). These cells and the hematopoietic microenvironment can give rise to persistent leukemia, which cannot be completely eradicated. The presented TAMs in the microenvironment of leukemia are called LAMs [274]. Acute lymphoblastic leukemia (ALL), AML, and CLL are three subtypes of leukemia. Recent studies have focused on the significance of TAMs in cancer development.

6.1.1. ALL

The activation of the CXCR4/CXCL12 axis has been shown to block the polarization of TAMs to the M1 phenotype [241]. Plerixafor is an inhibitor of CXCR4, which is reported to improve clinical scores in T-ALL. In a study by Song et al., 97 bone marrow (BM) samples from patients with acute leukemia (26/97 with ALL) were compared to 30 healthy control samples from individuals with iron-deficiency anemia [275]. The count of CD68-, CD163-, and CD206-positive macrophages was notably higher in the leukemic BM samples in comparison with the control group. These cells significantly decreased after therapy in patients who achieved complete remission. Nevertheless, they remained higher than the control group. Considering the CD68 as a pan-macrophage marker, the CD163+/CD68+ or CD206+/CD68+ ratio was enhanced in the leukemic BM samples, which further supports M2 polarization. Further, the amount of CD163+ cells was an autonomous prognostic issue in these patients. The T-ALL cells co-cultured with M2 macrophages led to significant induction of leukemic cell proliferation through IL-6, growth-related oncogene (GRO)-α, C5a, and TNFα [276]. Hohtari et al. analyzed the immune cell composition in the bone marrow of adult precursor B cell ALL patients. They found an increased amount of M2-like macrophages and myeloid-derived suppressor cells in the BM of ALL patients compared to healthy ones [277]. Various patterns of expressed TAM genes and phenotypes in the BM versus spleen were detected through analysis of multiple lymphoid organs in the Notch1 mouse model with overexpressed T-ALL. It was also demonstrated that splenic TAMs stimulate the growth of T-ALL cells better than bone marrow TAMs [278]. Several studies proposed the efficacy of leukemic cells and TAMs in the TME in the development of ALL. For example, Valencia et al. found that ALL cells release bone morphogenetic protein 4 (BMP4), which can generate M2-like macrophages and induce immunosuppressive dendritic cells. These cells could produce TNFα in low levels and great levels of IL-10, CCL2, and IL-6. [245]. Additionally, a recent report on malignant ALL cells demonstrated that the deletion of stromal interaction molecule 1 (STIM1) and STIM2 restores the pro-inflammatory status of TAMs through IFNγ and reduces the number of infiltrated macrophages [279]. These findings suggest that the interplay between TAMs and leukemic cells and TAMs may be involved in the promotion of tumorigenesis in ALL.

6.1.2. AML

AML is often associated with poor clinical outcomes [280]. One factor that contributes to the high rate of relapse, failure of targeted and traditional treatments, and mortality in AML patients is its resistance to therapy. The mechanisms of resistance in AML treatment are not fully understood. Therefore, finding novel strategies to overcome therapy resistance is essential for successful AML treatment [281]. Previous research has primarily focused on the mechanisms of therapy resistance that are inherent to leukemic cells, such as TP53 mutations. These studies have not extensively examined the mechanisms of acquired resistance that occur through exterior processes [282]. However, recent evidence suggested that the interplay between leukemic cells and other cells in the bone marrow microenvironment (BMME) can lead to acquired therapy resistance in AML.
Recently, Moore et al. found that bone marrow macrophages could decrease the growth of AML in animals through a process called LC3-associated phagocytosis. This process involves the phagocytosis of dying and dead leukemic cells, which includes the mitochondria within the leukemic blasts. These functions could activate the stimulator of IFN genes (STING) and lead to the production of inflammatory signals that enhance phagocytosis and inhibit the expansion of leukemic cells [283,284]. High levels of CD16/CD32 and CD64 are expressed in the spleen and bone marrow macrophages as Fc-activating receptors, which lead to the inhibition of AML through phagocytosis [285]. CD200 is a protein that is overexpressed in AML stem cells (LSCs). This protein can attenuate the response of macrophages to AML [285]. Moreover, treatment with an anti-CD200 antibody can specifically facilitate the phagocytosis of CD200+ AML cells by macrophages through a process called antibody-dependent cell phagocytosis (ADCP) [286]. AML cells that had mutated DNA (cytosine-5)-methyltransferase 3A (DNMT3A) were found to inhibit the polarization of M1 macrophages and resist their killing effect in the laboratory and animal models. In animals with xenografts (transplants of human tumors into mice), the experimental group had significantly larger tumor volumes and a higher proportion of M2 macrophages compared to the control group [287]. Interleukin 4 (IL4) has a powerful anti-leukemic effect in mice by promoting the phagocytosis of AML cells by macrophages. IL4 stimulation leads to the upregulation of CD47 in a STAT6-dependent manner. Moreover, the combination of IL4 stimulation with CD47 blockade further enhances the phagocytosis of AML cells by macrophages [288]. Chenodeoxycholic acid (CDCA) [289] is a type of bile acid, which can inhibit the polarization of M2 macrophages. These cells may have a synergistic effect on reducing the progression of AML. A potential target for chimeric antigen receptor T cell (CAR-T) therapy of AML is the C-type lectin domain family 12 member A (CLEC12A). Its expression level is closely linked to treatment response and patient survival outcomes. The expression of CLEC12A is positively correlated with the infiltration of type 2 macrophages and monocytes [290]. Peritoneal resident macrophages in AML-AF9-induced mice had an M2-like phenotype, which can contribute to cancer progression [291].
Studies determining the function of macrophages (Mφs) in AML have been limited by challenges in accurately distinguishing non-malignant from malignant or AML-associated Mφs. Conventional methods such as immunohistochemistry and flow cytometry have been routinely used for AML patients to determine M2-like Mφs/monocytes in the bone marrow or spleen based on myeloid markers such as CD163 and CD206. Nevertheless, these myeloid markers are also expressed on Mφs, non-malignant monocytes, and AML-associated Mφs. Recently, the detection of mutations and transcript expression discrepancy has been facilitated using single-cell RNA sequencing and genetic profiling. These methods allow for the specific characterization of malignant and non-malignant Mφs within the BMME of AML [292]. Despite improvements in the identification of TAMs/M2-like Mφs, our knowledge of the biology of Mφ in AML has just started to develop. Significant questions remain about the different Mφ groups within the BMME and their role in disease development. Given new technologies like CO-Detection by indEXing (CODEX) [293], the interplay between AML blasts and the surrounding BMME could be visualized. Single-cell sequencing technologies like MacSpectrum employ single-cell RNA sequencing data and can distinguish distinct macrophages derived from bone marrow and adipose tissue. These methods are also contributing to our knowledge of the complex function of macrophages in different diseases, like AML. It is important to comprehensively characterize tissue-resident Mφs and LAMs in the BMME to explore molecular differences for the precise targeting of LAMs. This will be crucial in generating novel Mφ targeting strategies with improved efficacy and declined toxicity. Possible combinations for the treatment of Mφ-mediated therapy resistance, such as selumetinib and/or AZD5991 or CYC065, could be considered as new therapeutic approaches to prevail Mφ- and MCL-1-driven therapy resistance in AML. In light of these facts, the future holds great promise for the development of unprecedented therapies targeting Mφ-mediated immunomodulation in AML [294].

6.1.3. CLL

CLL is a common and frequent type of leukemia in the elderly population. CLL and its related condition, known as small lymphocytic lymphoma (SLL), are recognized as belonging to the category of mature B cell neoplasms by the World Health Organization classification [295]. CLL can range from mild to aggressive in terms of its symptoms, and its treatment can range from a watchful waiting approach to immediate treatment [221]. TAMs, also known as NLCs (Nurse-like cells) in CLL [296], are a part of the TME and resemble M2-polarized macrophages. The level of TAM infiltration has been correlated with a poor prognosis, but this has not yet been proven for CLL [297]. Studies have shown that isolated CLL cells die in vitro, but when co-cultured with NLCs, they can proliferate. This observation suggests that the key to a cure for CLL may lie in the features of the TME and tumor cells. A better mechanistic grasp of the TME could lead to the development of efficient cancer therapies that target its modulation. These therapies could ring about personalized cancer treatments with better tolerance and fewer side effects [298].
The TNFR (tumor necrosis factor receptor) ligand, known as APRIL, has a remarkable role in the proliferation of CLL cells. However, the exact mechanism has not been revealed. Van Attekum et al. examined the role of APRIL in various aspects of CLL biology using a co-culture system with APRIL overexpression, recombinant APRIL, and APRIL reporter cells [299]. They found that APRIL had no effects on the survival of CLL cells in these systems and did not enhance the activation of NF-κB or affect CLL proliferation in single or combined stimuli. Additionally, the survival effect of macrophages on CLL cells was not affected by the APRIL decoy receptor transmembrane activator and CAML interactor-Fc [300]. These results suggest that the direct role of APRIL in CLL cell survival may have been overestimated due to the use of high levels of recombinant APRIL. Nurse-like cells (NLCs), also known as CLL-specific TAMs expressing CD68 and CD163 [301], have been shown to protect the CLL B cells from apoptosis through stromal cell-derived factor-1 [296]. NLC differentiation includes significant DNA methylation changes, which are MEK pathway dependent. MEK inhibitors reduce NLC numbers in vitro and may decrease the number of splenic monocytes/macrophages, which are mainly the M2-like population. The M2-like phenotype was observed in NLCs from high-viability CLL cultures. These cells can attract and facilitate contact with cancer cells, which has been linked to their protective function. In contrast, NLCs from low-viability CLL cell cultures show an M1-like phenotype and do not attract CLL cells. The addition of IL-10 to the culture can induce an M2-like phenotype in NLCs and increase CLL cell viability. On the other hand, TNF can depolarize protective M2-like NLCs into non-protective M1-like NLCs. IL-10 can repolarize TNF-depolarized NLCs and restore their protective effect on CLL cells [302].

6.2. Macrophage Role in Lymphoma

The progression of lymphoma could be supported by macrophages both in classic Hodgkin’s lymphoma (CHL) and non-Hodgkin’s lymphoma (NHL) (Figure 4).

6.2.1. Hodgkin Lymphoma

Studies have shown that the presence of CD163+ macrophages in tumor tissue is related to poor survival in patients with classical Hodgkin lymphoma (CHL) [303]. Additionally, a lower number of TAMs in CHL samples was correlated with a higher progression-free survival rate [304]. The lower level of M2 macrophages has been linked to a complete response and better survival [305]. The expressions of both TAM markers, CD68, and CD163 [306] are essential predictors of complete remission in CHL patients [307]. A high ratio of LAMs to Hodgkin–Reed–Sternberg cells at diagnosis is associated with a higher risk of CHL progression or death [308].

6.2.2. Non-Hodgkin Lymphoma

The number of CD68+ and CD163+ macrophages significantly increases in all three grades of follicular lymphoma [309]. A high PD-1 expression on TAMs in the T cells of non-Hodgkin lymphoma may predict a poor prognosis. It enhances the pro-tumor effects of the TME and inhibits the polarization of M1 macrophages and phagocytosis [310]. A high M2 TAM content at diagnosis, particularly in combination with an international prognostic index, may be a factor in the identification of diffused large B cell lymphoma patients [311]. GM-CSF amplifies the inhibitory effect of CHOP chemotherapy on DLBCL progression by promoting the polarization of M1 macrophages [312]. Enhanced M2 macrophage activation and lipid metabolism have been observed in the immunosuppressive tumor microenvironment of non-MYC/BCL2 double express or DLBCL [313]. The LXRα-related signaling pathways and functions are connected to M1 polarization and may increase the immune reactivity of macrophages in DLBCL [314].

6.3. Macrophage Role in Multiple Myeloma

Macrophages are a type of immune cells that are prevalent in the bone marrow of individuals with multiple myeloma (MM) and can support the proliferation, induce drug resistance, and contribute to the formation of an immunosuppressive environment. Beider et al. demonstrated that the interactions between macrophages and MM tumor cells result in the polarization of macrophages toward an M2 phenotype [315]. This process increases the production of CXCL13 and activates osteoclasts, which have the ability to resorb bone and promote MM progression. IL-32γ can promote drug resistance in MM through macrophages and modify macrophages towards an M2 phenotype [316]. Increased TAMs in MM patients can stop the functions of cytotoxic T lymphocytes (through the PD-1/PD-L1 pathway) and contribute to the evasion of the immune system by myeloma cells [317]. Exosomes, derived from MM containing IL-32γ, can increase the expression of PD-L1 by macrophages and lead to immune evasion. The PFKFB3-JAK1(6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3-Janus kinase 1) axis may also play a role in the expression of PD-L1 by macrophages [318]. Gao et al. found that daratumumab (DAR) has a significant anti-tumor effect on MM in mice through its interaction with macrophages via Fc-FcγR. DAR induces the activation of macrophages in mice and results in the phagocytosis of cancer cells through the Fc-FcγR interaction [319]. RGS12 (regulator of G-protein signaling 12) can inhibit the progression and metastasis of MM by the induction of M1 macrophage polarization and activation in the bone marrow microenvironment (BMME) (Figure 4) [320].

7. Challenges in TAM-Based Therapeutics (in Solid or Hematologic Tumors)

The reduction in negative side effects with TAM-based strategies is an ongoing challenge. Due to the complicated functions of macrophages in maintaining homeostasis, TAM depletion could increase the risk of infections or disorganize tissue-resident cells and prevent them from performing their usual functions. Therefore, discovering TAM-specific molecules or markers that are mainly created through metastasis-associated macrophages (MAMs) and/or activated M2 (AAMs) will enable therapeutic approaches to specifically target tumor cells without affecting the normal function of other immune cells which are tissue-resident [321]. The potency of wound healing and phagocytosis in non-tumor tissues should be preserved as a goal of the techniques which target macrophage reprogramming.

8. Conclusions

Macrophages are endowed with the ability to adapt and change their function based on external stimuli. They are prevalent in the TME and have an indispensable role in cancer progression. Various efforts have been made to alter the behavior of TAMs and inhibit their functions in the promotion of tumor growth. However, the ability of macrophages to travel to both primary tumors and metastatic sites presents an opportunity for their application as a means for the delivery of cellular therapies to cancer cells. As antigen-presenting cells, macrophages link innate immune responses with adaptive immunity. The development of gene engineering techniques, such as the use of Vpx-LV and Ad5f35 as vectors for modification of primary human macrophages, has opened the possibility of redirecting the function of these cells against tumors through synthetic biology. In addition, immune modulatory vaccines, which target TAMs in the TME, have emerged as an alternative to traditional antibodies or small molecule inhibitors and have shown promise in preclinical and clinical trials.

Author Contributions

Conceptualization, Z.P., S.K., F.A., F.Z. and B.K.; Writing—Original Draft Preparation, K.B., M.S.Z., Y.E. and K.F.; Writing—Review and Editing, N.A., A.A., S.K., F.A. and B.K.; Visualization, A.M.F., S.R.T. and A.F.N.; Supervision, Z.P.; Project Administration, Z.P. and F.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest. This study received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

References

  1. Qian, B.-Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [Green Version]
  2. Fu, L.Q.; Du, W.L.; Cai, M.H.; Yao, J.Y.; Zhao, Y.Y.; Mou, X.Z. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell. Immunol. 2020, 353, 104119. [Google Scholar] [CrossRef]
  3. Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef]
  4. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef]
  5. Poh, A.R.; Ernst, M. Targeting macrophages in cancer: From bench to bedside. Front. Oncol. 2018, 8, 49. [Google Scholar] [CrossRef] [Green Version]
  6. Petty, A.J.; Owen, D.H.; Yang, Y.; Huang, X. Targeting tumor-associated macrophages in cancer immunotherapy. Cancers 2021, 13, 5318. [Google Scholar] [CrossRef]
  7. Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef] [Green Version]
  8. Nishie, A.; Ono, M.; Shono, T.; Fukushi, J.; Otsubo, M.; Onoue, H.; Ito, Y.; Inamura, T.; Ikezaki, K.; Fukui, M. Macrophage infiltration and heme oxygenase-1 expression correlate with angiogenesis in human gliomas. Clin. Cancer Res. 1999, 5, 1107–1113. [Google Scholar]
  9. Torisu, H.; Ono, M.; Kiryu, H.; Furue, M.; Ohmoto, Y.; Nakayama, J.; Nishioka, Y.; Sone, S.; Kuwano, M. Macrophage infiltration correlates with tumor stage and angiogenesis in human malignant melanoma: Possible involvement of TNFα and IL-1α. Int. J. Cancer 2000, 85, 182–188. [Google Scholar] [CrossRef]
  10. Zhang, Y.; Cheng, S.; Zhang, M.; Zhen, L.; Pang, D.; Zhang, Q.; Li, Z. High-infiltration of tumor-associated macrophages predicts unfavorable clinical outcome for node-negative breast cancer. PLoS ONE 2013, 8, e76147. [Google Scholar] [CrossRef]
  11. Xue, Y.; Tong, L.; LiuAnwei Liu, F.; Liu, A.; Zeng, S.; Xiong, Q.; Yang, Z.; He, X.; Sun, Y.; Xu, C. Tumor-infiltrating M2 macrophages driven by specific genomic alterations are associated with prognosis in bladder cancer. Oncol. Rep. 2019, 42, 581–594. [Google Scholar] [CrossRef]
  12. Cao, J.; Liu, J.; Xu, R.; Zhu, X.; Zhao, X.; Qian, B.-Z. Prognostic role of tumour-associated macrophages and macrophage scavenger receptor 1 in prostate cancer: A systematic review and meta-analysis. Oncotarget 2017, 8, 83261. [Google Scholar] [CrossRef]
  13. Cortese, N.; Carriero, R.; Laghi, L.; Mantovani, A.; Marchesi, F. Prognostic significance of tumor-associated macrophages: Past, present and future. In Seminars in Immunology; Academic Press: Cambridge, MA, USA, 2020; p. 101408. [Google Scholar]
  14. Rhee, I. Diverse macrophages polarization in tumor microenvironment. Arch. Pharm. Res. 2016, 39, 1588–1596. [Google Scholar] [CrossRef]
  15. Callens, C.; Coulon, S.; Naudin, J.; Radford-Weiss, I.; Boissel, N.; Raffoux, E.; Wang, P.H.M.; Agarwal, S.; Tamouza, H.; Paubelle, E. Targeting iron homeostasis induces cellular differentiation and synergizes with differentiating agents in acute myeloid leukemia. J. Exp. Med. 2010, 207, 731–750. [Google Scholar] [CrossRef]
  16. Cassetta, L.; Pollard, J.W. Targeting macrophages: Therapeutic approaches in cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef]
  17. Kieler, M.; Hofmann, M.; Schabbauer, G. More than just protein building blocks: How amino acids and related metabolic pathways fuel macrophage polarization. FEBS J. 2021, 288, 3694–3714. [Google Scholar] [CrossRef]
  18. Henze, A.-T.; Mazzone, M. The impact of hypoxia on tumor-associated macrophages. J. Clin. Investig. 2016, 126, 3672–3679. [Google Scholar] [CrossRef]
  19. Komohara, Y.; Jinushi, M.; Takeya, M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci. 2014, 105, 1–8. [Google Scholar] [CrossRef] [Green Version]
  20. Lee, J.L.; Roh, S.A.; Kim, C.W.; Kwon, Y.H.; Ha, Y.J.; Kim, S.-K.; Kim, S.-Y.; Cho, D.-H.; Kim, Y.S.; Kim, J.C. Clinical assessment and identification of immuno-oncology markers concerning the 19-gene based risk classifier in stage IV colorectal cancer. World J. Gastroenterol. 2019, 25, 1341. [Google Scholar] [CrossRef]
  21. Liu, J.; Geng, X.; Hou, J.; Wu, G. New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 2021, 21, 1–7. [Google Scholar] [CrossRef]
  22. Zhou, D.; Huang, C.; Lin, Z.; Zhan, S.; Kong, L.; Fang, C.; Li, J. Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell. Signal. 2014, 26, 192–197. [Google Scholar] [CrossRef]
  23. Lim, S.Y.; Yuzhalin, A.E.; Gordon-Weeks, A.N.; Muschel, R.J. Tumor-infiltrating monocytes/macrophages promote tumor invasion and migration by upregulating S100A8 and S100A9 expression in cancer cells. Oncogene 2016, 35, 5735–5745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Peng, C.; Liu, J.; Yang, G.; Li, Y. Lysyl oxidase activates cancer stromal cells and promotes gastric cancer progression: Quantum dot-based identification of biomarkers in cancer stromal cells. Int. J. Nanomed. 2018, 13, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yang, C.; Wei, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Dou, R.; Xiong, B. Elevated CD163+/CD68+ ratio at tumor invasive front is closely associated with aggressive phenotype and poor prognosis in colorectal cancer. Int. J. Biol. Sci. 2019, 15, 984–998. [Google Scholar] [CrossRef] [Green Version]
  26. van Furth, R.; Cohn, Z.; Hirsch, J.; Humphrey, J.; Spector, W.; Langevoort, H. The mononuclear phagocyte system: A new classification of macrophages, monocytes, and their precursor cells. Bull. World Health Organ. 1972, 46, 845. [Google Scholar] [PubMed]
  27. Bruni, D.; Angell, H.K.; Galon, J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat. Rev. Cancer 2020, 20, 662–680. [Google Scholar] [CrossRef] [PubMed]
  28. Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic inflammation and cytokines in the tumor microenvironment. J. Immunol. Res. 2014, 2014, 149185. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, J.; Li, D.; Cang, H.; Guo, B. Crosstalk between cancer and immune cells: Role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019, 8, 4709–4721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Greten, F.R.; Grivennikov, S.I. Inflammation and cancer: Triggers, mechanisms, and consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  31. Cook, R.S.; Jacobsen, K.M.; Wofford, A.M.; DeRyckere, D.; Stanford, J.; Prieto, A.L.; Redente, E.; Sandahl, M.; Hunter, D.M.; Strunk, K.E. MerTK inhibition in tumor leukocytes decreases tumor growth and metastasis. J. Clin. Investig. 2013, 123, 3231–3242. [Google Scholar] [CrossRef] [Green Version]
  32. Xia, L.; Tan, S.; Zhou, Y.; Lin, J.; Wang, H.; Oyang, L.; Tian, Y.; Liu, L.; Su, M.; Wang, H. Role of the NFκB-signaling pathway in cancer. OncoTargets Ther. 2018, 11, 2063. [Google Scholar] [CrossRef] [Green Version]
  33. Baker, K.J.; Houston, A.; Brint, E. IL-1 family members in cancer; two sides to every story. Front. Immunol. 2019, 10, 1197. [Google Scholar] [CrossRef] [Green Version]
  34. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Cendrowicz, E.; Sas, Z.; Bremer, E.; Rygiel, T.P. The role of macrophages in cancer development and therapy. Cancers 2021, 13, 1946. [Google Scholar] [CrossRef] [PubMed]
  36. Oh, S.A.; Li, M.O. TGF-β: Guardian of T cell function. J. Immunol. 2013, 191, 3973–3979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Onodera, S.; Suzuki, K.; Matsuno, T.; Kaneda, K.; Takagi, M.; Nishihira, J. Macrophage migration inhibitory factor induces phagocytosis of foreign particles by macrophages in autocrine and paracrine fashion. Immunology 1997, 92, 131–137. [Google Scholar] [CrossRef]
  38. Pozzi, L.-A.M.; Weiser, W.Y. Human recombinant migration inhibitory factor activates human macrophages to kill tumor cells. Cell. Immunol. 1992, 145, 372–379. [Google Scholar] [CrossRef]
  39. Netea, M.G.; Kullberg, B.J.; Verschueren, I.; Meer, J.W.V.d. Interleukin-18 induces production of proinflammatory cytokines in mice: No intermediate role for the cytokines of the tumor necrosis factor family and interleukin-1β. Eur. J. Immunol. 2000, 30, 3057–3060. [Google Scholar] [CrossRef]
  40. Pyonteck, S.; Gadea, B.; Wang, H.; Gocheva, V.; Hunter, K.; Tang, L.; Joyce, J. Deficiency of the macrophage growth factor CSF-1 disrupts pancreatic neuroendocrine tumor development. Oncogene 2012, 31, 1459–1467. [Google Scholar] [CrossRef] [Green Version]
  41. Lin, E.Y.; Nguyen, A.V.; Russell, R.G.; Pollard, J.W. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J. Exp. Med. 2001, 193, 727–740. [Google Scholar] [CrossRef]
  42. Ryder, M.; Gild, M.; Hohl, T.M.; Pamer, E.; Knauf, J.; Ghossein, R.; Joyce, J.A.; Fagin, J.A. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid cancer progression. PLoS ONE 2013, 8, e54302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hanahan, D.; Christofori, G.; Naik, P.; Arbeit, J. Transgenic mouse models of tumour angiogenesis: The angiogenic switch, its molecular controls, and prospects for preclinical therapeutic models. Eur. J. Cancer 1996, 32, 2386–2393. [Google Scholar] [CrossRef] [PubMed]
  44. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Murdoch, C.; Muthana, M.; Coffelt, S.B.; Lewis, C.E. The role of myeloid cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer 2008, 8, 618–631. [Google Scholar] [CrossRef]
  46. Murdoch, C.; Giannoudis, A.; Lewis, C.E. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 2004, 104, 2224–2234. [Google Scholar] [CrossRef] [Green Version]
  47. De Palma, M.; Venneri, M.A.; Galli, R.; Sergi, L.S.; Politi, L.S.; Sampaolesi, M.; Naldini, L. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 2005, 8, 211–226. [Google Scholar] [CrossRef] [Green Version]
  48. Riabov, V.; Gudima, A.; Wang, N.; Mickley, A.; Orekhov, A.; Kzhyshkowska, J. Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front. Physiol. 2014, 5, 75. [Google Scholar] [CrossRef] [Green Version]
  49. Quintero-Fabián, S.; Arreola, R.; Becerril-Villanueva, E.; Torres-Romero, J.C.; Arana-Argáez, V.; Lara-Riegos, J.; Ramírez-Camacho, M.A.; Alvarez-Sánchez, M.E. Role of matrix metalloproteinases in angiogenesis and cancer. Front. Oncol. 2019, 9, 1370. [Google Scholar] [CrossRef] [Green Version]
  50. Giroux, V.; Rustgi, A.K. Metaplasia: Tissue injury adaptation and a precursor to the dysplasia–cancer sequence. Nat. Rev. Cancer 2017, 17, 594–604. [Google Scholar] [CrossRef]
  51. Stoletov, K.; Beatty, P.H.; Lewis, J.D. Novel therapeutic targets for cancer metastasis. Expert Rev. Anticancer Ther. 2020, 20, 97–109. [Google Scholar] [CrossRef] [Green Version]
  52. Qian, C.N.; Mei, Y.; Zhang, J. Cancer metastasis: Issues and challenges. Chin. J. Cancer 2017, 36, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xiong, K.; Qi, M.; Stoeger, T.; Zhang, J.; Chen, S. The role of tumor-associated macrophages and soluble mediators in pulmonary metastatic melanoma. Front. Immunol. 2022, 13, 1000927. [Google Scholar] [CrossRef] [PubMed]
  54. Yang, J.; Tan, C.L.; Long, D.; Liang, Y.; Zhou, L.; Liu, X.B.; Chen, Y.H. Analysis of invasiveness and tumor-associated macrophages infiltration in solid pseudopapillary tumors of pancreas. World J. Gastroenterol. 2022, 28, 5047–5057. [Google Scholar] [CrossRef] [PubMed]
  55. Fu, X.T.; Dai, Z.; Song, K.; Zhang, Z.J.; Zhou, Z.J.; Zhou, S.L.; Zhao, Y.M.; Xiao, Y.S.; Sun, Q.M.; Ding, Z.B.; et al. Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway. Int. J. Oncol. 2015, 46, 587–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef] [PubMed]
  57. Orfali, R.; Rateb, M.E.; Hassan, H.M.; Alonazi, M.; Gomaa, M.R.; Mahrous, N.; GabAllah, M.; Kandeil, A.; Perveen, S.; Abdelmohsen, U.R. Sinapic Acid Suppresses SARS CoV-2 Replication by Targeting Its Envelope Protein. Antibiotics 2021, 10, 420. [Google Scholar] [CrossRef]
  58. Konstantinov, A.S.; Kovaleva, O.V.; Samoilova, D.V.; Shelekhova, K.V. Role of macrophages in progression of colorectal cancer: A contrast with the traditional paradigm. Int. J. Clin. Exp. Pathol. 2022, 15, 403–411. [Google Scholar]
  59. Fridrichova, I.; Kalinkova, L.; Ciernikova, S. Clinical Relevancy of Circulating Tumor Cells in Breast Cancer: Epithelial or Mesenchymal Characteristics, Single Cells or Clusters? Int. J. Mol. Sci. 2022, 23, 12141. [Google Scholar] [CrossRef]
  60. El-Kenawi, A.; Hänggi, K.; Ruffell, B. The Immune Microenvironment and Cancer Metastasis. Cold Spring Harb. Perspect. Med. 2020, 10, a036541. [Google Scholar] [CrossRef]
  61. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  62. Wang, H.; Fang, R.; Wang, X.F.; Zhang, F.; Chen, D.Y.; Zhou, B.; Wang, H.S.; Cai, S.H.; Du, J. Stabilization of Snail through AKT/GSK-3β signaling pathway is required for TNF-α-induced epithelial-mesenchymal transition in prostate cancer PC3 cells. Eur. J. Pharmacol. 2013, 714, 48–55. [Google Scholar] [CrossRef] [PubMed]
  63. Annamalai, R.T.; Turner, P.A.; Carson, W.F.t.; Levi, B.; Kunkel, S.; Stegemann, J.P. Harnessing macrophage-mediated degradation of gelatin microspheres for spatiotemporal control of BMP2 release. Biomaterials 2018, 161, 216–227. [Google Scholar] [CrossRef] [PubMed]
  64. Wei, C.; Yang, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Liu, Q.; Dou, R.; Xiong, B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol. Cancer 2019, 18, 64. [Google Scholar] [CrossRef]
  65. Chen, Y.; Zhang, S.; Wang, Q.; Zhang, X. Tumor-recruited M2 macrophages promote gastric and breast cancer metastasis via M2 macrophage-secreted CHI3L1 protein. J. Hematol. Oncol. 2017, 10, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Li, D.; Ji, H.; Niu, X.; Yin, L.; Wang, Y.; Gu, Y.; Wang, J.; Zhou, X.; Zhang, H.; Zhang, Q. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. 2020, 111, 47–58. [Google Scholar] [CrossRef] [Green Version]
  67. Chen, Q.; Zhang, X.H.; Massagué, J. Macrophage binding to receptor VCAM-1 transmits survival signals in breast cancer cells that invade the lungs. Cancer Cell 2011, 20, 538–549. [Google Scholar] [CrossRef] [Green Version]
  68. Gil-Bernabé, A.M.; Ferjancic, S.; Tlalka, M.; Zhao, L.; Allen, P.D.; Im, J.H.; Watson, K.; Hill, S.A.; Amirkhosravi, A.; Francis, J.L.; et al. Recruitment of monocytes/macrophages by tissue factor-mediated coagulation is essential for metastatic cell survival and premetastatic niche establishment in mice. Blood 2012, 119, 3164–3175. [Google Scholar] [CrossRef] [Green Version]
  69. Sano, M.; Takahashi, R.; Ijichi, H.; Ishigaki, K.; Yamada, T.; Miyabayashi, K.; Kimura, G.; Mizuno, S.; Kato, H.; Fujiwara, H.; et al. Blocking VCAM-1 inhibits pancreatic tumour progression and cancer-associated thrombosis/thromboembolism. Gut 2021, 70, 1713–1723. [Google Scholar] [CrossRef]
  70. Ye, H.; Zhou, Q.; Zheng, S.; Li, G.; Lin, Q.; Wei, L.; Fu, Z.; Zhang, B.; Liu, Y.; Li, Z.; et al. Tumor-associated macrophages promote progression and the Warburg effect via CCL18/NF-kB/VCAM-1 pathway in pancreatic ductal adenocarcinoma. Cell Death Dis. 2018, 9, 453. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, M.; Liu, Z.Z.; Aoshima, K.; Cai, W.L.; Sun, H.; Xu, T.; Zhang, Y.; An, Y.; Chen, J.F.; Chan, L.H.; et al. CECR2 drives breast cancer metastasis by promoting NF-κB signaling and macrophage-mediated immune suppression. Sci. Transl. Med. 2022, 14, eabf5473. [Google Scholar] [CrossRef]
  72. Morrissey, S.M.; Zhang, F.; Ding, C.; Montoya-Durango, D.E.; Hu, X.; Yang, C.; Wang, Z.; Yuan, F.; Fox, M.; Zhang, H.G.; et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021, 33, 2040–2058. [Google Scholar] [CrossRef] [PubMed]
  73. Zhao, S.; Mi, Y.; Guan, B.; Zheng, B.; Wei, P.; Gu, Y.; Zhang, Z.; Cai, S.; Xu, Y.; Li, X.; et al. Tumor-derived exosomal miR-934 induces macrophage M2 polarization to promote liver metastasis of colorectal cancer. J. Hematol. Oncol. 2020, 13, 156. [Google Scholar] [CrossRef] [PubMed]
  74. Bader, J.E.; Enos, R.T.; Velázquez, K.T.; Carson, M.S.; Nagarkatti, M.; Nagarkatti, P.S.; Chatzistamou, I.; Davis, J.M.; Carson, J.A.; Robinson, C.M.; et al. Macrophage depletion using clodronate liposomes decreases tumorigenesis and alters gut microbiota in the AOM/DSS mouse model of colon cancer. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 314, G22–G31. [Google Scholar] [CrossRef]
  75. Linde, N.; Casanova-Acebes, M.; Sosa, M.S.; Mortha, A.; Rahman, A.; Farias, E.; Harper, K.; Tardio, E.; Reyes Torres, I.; Jones, J.; et al. Macrophages orchestrate breast cancer early dissemination and metastasis. Nat. Commun. 2018, 9, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kumari, N.; Choi, S.H. Tumor-associated macrophages in cancer: Recent advancements in cancer nanoimmunotherapies. J. Exp. Clin. Cancer Res. CR 2022, 41, 68. [Google Scholar] [CrossRef]
  77. Romero, D. Immunotherapy: CAR T cells ready to go mainstream. Nat. Rev. Clin. Oncol. 2016, 13, 396–397. [Google Scholar] [CrossRef]
  78. Zhang, L.; Tian, L.; Dai, X.; Yu, H.; Wang, J.; Lei, A.; Zhu, M.; Xu, J.; Zhao, W.; Zhu, Y.; et al. Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions. J. Hematol. Oncol. 2020, 13, 153. [Google Scholar] [CrossRef]
  79. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E.; et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef]
  80. Niu, Z.; Chen, G.; Chang, W.; Sun, P.; Luo, Z.; Zhang, H.; Zhi, L.; Guo, C.; Chen, H.; Yin, M.; et al. Chimeric antigen receptor-modified macrophages trigger systemic anti-tumour immunity. J. Pathol. 2021, 253, 247–257. [Google Scholar] [CrossRef]
  81. Pukrop, T.; Klemm, F.; Hagemann, T.; Gradl, D.; Schulz, M.; Siemes, S.; Trümper, L.; Binder, C. Wnt 5a signaling is critical for macrophage-induced invasion of breast cancer cell lines. Proc. Natl. Acad. Sci. USA 2006, 103, 5454–5459. [Google Scholar] [CrossRef]
  82. Gilles, C.; Bassuk, J.A.; Pulyaeva, H.; Sage, E.H.; Foidart, J.-M.; Thompson, E.W. SPARC/osteonectin induces matrix metalloproteinase 2 activation in human breast cancer cell lines. Cancer Res. 1998, 58, 5529–5536. [Google Scholar] [PubMed]
  83. Kitamura, T.; Qian, B.-Z.; Soong, D.; Cassetta, L.; Noy, R.; Sugano, G.; Kato, Y.; Li, J.; Pollard, J.W. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J. Exp. Med. 2015, 212, 1043–1059. [Google Scholar] [CrossRef] [PubMed]
  84. Mizutani, K.; Sud, S.; McGregor, N.A.; Martinovski, G.; Rice, B.T.; Craig, M.J.; Varsos, Z.S.; Roca, H.; Pienta, K.J. The chemokine CCL2 increases prostate tumor growth and bone metastasis through macrophage and osteoclast recruitment. Neoplasia 2009, 11, 1235–1242. [Google Scholar] [CrossRef] [Green Version]
  85. Westendorf, J.J.; Kahler, R.A.; Schroeder, T.M. Wnt signaling in osteoblasts and bone diseases. Gene 2004, 341, 19–39. [Google Scholar] [CrossRef]
  86. Lu, Y.; Chen, Q.; Corey, E.; Xie, W.; Fan, J.; Mizokami, A.; Zhang, J. Activation of MCP-1/CCR2 axis promotes prostate cancer growth in bone. Clin. Exp. Metastasis 2009, 26, 161–169. [Google Scholar] [CrossRef] [PubMed]
  87. Räihä, M.R.; Puolakkainen, P.A. Tumor-associated macrophages (TAMs) as biomarkers for gastric cancer: A review. Chronic Dis. Transl. Med. 2018, 4, 156–163. [Google Scholar] [CrossRef]
  88. Di Caro, G.; Cortese, N.; Castino, G.F.; Grizzi, F.; Gavazzi, F.; Ridolfi, C.; Capretti, G.; Mineri, R.; Todoric, J.; Zerbi, A. Dual prognostic significance of tumour-associated macrophages in human pancreatic adenocarcinoma treated or untreated with chemotherapy. Gut 2016, 65, 1710–1720. [Google Scholar] [CrossRef]
  89. Vogel, D.Y.; Glim, J.E.; Stavenuiter, A.W.; Breur, M.; Heijnen, P.; Amor, S.; Dijkstra, C.D.; Beelen, R.H. Human macrophage polarization in vitro: Maturation and activation methods compared. Immunobiology 2014, 219, 695–703. [Google Scholar] [CrossRef]
  90. Buchacher, T.; Ohradanova-Repic, A.; Stockinger, H.; Fischer, M.B.; Weber, V. M2 polarization of human macrophages favors survival of the intracellular pathogen Chlamydia pneumoniae. PLoS ONE 2015, 10, e0143593. [Google Scholar] [CrossRef] [Green Version]
  91. Forssell, J.; Öberg, A.K.; Henriksson, M.L.; Stenling, R.; Jung, A.; Palmqvist, R. High macrophage infiltration along the tumor front correlates with improved survival in colon cancer. Clin. Cancer Res. 2007, 13, 1472–1479. [Google Scholar] [CrossRef] [Green Version]
  92. Steidl, C.; Lee, T.; Shah, S.P.; Farinha, P.; Han, G.; Nayar, T.; Delaney, A.; Jones, S.J.; Iqbal, J.; Weisenburger, D.D. Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N. Engl. J. Med. 2010, 362, 875–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Mantovani, A.; Allavena, P.; Marchesi, F.; Garlanda, C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 2022, 21, 799–820. [Google Scholar] [CrossRef] [PubMed]
  94. Tian, L.; Yi, X.; Dong, Z.; Xu, J.; Liang, C.; Chao, Y.; Wang, Y.; Yang, K.; Liu, Z. Calcium bisphosphonate nanoparticles with chelator-free radiolabeling to deplete tumor-associated macrophages for enhanced cancer radioisotope therapy. ACS Nano 2018, 12, 11541–11551. [Google Scholar] [CrossRef] [PubMed]
  95. Tap, W. ENLIVEN study: Pexidartinib for tenosynovial giant cell tumor (TGCT). Future Oncol. 2020, 16, 1875–1878. [Google Scholar] [CrossRef]
  96. Manji, G.A.; Van Tine, B.A.; Lee, S.M.; Raufi, A.G.; Pellicciotta, I.; Hirbe, A.C.; Pradhan, J.; Chen, A.; Rabadan, R.; Schwartz, G.K. A phase I study of the combination of pexidartinib and sirolimus to target tumor-associated macrophages in unresectable sarcoma and malignant peripheral nerve sheath tumors. Clin. Cancer Res. 2021, 27, 5519–5527. [Google Scholar] [CrossRef]
  97. Huang, Z.; Yao, D.; Ye, Q.; Jiang, H.; Gu, R.; Ji, C.; Wu, J.; Hu, Y.; Yuan, A. Zoledronic Acid–Gadolinium Coordination Polymer Nanorods for Improved Tumor Radioimmunotherapy by Synergetically Inducing Immunogenic Cell Death and Reprogramming the Immunosuppressive Microenvironment. ACS Nano 2021, 15, 8450–8465. [Google Scholar] [CrossRef]
  98. Barone, A.; Chi, D.-C.; Theoret, M.R.; Chen, H.; He, K.; Kufrin, D.; Helms, W.S.; Subramaniam, S.; Zhao, H.; Patel, A. FDA Approval Summary: Trabectedin for Unresectable or Metastatic Liposarcoma or Leiomyosarcoma Following an Anthracycline-Containing RegimenFDA Approval Summary: Trabectedin. Clin. Cancer Res. 2017, 23, 7448–7453. [Google Scholar] [CrossRef] [Green Version]
  99. Zijoo, R.; von Mehren, M. Efficacy of trabectedin for the treatment of liposarcoma. Expert Opin. Pharmacother. 2016, 17, 1953–1962. [Google Scholar] [CrossRef]
  100. Matsuda, S.; Tanaka, K.; Kawano, M.; Iwasaki, T.; Itonaga, I.; Tsumura, H. Long-term disease control by trabectedin in a patient with dedifferentiated liposarcoma: A case report. Medicine 2020, 99, e18689. [Google Scholar] [CrossRef]
  101. Liguori, M.; Buracchi, C.; Pasqualini, F.; Bergomas, F.; Pesce, S.; Sironi, M.; Grizzi, F.; Mantovani, A.; Belgiovine, C.; Allavena, P. Functional TRAIL receptors in monocytes and tumor-associated macrophages: A possible targeting pathway in the tumor microenvironment. Oncotarget 2016, 7, 41662. [Google Scholar] [CrossRef] [Green Version]
  102. Germano, G.; Frapolli, R.; Belgiovine, C.; Anselmo, A.; Pesce, S.; Liguori, M.; Erba, E.; Uboldi, S.; Zucchetti, M.; Pasqualini, F. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013, 23, 249–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Elez, M.E.; Tabernero, J.; Geary, D.; Macarulla, T.; Kang, S.P.; Kahatt, C.; Pita, A.S.-M.; Teruel, C.F.; Siguero, M.; Cullell-Young, M. First-In-Human Phase I Study of Lurbinectedin (PM01183) in Patients with Advanced Solid TumorsFirst-In-Human Phase I Study of Lurbinectedin (PM01183). Clin. Cancer Res. 2014, 20, 2205–2214. [Google Scholar] [CrossRef] [Green Version]
  104. Poveda, A.; Del Campo, J.; Ray-Coquard, I.; Alexandre, J.; Provansal, M.; Alía, E.G.; Casado, A.; Gonzalez-Martin, A.; Fernández, C.; Rodriguez, I. Phase II randomized study of PM01183 versus topotecan in patients with platinum-resistant/refractory advanced ovarian cancer. Ann. Oncol. 2017, 28, 1280–1287. [Google Scholar] [CrossRef]
  105. Belgiovine, C.; Bello, E.; Liguori, M.; Craparotta, I.; Mannarino, L.; Paracchini, L.; Beltrame, L.; Marchini, S.; Galmarini, C.M.; Mantovani, A. Lurbinectedin reduces tumour-associated macrophages and the inflammatory tumour microenvironment in preclinical models. Br. J. Cancer 2017, 117, 628–638. [Google Scholar] [CrossRef]
  106. Razak, A.R.; Cleary, J.M.; Moreno, V.; Boyer, M.; Aller, E.C.; Edenfield, W.; Tie, J.; Harvey, R.D.; Rutten, A.; Shah, M.A. Safety and efficacy of AMG 820, an anti-colony-stimulating factor 1 receptor antibody, in combination with pembrolizumab in adults with advanced solid tumors. J. Immunother. Cancer 2020, 8, e001006. [Google Scholar] [CrossRef]
  107. Papadopoulos, K.P.; Gluck, L.; Martin, L.P.; Olszanski, A.J.; Tolcher, A.W.; Ngarmchamnanrith, G.; Rasmussen, E.; Amore, B.M.; Nagorsen, D.; Hill, J.S. First-in-human study of AMG 820, a monoclonal anti-colony-stimulating factor 1 receptor antibody, in patients with advanced solid tumors. Clin. Cancer Res. 2017, 23, 5703–5710. [Google Scholar] [CrossRef] [Green Version]
  108. Cassier, P.A.; Italiano, A.; Gomez-Roca, C.A.; Le Tourneau, C.; Toulmonde, M.; Cannarile, M.A.; Ries, C.; Brillouet, A.; Müller, C.; Jegg, A.-M. CSF1R inhibition with emactuzumab in locally advanced diffuse-type tenosynovial giant cell tumours of the soft tissue: A dose-escalation and dose-expansion phase 1 study. Lancet Oncol. 2015, 16, 949–956. [Google Scholar] [CrossRef] [PubMed]
  109. Deng, C.; Zhang, Q.; Jia, M.; Zhao, J.; Sun, X.; Gong, T.; Zhang, Z. Tumors and their microenvironment dual-targeting chemotherapy with local immune adjuvant therapy for effective antitumor immunity against breast cancer. Adv. Sci. 2019, 6, 1801868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Unger, W.W.; Van Beelen, A.J.; Bruijns, S.C.; Joshi, M.; Fehres, C.M.; Van Bloois, L.; Verstege, M.I.; Ambrosini, M.; Kalay, H.; Nazmi, K. Glycan-modified liposomes boost CD4+ and CD8+ T-cell responses by targeting DC-SIGN on dendritic cells. J. Control. Release 2012, 160, 88–95. [Google Scholar] [CrossRef]
  111. Yang, L.; Zhang, Y. Tumor-associated macrophages: From basic research to clinical application. J. Hematol. Oncol. 2017, 10, 58. [Google Scholar] [CrossRef] [Green Version]
  112. Brana, I.; Calles, A.; LoRusso, P.M.; Yee, L.K.; Puchalski, T.A.; Seetharam, S.; Zhong, B.; de Boer, C.J.; Tabernero, J.; Calvo, E. Carlumab, an anti-CC chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: An open-label, multicenter phase 1b study. Target. Oncol. 2015, 10, 111–123. [Google Scholar] [CrossRef]
  113. Masuda, T.; Noda, M.; Kogawa, T.; Kitagawa, D.; Hayashi, N.; Jomori, T.; Nakanishi, Y.; Nakayama, K.I.; Ohno, S.; Mimori, K. Phase I dose-escalation trial to repurpose propagermanium, an oral CCL2 inhibitor, in patients with breast cancer. Cancer Sci. 2020, 111, 924–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Nywening, T.M.; Wang-Gillam, A.; Sanford, D.E.; Belt, B.A.; Panni, R.Z.; Cusworth, B.M.; Toriola, A.T.; Nieman, R.K.; Worley, L.A.; Yano, M. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: A single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016, 17, 651–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Cherney, R.J.; Anjanappa, P.; Selvakumar, K.; Batt, D.G.; Brown, G.D.; Rose, A.V.; Vuppugalla, R.; Chen, J.; Pang, J.; Xu, S. BMS-813160: A Potent CCR2 and CCR5 Dual Antagonist Selected as a Clinical Candidate. ACS Med. Chem. Lett. 2021, 12, 1753–1758. [Google Scholar] [CrossRef]
  116. Cristofanilli, M.; Dolezal, M.; Lalezari, J.; Rui, H.; Patterson, B.; Tang, C.-M.; Adams, D.; Zhang, Q.; Kazempour, K.; Pourhassan, N. Abstract CT233: Phase Ib/II study of leronlimab (PRO 140) combined with carboplatin in CCR5+ mTNBC patients. Cancer Res. 2020, 80, CT233. [Google Scholar] [CrossRef]
  117. Qi, B.; Fang, Q.; Liu, S.; Hou, W.; Li, J.; Huang, Y.; Shi, J. Advances of CCR5 antagonists: From small molecules to macromolecules. Eur. J. Med. Chem. 2020, 208, 112819. [Google Scholar] [CrossRef]
  118. Haag, G.M.; Halama, N.; Springfeld, C.; Grün, B.; Apostolidis, L.; Zschaebitz, S.; Dietrich, M.; Berger, A.-K.; Weber, T.F.; Zoernig, I. Combined PD-1 inhibition (Pembrolizumab) and CCR5 inhibition (Maraviroc) for the treatment of refractory microsatellite stable (MSS) metastatic colorectal cancer (mCRC): First results of the PICCASSO phase I trial. J. Clin. Oncol. 2020, 38, 3010. [Google Scholar] [CrossRef]
  119. Jiao, X.; Nawab, O.; Patel, T.; Kossenkov, A.V.; Halama, N.; Jaeger, D.; Pestell, R.G. Recent Advances Targeting CCR5 for Cancer and Its Role in Immuno-OncologyTargeting CCR5 for Cancer. Cancer Res. 2019, 79, 4801–4807. [Google Scholar] [CrossRef] [Green Version]
  120. Cambien, B.; Richard-Fiardo, P.; Karimdjee, B.F.; Martini, V.; Ferrua, B.; Pitard, B.; Schmid-Antomarchi, H.; Schmid-Alliana, A. CCL5 neutralization restricts cancer growth and potentiates the targeting of PDGFRβ in colorectal carcinoma. PLoS ONE 2011, 6, e28842. [Google Scholar] [CrossRef]
  121. Zhang, Y.; Arnatt, C.K.; Zhang, F.; Wang, J.; Haney, K.M.; Fang, X. The potential role of anibamine, a natural product CCR5 antagonist, and its analogues as leads toward development of anti-ovarian cancer agents. Bioorg. Med. Chem. Lett. 2012, 22, 5093–5097. [Google Scholar] [CrossRef]
  122. Donatella, A.; Borghese, C.; Casagrande, N. The CCL5/CCR5 axis in cancer progression. Cancers 2020, 12, 1765. [Google Scholar]
  123. Jung, K.; Heishi, T.; Khan, O.F.; Kowalski, P.S.; Incio, J.; Rahbari, N.N.; Chung, E.; Clark, J.W.; Willett, C.G.; Luster, A.D. Ly6C lo monocytes drive immunosuppression and confer resistance to anti-VEGFR2 cancer therapy. J. Clin. Investig. 2017, 127, 3039–3051. [Google Scholar] [CrossRef] [PubMed]
  124. Lamb, R.Y.N. First approval. Drugs 2020, 80, 841–848. [Google Scholar] [CrossRef]
  125. Sun, Y.; Yang, L.; Hao, X.; Liu, Y.; Zhang, J.; Ning, Z.; Shi, Y. Phase I dose-escalation study of chiauranib, a novel angiogenic, mitotic, and chronic inflammation inhibitor, in patients with advanced solid tumors. J. Hematol. Oncol. 2019, 12, 9. [Google Scholar] [CrossRef] [Green Version]
  126. Ries, C.H.; Cannarile, M.A.; Hoves, S.; Benz, J.; Wartha, K.; Runza, V.; Rey-Giraud, F.; Pradel, L.P.; Feuerhake, F.; Klaman, I. Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy. Cancer Cell 2014, 25, 846–859. [Google Scholar] [CrossRef] [PubMed]
  127. Weiss, S.A.; Djureinovic, D.; Jessel, S.; Krykbaeva, I.; Zhang, L.; Jilaveanu, L.; Ralabate, A.; Johnson, B.; Levit, N.S.; Anderson, G. A Phase I Study of APX005M and Cabiralizumab with or without Nivolumab in Patients with Melanoma, Kidney Cancer, or Non–Small Cell Lung Cancer Resistant to Anti-PD-1/PD-L1Phase I Study of APX005M, Cabiralizumab, and Nivolumab. Clin. Cancer Res. 2021, 27, 4757–4767. [Google Scholar] [CrossRef]
  128. Halbrook, C.J.; Pontious, C.; Kovalenko, I.; Lapienyte, L.; Dreyer, S.; Lee, H.-J.; Thurston, G.; Zhang, Y.; Lazarus, J.; Sajjakulnukit, P. Macrophage-released pyrimidines inhibit gemcitabine therapy in pancreatic cancer. Cell Metab. 2019, 29, 1390–1399. [Google Scholar] [CrossRef]
  129. Shen, F.; Zhang, Y.; Jernigan, D.L.; Feng, X.; Yan, J.; Garcia, F.U.; Meucci, O.; Salvino, J.M.; Fatatis, A. Novel Small-Molecule CX3CR1 Antagonist Impairs Metastatic Seeding and Colonization of Breast Cancer CellsTargeting CX3CR1 Reduces Breast Cancer Metastasis. Mol. Cancer Res. 2016, 14, 518–527. [Google Scholar] [CrossRef] [Green Version]
  130. Vonderheide, R.H.; Flaherty, K.T.; Khalil, M.; Stumacher, M.S.; Bajor, D.L.; Hutnick, N.A.; Sullivan, P.; Mahany, J.J.; Gallagher, M.; Kramer, A. Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J. Clin. Oncol. 2007, 25, 876–883. [Google Scholar] [CrossRef]
  131. Beatty, G.L.; Torigian, D.A.; Chiorean, E.G.; Saboury, B.; Brothers, A.; Alavi, A.; Troxel, A.B.; Sun, W.; Teitelbaum, U.R.; Vonderheide, R.H. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin. Cancer Res. 2013, 19, 6286–6295. [Google Scholar] [CrossRef] [Green Version]
  132. Buhtoiarov, I.N.; Lum, H.; Berke, G.; Paulnock, D.M.; Sondel, P.M.; Rakhmilevich, A.L. CD40 ligation activates murine macrophages via an IFN-γ-dependent mechanism resulting in tumor cell destruction in vitro. J. Immunol. 2005, 174, 6013–6022. [Google Scholar] [CrossRef]
  133. Rakhmilevich, A.L.; Alderson, K.L.; Sondel, P.M. T-cell-independent antitumor effects of CD40 ligation. Int. Rev. Immunol. 2012, 31, 267–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. O’Hara, M.H.; O’Reilly, E.M.; Varadhachary, G.; Wolff, R.A.; Wainberg, Z.A.; Ko, A.H.; Fisher, G.; Rahma, O.; Lyman, J.P.; Cabanski, C.R. CD40 agonistic monoclonal antibody APX005M (sotigalimab) and chemotherapy, with or without nivolumab, for the treatment of metastatic pancreatic adenocarcinoma: An open-label, multicentre, phase 1b study. Lancet Oncol. 2021, 22, 118–131. [Google Scholar] [CrossRef] [PubMed]
  135. Sanborn, R.E.; Gabrail, N.Y.; Bhardwaj, N.; Gordon, M.S.; O’Hara, M.; Khalil, D.; Hawthorne, T.; Gedrich, R.; Vitale, L.; Rogalski, M. Abstract LB-194: First-in-human Phase I study of the CD40 agonist mAb CDX-1140 and in combination with CDX-301 (rhFLT3L) in patients with advanced cancers: Interim results. Cancer Res. 2019, 79, LB-194. [Google Scholar] [CrossRef]
  136. Bajor, D.L.; Gutierrez, M.; Vaccaro, G.M.; Masood, A.; Brown-Glaberman, U.; Grilley-Olson, J.E.; Kindler, H.L.; Zalupski, M.; Heath, E.I.; Piha-Paul, S.A. Preliminary results of a phase 1 study of sea-CD40, gemcitabine, nab-paclitaxel, and pembrolizumab in patients with metastatic pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2022, 40, 559. [Google Scholar] [CrossRef]
  137. Ansell, S.M.; Maris, M.B.; Lesokhin, A.M.; Chen, R.W.; Flinn, I.W.; Sawas, A.; Minden, M.D.; Villa, D.; Percival, M.-E.M.; Advani, A.S. Phase I study of the CD47 blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin. Cancer Res. 2021, 27, 2190–2199. [Google Scholar] [CrossRef] [PubMed]
  138. Dheilly, E.; Majocchi, S.; Moine, V.; Didelot, G.; Broyer, L.; Calloud, S.; Malinge, P.; Chatel, L.; Ferlin, W.G.; Kosco-Vilbois, M.H. Tumor-directed blockade of CD47 with bispecific antibodies induces adaptive antitumor immunity. Antibodies 2018, 7, 3. [Google Scholar] [CrossRef] [Green Version]
  139. Sikic, B.I.; Lakhani, N.; Patnaik, A.; Shah, S.A.; Chandana, S.R.; Rasco, D.; Colevas, A.D.; O’Rourke, T.; Narayanan, S.; Papadopoulos, K. First-in-human, first-in-class phase I trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J. Clin. Oncol. 2019, 37, 946. [Google Scholar] [CrossRef]
  140. Fisher, G.A.; Lakhani, N.J.; Eng, C.; Hecht, J.R.; Bendell, J.C.; Philip, P.A.; O’Dwyer, P.J.; Johnson, B.; Kardosh, A.; Ippolito, T.M. A phase Ib/II study of the anti-CD47 antibody magrolimab with cetuximab in solid tumor and colorectal cancer patients. J. Clin. Oncol. 2020, 38, 114. [Google Scholar] [CrossRef]
  141. Hourani, T.; Holden, J.A.; Li, W.; Lenzo, J.C.; Hadjigol, S.; O’Brien-Simpson, N.M. Tumor associated macrophages: Origin, recruitment, phenotypic diversity, and targeting. Front. Oncol. 2021, 11, 788365. [Google Scholar] [CrossRef]
  142. Johnson, P.; Challis, R.; Chowdhury, F.; Gao, Y.; Harvey, M.; Geldart, T.; Kerr, P.; Chan, C.; Smith, A.; Steven, N. Clinical and Biological Effects of an Agonist Anti-CD40 Antibody: A Cancer Research UK Phase I StudyAnti-CD40 Phase I Study. Clin. Cancer Res. 2015, 21, 1321–1328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Yan, D.; Kowal, J.; Akkari, L.; Schuhmacher, A.; Huse, J.; West, B.; Joyce, J. Inhibition of colony stimulating factor-1 receptor abrogates microenvironment-mediated therapeutic resistance in gliomas. Oncogene 2017, 36, 6049–6058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Yu, G.T.; Rao, L.; Wu, H.; Yang, L.L.; Bu, L.L.; Deng, W.W.; Wu, L.; Nan, X.; Zhang, W.F.; Zhao, X.Z. Myeloid-derived suppressor cell membrane-coated magnetic nanoparticles for cancer theranostics by inducing macrophage polarization and synergizing immunogenic cell death. Adv. Funct. Mater. 2018, 28, 1801389. [Google Scholar] [CrossRef]
  146. Sabado, R.L.; Pavlick, A.; Gnjatic, S.; Cruz, C.; Vengco, I.; Farah, H.; Darvishian, F.; Chiriboga, L.; Holman, R.M.; Escalon, J. Phase I/II study of Resiquimod as an immunologic adjuvant for NY-ESO-1 protein vaccination in patients with melanoma. Proc. J. ImmunoTher. Cancer 2013, 1, P272. [Google Scholar] [CrossRef] [Green Version]
  147. Li, H.; Somiya, M.; Kuroda, S.i. Enhancing antibody-dependent cellular phagocytosis by Re-education of tumor-associated macrophages with resiquimod-encapsulated liposomes. Biomaterials 2021, 268, 120601. [Google Scholar] [CrossRef]
  148. Sharma, M.; Carvajal, R.D.; Hanna, G.J.; Li, B.T.; Moore, K.N.; Pegram, M.D.; Rasco, D.W.; Spira, A.I.; Alonso, M.; Fang, L. Preliminary results from a phase 1/2 study of BDC-1001, a novel HER2 targeting TLR7/8 immune-stimulating antibody conjugate (ISAC), in patients (pts) with advanced HER2-expressing solid tumors. J. Clin. Oncol. 2021, 39, 2549. [Google Scholar] [CrossRef]
  149. Wang, S.; Astsaturov, I.A.; Bingham, C.A.; McCarthy, K.M.; von Mehren, M.; Xu, W.; Alpaugh, R.K.; Tang, Y.; Littlefield, B.A.; Hawkins, L.D. Effective antibody therapy induces host-protective antitumor immunity that is augmented by TLR4 agonist treatment. Cancer Immunol. Immunother. 2012, 61, 49–61. [Google Scholar] [CrossRef] [Green Version]
  150. Fennell, D.A.; Casbard, A.C.; Porter, C.; Rudd, R.; Lester, J.F.; Nicolson, M.; Morgan, B.; Steele, J.P.; Darlison, L.; Gardner, G.M. A randomized phase II trial of oral vinorelbine as second-line therapy for patients with malignant pleural mesothelioma. J. Clin. Oncol. 2021, 39, 8507. [Google Scholar] [CrossRef]
  151. Ribas, A.; Medina, T.; Kirkwood, J.M.; Zakharia, Y.; Gonzalez, R.; Davar, D.; Chmielowski, B.; Campbell, K.M.; Bao, R.; Kelley, H. Overcoming PD-1 blockade resistance with CpG-A toll-like receptor 9 agonist vidutolimod in patients with metastatic melanoma. Cancer Discov. 2021, 11, 2998–3007. [Google Scholar] [CrossRef]
  152. Sullivan, R.J.; Hong, D.S.; Tolcher, A.W.; Patnaik, A.; Shapiro, G.; Chmielowski, B.; Ribas, A.; Brail, L.H.; Roberts, J.; Lee, L. Initial results from first-in-human study of IPI-549, a tumor macrophage-targeting agent, combined with nivolumab in advanced solid tumors. J. Clin. Oncol. 2018, 36, 3013. [Google Scholar] [CrossRef]
  153. Kaneda, M.M.; Messer, K.S.; Ralainirina, N.; Li, H.; Leem, C.J.; Gorjestani, S.; Woo, G.; Nguyen, A.V.; Figueiredo, C.C.; Foubert, P. PI3Kγ is a molecular switch that controls immune suppression. Nature 2016, 539, 437–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. La Fleur, L.; Botling, J.; He, F.; Pelicano, C.; Zhou, C.; He, C.; Palano, G.; Mezheyeuski, A.; Micke, P.; Ravetch, J.V. Targeting MARCO and IL37R on immunosuppressive macrophages in lung cancer blocks regulatory T cells and supports cytotoxic lymphocyte function. Cancer Res. 2021, 81, 956–967. [Google Scholar] [CrossRef] [PubMed]
  155. Georgoudaki, A.-M.; Prokopec, K.E.; Boura, V.F.; Hellqvist, E.; Sohn, S.; Östling, J.; Dahan, R.; Harris, R.A.; Rantalainen, M.; Klevebring, D. Reprogramming tumor-associated macrophages by antibody targeting inhibits cancer progression and metastasis. Cell Rep. 2016, 15, 2000–2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Zanganeh, S.; Hutter, G.; Spitler, R.; Lenkov, O.; Mahmoudi, M.; Shaw, A.; Pajarinen, J.S.; Nejadnik, H.; Goodman, S.; Moseley, M.; et al. Iron Oxide Nanoparticles Inhibit Tumour Growth by Inducing Pro-Inflammatory Macrophage Polarization in Tumour Tissues. Nat. Nanotechnol. 2016, 11, 986–994. [Google Scholar] [CrossRef] [PubMed]
  157. Fu, X.; Yu, J.; Yuan, A.; Liu, L.; Zhao, H.; Huang, Y.; Shen, S.; Lv, F.; Wang, S. Polymer nanoparticles regulate macrophage repolarization for antitumor treatment. Chem. Commun. 2021, 57, 6919–6922. [Google Scholar] [CrossRef] [PubMed]
  158. Huang, Z.; Yang, Y.; Jiang, Y.; Shao, J.; Sun, X.; Chen, J.; Dong, L.; Zhang, J. Anti-tumor immune responses of tumor-associated macrophages via toll-like receptor 4 triggered by cationic polymers. Biomaterials 2013, 34, 746–755. [Google Scholar] [CrossRef]
  159. Rodell, C.B.; Arlauckas, S.P.; Cuccarese, M.F.; Garris, C.S.; Li, R.; Ahmed, M.S.; Kohler, R.H.; Pittet, M.J.; Weissleder, R. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat. Biomed. Eng. 2018, 2, 578–588. [Google Scholar] [CrossRef]
  160. Finocchiaro, G.; Gentner, B.; Farina, F.; Capotondo, A.; Eoli, M.; Anghileri, E.; Carabba, M.G.; Cuccarini, V.; Di Meco, F.; Legnani, F. A phase I-IIa study of genetically modified Tie-2 expressing monocytes in patients with glioblastoma multiforme (TEM-GBM Study). J. Clin. Oncol. 2021, 39, 2532. [Google Scholar] [CrossRef]
  161. Tang, H.; Liang, Y.; Anders, R.A.; Taube, J.M.; Qiu, X.; Mulgaonkar, A.; Liu, X.; Harrington, S.M.; Guo, J.; Xin, Y. PD-L1 on host cells is essential for PD-L1 blockade–mediated tumor regression. J. Clin. Investig. 2018, 128, 580–588. [Google Scholar] [CrossRef] [Green Version]
  162. Rogers, B.M.; Smith, L.; Dezso, Z.; Shi, X.; DiGiammarino, E.; Nguyen, D.; Sethuraman, S.; Zheng, P.; Choi, D.; Zhang, D. VISTA is an activating receptor in human monocytes. J. Exp. Med. 2021, 218, e20201601. [Google Scholar] [CrossRef] [PubMed]
  163. Chow, A.; Schad, S.; Green, M.D.; Hellmann, M.D.; Allaj, V.; Ceglia, N.; Zago, G.; Shah, N.S.; Sharma, S.K.; Mattar, M. Tim-4+ cavity-resident macrophages impair anti-tumor CD8+ T cell immunity. Cancer Cell 2021, 39, 973–988.e9. [Google Scholar] [CrossRef] [PubMed]
  164. Cheng, L.; Wang, Y.; Huang, L. Exosomes from M1-polarized macrophages potentiate the cancer vaccine by creating a pro-inflammatory microenvironment in the lymph node. Mol. Ther. 2017, 25, 1665–1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Dey, S.; Sutanto-Ward, E.; Kopp, K.L.; DuHadaway, J.; Mondal, A.; Ghaban, D.; Lecoq, I.; Zocca, M.-B.; Merlo, L.M.; Mandik-Nayak, L. Peptide vaccination directed against IDO1-expressing immune cells elicits CD8+ and CD4+ T-cell-mediated antitumor immunity and enhanced anti-PD1 responses. J. Immunother. Cancer 2020, 8, e000605. [Google Scholar] [CrossRef]
  166. Cheever, M.A.; Higano, C.S. PROVENGE (Sipuleucel-T) in Prostate Cancer: The First FDA-Approved Therapeutic Cancer VaccineTherapeutic Prostate Cancer Vaccine. Clin. Cancer Res. 2011, 17, 3520–3526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Fu, J.; Kanne, D.B.; Leong, M.; Glickman, L.H.; McWhirter, S.M.; Lemmens, E.; Mechette, K.; Leong, J.J.; Lauer, P.; Liu, W. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci. Transl. Med. 2015, 7, ra252–ra283. [Google Scholar] [CrossRef] [Green Version]
  168. Cieslewicz, M.; Tang, J.; Yu, J.L.; Cao, H.; Zavaljevski, M.; Motoyama, K.; Lieber, A.; Raines, E.W.; Pun, S.H. Targeted delivery of proapoptotic peptides to tumor-associated macrophages improves survival. Proc. Natl. Acad. Sci. USA 2013, 110, 15919–15924. [Google Scholar] [CrossRef]
  169. Scodeller, P.; Simón-Gracia, L.; Kopanchuk, S.; Tobi, A.; Kilk, K.; Säälik, P.; Kurm, K.; Squadrito, M.L.; Kotamraju, V.R.; Rinken, A. Precision targeting of tumor macrophages with a CD206 binding peptide. Sci. Rep. 2017, 7, 14655. [Google Scholar] [CrossRef] [Green Version]
  170. Lee, C.; Bae, S.-J.S.; Joo, H.; Bae, H. Melittin suppresses tumor progression by regulating tumor-associated macrophages in a Lewis lung carcinoma mouse model. Oncotarget 2017, 8, 54951. [Google Scholar] [CrossRef] [Green Version]
  171. Jaynes, J.M.; Sable, R.; Ronzetti, M.; Bautista, W.; Knotts, Z.; Abisoye-Ogunniyan, A.; Li, D.; Calvo, R.; Dashnyam, M.; Singh, A. Mannose receptor (CD206) activation in tumor-associated macrophages enhances adaptive and innate antitumor immune responses. Sci. Transl. Med. 2020, 12, eaax6337. [Google Scholar] [CrossRef]
  172. Vadevoo, S.M.P.; Kim, J.-E.; Gunassekaran, G.R.; Jung, H.-K.; Chi, L.; Kim, D.E.; Lee, S.-H.; Im, S.-H.; Lee, B. IL4 Receptor–Targeted Proapoptotic Peptide Blocks Tumor Growth and Metastasis by Enhancing Antitumor ImmunityAnticancer Activity of IL4R-Targeted Proapoptotic Peptide. Mol. Cancer Ther. 2017, 16, 2803–2816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Zhang, L.; Qi, Y.; Min, H.; Ni, C.; Wang, F.; Wang, B.; Qin, H.; Zhang, Y.; Liu, G.; Qin, Y. Cooperatively responsive peptide nanotherapeutic that regulates angiopoietin receptor Tie2 activity in tumor microenvironment to prevent breast tumor relapse after chemotherapy. ACS Nano 2019, 13, 5091–5102. [Google Scholar] [CrossRef]
  174. Wang, H.; Sun, Y.; Zhou, X.; Chen, C.; Jiao, L.; Li, W.; Gou, S.; Li, Y.; Du, J.; Chen, G. CD47/SIRPα blocking peptide identification and synergistic effect with irradiation for cancer immunotherapy. J. Immunother. Cancer 2020, 8, e000905. [Google Scholar] [CrossRef]
  175. Tang, T.; Wei, Y.; Kang, J.; She, Z.-G.; Kim, D.; Sailor, M.J.; Ruoslahti, E.; Pang, H.-B. Tumor-specific macrophage targeting through recognition of retinoid X receptor beta. J. Control. Release 2019, 301, 42–53. [Google Scholar] [CrossRef] [PubMed]
  176. Galluzzi, L.; Humeau, J.; Buqué, A.; Zitvogel, L.; Kroemer, G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2020, 17, 725–741. [Google Scholar] [CrossRef]
  177. Heath, O.; Berlato, C.; Maniati, E.; Lakhani, A.; Pegrum, C.; Kotantaki, P.; Elorbany, S.; Böhm, S.; Barry, S.T.; Annibaldi, A. Chemotherapy induces tumor-associated macrophages that aid adaptive immune responses in ovarian cancer. Cancer Immunol. Res. 2021, 9, 665–681. [Google Scholar] [CrossRef] [PubMed]
  178. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef]
  179. Rogers, T.L.; Holen, I. Tumour macrophages as potential targets of bisphosphonates. J. Transl. Med. 2011, 9, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Salvagno, C.; Ciampricotti, M.; Tuit, S.; Hau, C.-S.; van Weverwijk, A.; Coffelt, S.B.; Kersten, K.; Vrijland, K.; Kos, K.; Ulas, T. Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response. Nat. Cell Biol. 2019, 21, 511–521. [Google Scholar] [CrossRef] [PubMed]
  181. Weng, W.-K.; Levy, R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 2003, 21, 3940–3947. [Google Scholar] [CrossRef]
  182. Bibeau, F.; Lopez-Crapez, E.; Di Fiore, F.; Thezenas, S.; Ychou, M.; Blanchard, F.; Lamy, A.; Penault-Llorca, F.; Frébourg, T.; Michel, P. Impact of FcγRIIa-FcγRIIIa polymorphisms and KRAS mutations on the clinical outcome of patients with metastatic colorectal cancer treated with cetuximab plus irinotecan. J. Clin. Oncol. 2009, 27, 1122–1129. [Google Scholar] [CrossRef] [PubMed]
  183. DiLillo, D.J.; Ravetch, J.V. Fc-receptor interactions regulate both cytotoxic and immunomodulatory therapeutic antibody effector functions. Cancer Immunol. Res. 2015, 3, 704–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Musolino, A.; Naldi, N.; Bortesi, B.; Pezzuolo, D.; Capelletti, M.; Missale, G.; Laccabue, D.; Zerbini, A.; Camisa, R.; Bisagni, G. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu–positive metastatic breast cancer. J. Clin. Oncol. 2008, 26, 1789–1796. [Google Scholar] [CrossRef] [PubMed]
  185. Lapenna, A.; De Palma, M.; Lewis, C.E. Perivascular macrophages in health and disease. Nat. Rev. Immunol. 2018, 18, 689–702. [Google Scholar] [CrossRef]
  186. Kloepper, J.; Riedemann, L.; Amoozgar, Z.; Seano, G.; Susek, K.; Yu, V.; Dalvie, N.; Amelung, R.L.; Datta, M.; Song, J.W. Ang-2/VEGF bispecific antibody reprograms macrophages and resident microglia to anti-tumor phenotype and prolongs glioblastoma survival. Proc. Natl. Acad. Sci. USA 2016, 113, 4476–4481. [Google Scholar] [CrossRef]
  187. Herroon, M.; Rajagurubandara, E.; Rudy, D.; Chalasani, A.; Hardaway, A.; Podgorski, I. Macrophage cathepsin K promotes prostate tumor progression in bone. Oncogene 2013, 32, 1580–1593. [Google Scholar] [CrossRef] [Green Version]
  188. Vasiljeva, O.; Papazoglou, A.; Krüger, A.; Brodoefel, H.; Korovin, M.; Deussing, J.; Augustin, N.; Nielsen, B.S.; Almholt, K.; Bogyo, M. Tumor cell–derived and macrophage-derived cathepsin B promotes progression and lung metastasis of mammary cancer. Cancer Res. 2006, 66, 5242–5250. [Google Scholar] [CrossRef] [Green Version]
  189. Barth, N.D.; Van Dalen, F.J.; Karmakar, U.; Bertolini, M.; Mendive-Tapia, L.; Kitamura, T.; Verdoes, M.; Vendrell, M. Enzyme-Activatable Chemokine Conjugates for In Vivo Targeting of Tumor-Associated Macrophages. Angew. Chem. Int. Ed. 2022, 61, e202207508. [Google Scholar] [CrossRef]
  190. Xu, R.-H.; Kalechman, Y.; Albeck, M.; Sredni, B. The cytoprotective effect of the immunomodulator AS101 against hydrochloride induced gastric lesions. Res. Commun. Mol. Pathol. Pharmacol. 1995, 87, 4–20. [Google Scholar]
  191. Dalton, H.J.; Pradeep, S.; McGuire, M.; Hailemichael, Y.; Ma, S.; Lyons, Y.; Armaiz-Pena, G.N.; Previs, R.A.; Hansen, J.M.; Rupaimoole, R. Macrophages Facilitate Resistance to Anti-VEGF Therapy by Altered VEGFR ExpressionMacrophages Adapt to Anti-VEGF Therapy. Clin. Cancer Res. 2017, 23, 7034–7046. [Google Scholar] [CrossRef] [Green Version]
  192. Mitamura, T.; Pradeep, S.; McGuire, M.; Wu, S.; Ma, S.; Hatakeyama, H.; Lyons, Y.A.; Hisamatsu, T.; Noh, K.; Villar-Prados, A. Induction of anti-VEGF therapy resistance by upregulated expression of microseminoprotein (MSMP). Oncogene 2018, 37, 722–731. [Google Scholar] [CrossRef]
  193. Wu, Y.; Jennings, N.B.; Sun, Y.; Dasari, S.K.; Bayraktar, E.; Corvigno, S.; Stur, E.; Glassman, D.; Mangala, L.S.; Lankenau Ahumada, A. Targeting CCR2+ macrophages with BET inhibitor overcomes adaptive resistance to anti-VEGF therapy in ovarian cancer. J. Cancer Res. Clin. Oncol. 2022, 148, 803–821. [Google Scholar] [CrossRef] [PubMed]
  194. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef] [PubMed]
  195. Kuang, D.-M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.-P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef]
  196. Bloch, O.; Crane, C.A.; Kaur, R.; Safaee, M.; Rutkowski, M.J.; Parsa, A.T. Gliomas Promote Immunosuppression through Induction of B7-H1 Expression in Tumor-Associated MacrophagesB7-H1 in Glioma TAM. Clin. Cancer Res. 2013, 19, 3165–3175. [Google Scholar] [CrossRef] [Green Version]
  197. Havel, J.J.; Chowell, D.; Chan, T.A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 2019, 19, 133–150. [Google Scholar] [CrossRef]
  198. Strauss, L.; Mahmoud, M.A.; Weaver, J.D.; Tijaro-Ovalle, N.M.; Christofides, A.; Wang, Q.; Pal, R.; Yuan, M.; Asara, J.; Patsoukis, N. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci. Immunol. 2020, 5, eaay1863. [Google Scholar] [CrossRef] [PubMed]
  199. Laba, S.; Mallett, G.; Amarnath, S. The depths of PD-1 function within the tumor microenvironment beyond CD8+ T cells. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2021. [Google Scholar]
  200. Wang, L.; Rubinstein, R.; Lines, J.L.; Wasiuk, A.; Ahonen, C.; Guo, Y.; Lu, L.-F.; Gondek, D.; Wang, Y.; Fava, R.A. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 2011, 208, 577–592. [Google Scholar] [CrossRef]
  201. Johnston, R.J.; Su, L.J.; Pinckney, J.; Critton, D.; Boyer, E.; Krishnakumar, A.; Corbett, M.; Rankin, A.L.; Dibella, R.; Campbell, L. VISTA is an acidic pH-selective ligand for PSGL-1. Nature 2019, 574, 565–570. [Google Scholar] [CrossRef]
  202. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [Green Version]
  203. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.; Karpinets, T.; Prieto, P.; Vicente, D.; Hoffman, K.; Wei, S.C. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
  204. Fridlender, Z.; Buchlis, G.; Kapoor, V.; Cheng, G.; Sun, J.; Singhal, S.; Crisanti, M.; Wang, L.C.; Heitjan, D.; Snyder, L. Correction: CCL2 blockade augments cancer immunotherapy (Cancer Research (2010) 70, (109–118). Cancer Res. 2010, 70, 2569. [Google Scholar] [CrossRef] [Green Version]
  205. Neubert, N.J.; Schmittnaegel, M.; Bordry, N.; Nassiri, S.; Wald, N.; Martignier, C.; Tillé, L.; Homicsko, K.; Damsky, W.; Maby-El Hajjami, H. T cell–induced CSF1 promotes melanoma resistance to PD1 blockade. Sci. Transl. Med. 2018, 10, eaan3311. [Google Scholar] [CrossRef] [Green Version]
  206. Peranzoni, E.; Lemoine, J.; Vimeux, L.; Feuillet, V.; Barrin, S.; Kantari-Mimoun, C.; Bercovici, N.; Guérin, M.; Biton, J.; Ouakrim, H. Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti–PD-1 treatment. Proc. Natl. Acad. Sci. USA 2018, 115, E4041–E4050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Andersen, M.H. The balance players of the adaptive immune system. Cancer Res. 2018, 78, 1379–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Iversen, T.Z.; Engell-Noerregaard, L.; Ellebaek, E.; Andersen, R.; Larsen, S.K.; Bjoern, J.; Zeyher, C.; Gouttefangeas, C.; Thomsen, B.M.; Holm, B. Long-lasting Disease Stabilization in the Absence of Toxicity in Metastatic Lung Cancer Patients Vaccinated with an Epitope Derived from Indoleamine 2, 3 DioxygenasePeptide Vaccination Targeting IDO in Metastatic NSCLC Patients. Clin. Cancer Res. 2014, 20, 221–232. [Google Scholar] [CrossRef] [Green Version]
  209. Kjeldsen, J.W.; Iversen, T.Z.; Engell-Noerregaard, L.; Mellemgaard, A.; Andersen, M.H.; Svane, I.M. Durable clinical responses and long-term follow-up of stage III–IV non-small-cell lung cancer (NSCLC) patients treated with IDO peptide vaccine in a phase I study—A brief research report. Front. Immunol. 2018, 9, 2145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Jørgensen, N.G.; Kaae, J.; Grauslund, J.H.; Met, Ö.; Svane, I.M.; Ehrnrooth, E.; Andersen, M.H.; Zachariae, C.; Skov, L. Efficacy and safety of IO103 a novel anti PD-L1 vaccine in basal cell carcinoma. J. Clin. Oncol. 2020, 38, e22070. [Google Scholar] [CrossRef]
  211. Svane, I.; Kjeldsen, J.; Lorentzen, C.; Martinenaite, E.; Andersen, M. LBA48 Clinical efficacy and immunity of combination therapy with nivolumab and IDO/PD-L1 peptide vaccine in patients with metastatic melanoma: A phase I/II trial. Ann. Oncol. 2020, 31, S1176. [Google Scholar] [CrossRef]
  212. Andersen, M.H. The targeting of tumor-associated macrophages by vaccination. Cell Stress 2019, 3, 139. [Google Scholar] [CrossRef]
  213. Allavena, P.; Palmioli, A.; Avigni, R.; Sironi, M.; La Ferla, B.; Maeda, A. PLGA based nanoparticles for the monocyte-mediated anti-tumor drug delivery system. J. Biomed. Nanotechnol. 2020, 16, 212–223. [Google Scholar] [CrossRef] [PubMed]
  214. De Palma, M.; Mazzieri, R.; Politi, L.S.; Pucci, F.; Zonari, E.; Sitia, G.; Mazzoleni, S.; Moi, D.; Venneri, M.A.; Indraccolo, S. Tumor-targeted interferon-α delivery by Tie2-expressing monocytes inhibits tumor growth and metastasis. Cancer Cell 2008, 14, 299–311. [Google Scholar] [CrossRef] [Green Version]
  215. Shields IV, C.W.; Evans, M.A.; Wang, L.L.-W.; Baugh, N.; Iyer, S.; Wu, D.; Zhao, Z.; Pusuluri, A.; Ukidve, A.; Pan, D.C. Cellular backpacks for macrophage immunotherapy. Sci. Adv. 2020, 6, eaaz6579. [Google Scholar] [CrossRef] [PubMed]
  216. Kaczanowska, S.; Beury, D.W.; Gopalan, V.; Tycko, A.K.; Qin, H.; Clements, M.E.; Drake, J.; Nwanze, C.; Murgai, M.; Rae, Z. Genetically engineered myeloid cells rebalance the core immune suppression program in metastasis. Cell 2021, 184, 2033–2052.e21. [Google Scholar] [CrossRef]
  217. Tanito, K.; Nii, T.; Yokoyama, Y.; Oishi, H.; Shibata, M.; Hijii, S.; Kaneko, R.; Tateishi, C.; Ito, S.; Kishimura, A. Engineered macrophages acting as a trigger to induce inflammation only in tumor tissues. J. Control. Release 2023, in press. [Google Scholar] [CrossRef]
  218. Sunseri, N.; O’Brien, M.; Bhardwaj, N.; Landau, N.R. Human immunodeficiency virus type 1 modified to package Simian immunodeficiency virus Vpx efficiently infects macrophages and dendritic cells. J. Virol. 2011, 85, 6263–6274. [Google Scholar] [CrossRef] [Green Version]
  219. Bobadilla, S.; Sunseri, N.; Landau, N.R. Efficient transduction of myeloid cells by an HIV-1-derived lentiviral vector that packages the Vpx accessory protein. Gene Ther. 2013, 20, 514–520. [Google Scholar] [CrossRef] [Green Version]
  220. Morrissey, M.A.; Williamson, A.P.; Steinbach, A.M.; Roberts, E.W.; Kern, N.; Headley, M.B.; Vale, R.D. Chimeric antigen receptors that trigger phagocytosis. eLife 2018, 7, e36688. [Google Scholar] [CrossRef] [PubMed]
  221. Hallek, M. Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am. J. Hematol. 2019, 94, 1266–1287. [Google Scholar] [CrossRef] [Green Version]
  222. Aalipour, A.; Chuang, H.-Y.; Murty, S.; D’Souza, A.L.; Park, S.-m.; Gulati, G.S.; Patel, C.B.; Beinat, C.; Simonetta, F.; Martinić, I. Engineered immune cells as highly sensitive cancer diagnostics. Nat. Biotechnol. 2019, 37, 531–539. [Google Scholar] [CrossRef]
  223. Dai, L.; Bai, A.; Smith, C.D.; Rodriguez, P.C.; Yu, F.; Qin, Z. ABC294640, A Novel Sphingosine Kinase 2 Inhibitor, Induces Oncogenic Virus–Infected Cell Autophagic Death and Represses Tumor GrowthABC294640 and Autophagic Death. Mol. Cancer Ther. 2017, 16, 2724–2734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Pang, L.; Pei, Y.; Uzunalli, G.; Hyun, H.; Lyle, L.T.; Yeo, Y. Surface modification of polymeric nanoparticles with M2pep peptide for drug delivery to tumor-associated macrophages. Pharm. Res. 2019, 36, 65. [Google Scholar] [CrossRef] [PubMed]
  225. Ngambenjawong, C.; Gustafson, H.H.; Pineda, J.M.; Kacherovsky, N.A.; Cieslewicz, M.; Pun, S.H. Serum stability and affinity optimization of an M2 macrophage-targeting peptide (M2pep). Theranostics 2016, 6, 1403. [Google Scholar] [CrossRef]
  226. Ngambenjawong, C.; Sylvestre, M.; Gustafson, H.H.; Pineda, J.M.B.; Pun, S.H. Reversibly switchable, pH-dependent peptide ligand binding via 3, 5-diiodotyrosine substitutions. ACS Chem. Biol. 2018, 13, 995–1002. [Google Scholar] [CrossRef] [PubMed]
  227. He, Y.; de Araújo Júnior, R.F.; Cruz, L.J.; Eich, C. Functionalized nanoparticles targeting tumor-associated macrophages as cancer therapy. Pharmaceutics 2021, 13, 1670. [Google Scholar] [CrossRef] [PubMed]
  228. Ovais, M.; Guo, M.; Chen, C. Tailoring nanomaterials for targeting tumor-associated macrophages. Adv. Mater. 2019, 31, 1808303. [Google Scholar] [CrossRef] [PubMed]
  229. Lim, S.Y.; Yuzhalin, A.E.; Gordon-Weeks, A.N.; Muschel, R.J. Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget 2016, 7, 28697. [Google Scholar] [CrossRef] [Green Version]
  230. Xiao, H.; Guo, Y.; Li, B.; Li, X.; Wang, Y.; Han, S.; Cheng, D.; Shuai, X. M2-like tumor-associated macrophage-targeted codelivery of STAT6 inhibitor and IKKβ siRNA induces M2-to-M1 repolarization for cancer immunotherapy with low immune side effects. ACS Cent. Sci. 2020, 6, 1208–1222. [Google Scholar] [CrossRef]
  231. Hu, G.; Guo, M.; Xu, J.; Wu, F.; Fan, J.; Huang, Q.; Yang, G.; Lv, Z.; Wang, X.; Jin, Y. Nanoparticles targeting macrophages as potential clinical therapeutic agents against cancer and inflammation. Front. Immunol. 2019, 10, 1998. [Google Scholar] [CrossRef]
  232. Pham, L.V.; Pogue, E.; Ford, R.J. The role of macrophage/B-cell interactions in the pathophysiology of B-cell lymphomas. Front. Oncol. 2018, 8, 147. [Google Scholar] [CrossRef]
  233. Banerjee, P.; Zhang, R.; Ivan, C.; Galletti, G.; Clise-Dwyer, K.; Barbaglio, F.; Scarfò, L.; Aracil, M.; Klein, C.; Wierda, W. Trabectedin reveals a strategy of immunomodulation in chronic lymphocytic leukemia. Cancer Immunol. Res. 2019, 7, 2036–2051. [Google Scholar] [CrossRef] [PubMed]
  234. Polk, A.; Lu, Y.; Wang, T.; Seymour, E.; Bailey, N.G.; Singer, J.W.; Boonstra, P.S.; Lim, M.S.; Malek, S.; Wilcox, R.A. Colony-stimulating factor-1 receptor is required for nurse-like cell survival in chronic lymphocytic leukemia. Clin. Cancer Res. 2016, 22, 6118–6128. [Google Scholar] [CrossRef] [Green Version]
  235. Sweeney, D.T.; Ho, H.; Eide, C.A.; Rofelty, A.; Agarwal, A.; Liu, S.Q.; Danilov, A.V.; Lee, P.; Chantry, D.; McWeeney, S.K. Targeting of colony-stimulating factor 1 receptor (CSF1R) in the CLL microenvironment yields antineoplastic activity in primary patient samples. Oncotarget 2018, 9, 24576. [Google Scholar]
  236. Kriston, C.; Hernádfői, M.; Plander, M.; Márk, Á.; Takács, F.; Czeti, Á.; Szalóki, G.; Szabó, O.; Matolcsy, A.; Barna, G. Lenalidomide abrogates the survival effect of bone marrow stromal cells in chronic lymphocytic leukemia. Hematol. Oncol. 2021, 39, 513–520. [Google Scholar] [CrossRef]
  237. Normant, E.; Ribeiro, M.L.; Profitos-Peleja, N.; Blecua, P.; Reyes-Garau, D.; Santos, J.C.; Armengol, M.; Fernández-Serrano, M.; Miskin, H.P.; Roue, G. The Ublituximab-Umbralisib (U2) Drug Regimen Potentiates the Activity of the Novel CD47-CD19 Bispecific Antibody, TG-1801, through the Activation of the G Protein-Coupled Receptor EBI2/GPR183. Blood 2021, 138, 1196. [Google Scholar] [CrossRef]
  238. Valentin, R.; Peluso, M.O.; Lehmberg, T.Z.; Adam, A.; Zhang, L.; Armet, C.M.; Guerriero, J.L.; Lee, B.H.; Palombella, V.J.; Holland, P.M. The fully human anti-CD47 antibody SRF231 has dual-mechanism antitumor activity against chronic lymphocytic leukemia (CLL) Cells and increases the activity of both rituximab and venetoclax. Blood 2018, 132, 4393. [Google Scholar] [CrossRef]
  239. Li, K.; Xu, W.; Lu, K.; Wen, Y.; Xin, T.; Shen, Y.; Lv, X.; Hu, S.; Jin, R.; Wu, X. CSF-1R inhibition disrupts the dialog between leukaemia cells and macrophages and delays leukaemia progression. J. Cell. Mol. Med. 2020, 24, 13115–13128. [Google Scholar] [CrossRef]
  240. Komohara, Y.; Noyori, O.; Saito, Y.; Takeya, H.; Baghdadi, M.; Kitagawa, F.; Hama, N.; Ishikawa, K.; Okuno, Y.; Nosaka, K. Potential anti-lymphoma effect of M-CSFR inhibitor in adult T-cell leukemia/lymphoma. J. Clin. Exp. Hematopathol. JCEH 2018, 58, 152. [Google Scholar] [CrossRef] [Green Version]
  241. Walker, K.L.; Rinella, S.P.; Hess, N.J.; Turicek, D.P.; Kabakov, S.A.; Zhu, F.; Bouchlaka, M.N.; Olson, S.L.; Cho, M.M.; Quamine, A.E.; et al. CXCR4 allows T cell acute lymphoblastic leukemia to escape from JAK1/2 and BCL2 inhibition through CNS infiltration. Leuk. Lymphoma 2021, 62, 1167–1177. [Google Scholar] [CrossRef]
  242. Liu, S.; Luo, X.; Zhang, X.; Xu, L.; Wang, Y.; Yan, C.; Chen, H.; Chen, Y.; Han, W.; Wang, F. Preemptive interferon-α treatment could protect against relapse and improve long-term survival of ALL patients after allo-HSCT. Sci. Rep. 2020, 10, 20148. [Google Scholar] [CrossRef]
  243. Chao, M.P.; Alizadeh, A.A.; Tang, C.; Myklebust, J.H.; Varghese, B.; Gill, S.; Jan, M.; Cha, A.C.; Chan, C.K.; Tan, B.T. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 2010, 142, 699–713. [Google Scholar] [CrossRef] [Green Version]
  244. Saito, Y.; Komohara, Y.; Niino, D.; Horlad, H.; Ohnishi, K.; Takeya, H.; Kawaguchi, H.; Shimizu, H.; Ohshima, K.; Takeya, M. Role of CD204-positive tumor-associated macrophages in adult T-cell leukemia/lymphoma. J. Clin. Exp. Hematop. 2014, 54, 59–65. [Google Scholar] [CrossRef] [Green Version]
  245. Valencia, J.; Fernández-Sevilla, L.M.; Fraile-Ramos, A.; Sacedón, R.; Jiménez, E.; Vicente, A.; Varas, A. Acute lymphoblastic leukaemia cells impair dendritic cell and macrophage differentiation: Role of BMP4. Cells 2019, 8, 722. [Google Scholar] [CrossRef] [Green Version]
  246. McClellan, J.S.; Dove, C.; Gentles, A.J.; Ryan, C.E.; Majeti, R. Reprogramming of primary human Philadelphia chromosome-positive B cell acute lymphoblastic leukemia cells into nonleukemic macrophages. Proc. Natl. Acad. Sci. USA 2015, 112, 4074–4079. [Google Scholar] [CrossRef]
  247. Zhang, J.; Wang, Y.; Yin, C.; Gong, P.; Zhang, Z.; Zhao, L.; Waxman, S.; Jing, Y. Artesunate improves venetoclax plus cytarabine AML cell targeting by regulating the Noxa/Bim/Mcl-1/p-Chk1 axis. Cell Death Dis. 2022, 13, 379. [Google Scholar] [CrossRef] [PubMed]
  248. Hart, S.; Goh, K.; Novotny-Diermayr, V.; Tan, Y.; Madan, B.; Amalini, C.; Ong, L.; Kheng, B.; Cheong, A.; Zhou, J. Pacritinib (SB1518), a JAK2/FLT3 inhibitor for the treatment of acute myeloid leukemia. Blood Cancer J. 2011, 1, e44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Russ, A.; Hua, A.B.; Montfort, W.R.; Rahman, B.; Riaz, I.B.; Khalid, M.U.; Carew, J.S.; Nawrocki, S.T.; Persky, D.; Anwer, F. Blocking “don’t eat me” signal of CD47-SIRPα in hematological malignancies, an in-depth review. Blood Rev. 2018, 32, 480–489. [Google Scholar] [CrossRef]
  250. Daver, N.; Jonas, B.A.; Medeiros, B.C.; Patil, U.; Yan, M. Phase 1b, open-label study evaluating the safety and pharmacokinetics of atezolizumab (anti–PD-L1 antibody) administered in combination with Hu5F9-G4 to patients with relapsed and/or refractory acute myeloid leukemia. Leuk. Lymphoma 2022, 63, 2711–2714. [Google Scholar] [CrossRef] [PubMed]
  251. Kauder, S.E.; Kuo, T.C.; Harrabi, O.; Chen, A.; Sangalang, E.; Doyle, L.; Rocha, S.S.; Bollini, S.; Han, B.; Sim, J. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS ONE 2018, 13, e0201832. [Google Scholar] [CrossRef] [Green Version]
  252. Prawira, A.; Coward, J.; Mislang, A.; Nagrial, A.; Gan, H.; Jin, X.; Li, B.; Wang, Z.M.; Kwek, K.Y.; Xia, D. 384 A Phase 1 study to evaluate the safety, PK, and antitumor activity of AK117, an anti-CD47 monoclonal antibody, in subjects with relapsed/refractory advanced or metastatic solid tumors or lymphomas. J. Immunother. Cancer 2020, 8 (Suppl. 3), A1–A559. [Google Scholar]
  253. Lakhani, N.; Orloff, M.; Fu, S.; Liu, Y.; Wang, Y.; Zhou, H.; Lin, K.; Liu, F.; Yan, S.; Patnaik, A. 295 First-in-human Phase I trial of IBI188, an anti-CD47 targeting monoclonal antibody, in patients with advanced solid tumors and lymphomas. J. ImmunoTher. Cancer 2020, 8, A322. [Google Scholar]
  254. Sallman, D.A.; Donnellan, W.B.; Asch, A.S.; Lee, D.J.; Al Malki, M.; Marcucci, G.; Pollyea, D.A.; Kambhampati, S.; Komrokji, R.S.; Van Elk, J. The first-in-class anti-CD47 antibody Hu5F9-G4 is active and well tolerated alone or with azacitidine in AML and MDS patients: Initial phase 1b results. J. Clin. Oncol. 2019, 37, 7009. [Google Scholar] [CrossRef]
  255. Li, M.; Yu, H.; Qi, F.; Ye, Y.; Hu, D.; Cao, J.; Wang, D.; Mi, L.; Wang, Z.; Ding, N. Anti-CD47 immunotherapy in combination with BCL-2 inhibitor to enhance anti-tumor activity in B-cell lymphoma. Hematol. Oncol. 2022, 40, 596–608. [Google Scholar] [CrossRef] [PubMed]
  256. Sun, M.; Qi, J.; Zheng, W.; Song, L.; Jiang, B.; Wang, Z.; Huang, C.; Tian, W.; Qiu, L. Preliminary results of a first-in-human phase I dtudy of IMM01, SIRPα Fc protein in patients with relapsed or refractory lymphoma. J. Clin. Oncol. 2021, 39, 2550. [Google Scholar] [CrossRef]
  257. Zeidan, A.M.; DeAngelo, D.J.; Palmer, J.M.; Seet, C.S.; Tallman, M.S.; Wei, X.; Li, Y.F.; Hock, N.; Burgess, M.R.; Hege, K. A phase I study of CC-90002, a monoclonal antibody targeting CD47, in patients with relapsed and/or refractory (R/R) acute myeloid leukemia (AML) and high-risk myelodysplastic syndromes (MDS): Final results. Blood 2019, 134, 1320. [Google Scholar] [CrossRef]
  258. Gozlan, Y.M.; Hilgendorf, S.; Aronin, A.; Sagiv, Y.; Ben-gigi-Tamir, L.; Amsili, S.; Tamir, A.; Pecker, I.; Greenwald, S.; Chajut, A. Abstract A076: DSP107—A novel SIRPα-4-1BBL dual signaling protein (DSP) for cancer immunotherapy. Cancer Immunol. Res. 2019, 7, A076. [Google Scholar] [CrossRef]
  259. Daver, N.; Vyas, P.; Chao, M.; Xing, G.; Renard, C.; Ramsingh, G.; Sallman, D.A.; Wei, A.H. A Phase 3, Randomized, Open-Label Study Evaluating the Safety and Efficacy of Magrolimab in Combination with Azacitidine in Previously Untreated Patients with TP53-Mutant Acute Myeloid Leukemia. Blood 2021, 138, 3426. [Google Scholar] [CrossRef]
  260. Moskowitz, C.H.; Younes, A.; de Vos, S.; Bociek, R.G.; Gordon, L.I.; Witzig, T.E.; Gascoyne, R.D.; West, B.; Nolop, K.; Steidl, C. CSF1R inhibition by PLX3397 in patients with relapsed or refractory Hodgkin lymphoma: Results from a phase 2 single agent clinical trial. Blood 2012, 120, 1638. [Google Scholar] [CrossRef]
  261. Straus, D.J.; Długosz-Danecka, M.; Connors, J.M.; Alekseev, S.; Illés, Á.; Picardi, M.; Lech-Maranda, E.; Feldman, T.; Smolewski, P.; Savage, K.J. Brentuximab vedotin with chemotherapy for stage III or IV classical Hodgkin lymphoma (ECHELON-1): 5-year update of an international, open-label, randomised, phase 3 trial. Lancet Haematol. 2021, 8, e410–e421. [Google Scholar] [CrossRef]
  262. Younes, A.; Oki, Y.; Bociek, R.G.; Kuruvilla, J.; Fanale, M.; Neelapu, S.; Copeland, A.; Buglio, D.; Galal, A.; Besterman, J. Mocetinostat for relapsed classical Hodgkin’s lymphoma: An open-label, single-arm, phase 2 trial. Lancet Oncol. 2011, 12, 1222–1228. [Google Scholar] [CrossRef] [Green Version]
  263. Advani, R.; Flinn, I.; Popplewell, L.; Forero, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.; Roschewski, M.; LaCasce, A.; Collins, G.P. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N. Engl. J. Med. 2018, 379, 1711–1721. [Google Scholar] [CrossRef]
  264. Roschewski, M.; Izumi, R.; Hamdy, A.; Patel, M.R.; Arkenau, H.-T.; de Vos, S.; Reagan, P.M.; Zinzani, P.L.; Davies, A.; Pagel, J.M. PRISM: A Platform Protocol for the Treatment of Relapsed/Refractory Aggressive Non-Hodgkin Lymphoma. Blood 2019, 134, 2869. [Google Scholar] [CrossRef]
  265. Yu, J.; Li, S.; Chen, D.; Liu, D.; Guo, H.; Yang, C.; Zhang, W.; Zhang, L.; Zhao, G.; Tu, X. IMM0306, a fusion protein of CD20 mAb with the CD47 binding domain of SIRPα, exerts excellent cancer killing efficacy by activating both macrophages and NK cells via blockade of CD47-SIRPα interaction and FcɣR engagement by simultaneously binding to CD47 and CD20 of B cells. Leukemia 2022, 37, 695–698. [Google Scholar]
  266. Kim, T.M.; Lakhani, N.; Gainor, J.; Kamdar, M.; Fanning, P.; Squifflet, P.; Jin, F.; Forgie, A.J.; Wan, H.; Pons, J. ALX148, a CD47 blocker, in combination with rituximab in patients with non-Hodgkin lymphoma. Blood 2020, 136, 13–14. [Google Scholar] [CrossRef]
  267. Behrens, L.M.; van den Berg, T.K.; van Egmond, M. Targeting the CD47-SIRPα Innate Immune Checkpoint to Potentiate Antibody Therapy in Cancer by Neutrophils. Cancers 2022, 14, 3366. [Google Scholar] [CrossRef]
  268. De Vos, S.; Forero-Torres, A.; Ansell, S.M.; Kahl, B.; Cheson, B.D.; Bartlett, N.L.; Furman, R.R.; Winter, J.N.; Kaplan, H.; Timmerman, J. A phase II study of dacetuzumab (SGN-40) in patients with relapsed diffuse large B-cell lymphoma (DLBCL) and correlative analyses of patient-specific factors. J. Hematol. Oncol. 2014, 7, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Patel, K.; Orlowski, R.Z.; Doucette, K.; Maris, M.; Pianko, M.J.; Ramchandren, R.; Stevens, D.A.; Vesole, D.H.; Uger, R.A.; Scheuber, A. TTI-622-01: A phase 1a/1b dose-escalation and expansion trial of TTI-622 in patients with advanced hematologic malignancies, including multiple myeloma. J. Clin. Oncol. 2022, 40, TPS8071. [Google Scholar] [CrossRef]
  270. Wilson, W.C.; Richards, J.; Puro, R.J.; Andrejeva, G.; Capoccia, B.J.; Donio, M.J.; Hiebsch, R.R.; Chakraborty, P.; Sung, V.; Pereira, D.S. AO-176, a highly differentiated clinical stage anti-CD47 antibody, exerts potent anti-tumor activity in preclinical models of multiple myeloma as a single agent and in combination with approved therapeutics. Blood 2020, 136, 3–4. [Google Scholar] [CrossRef]
  271. Li, W.; Wang, F.; Guo, R.; Bian, Z.; Song, Y. Targeting macrophages in hematological malignancies: Recent advances and future directions. J. Hematol. Oncol. 2022, 15, 110. [Google Scholar] [CrossRef]
  272. Hansson, M.; Gimsing, P.; Badros, A.; Niskanen, T.M.; Nahi, H.; Offner, F.; Salomo, M.; Sonesson, E.; Mau-Sorensen, M.; Stenberg, Y. A Phase I Dose-Escalation Study of Antibody BI-505 in Relapsed/Refractory Multiple MyelomaPhase I Trial with Antibody BI-505 in Myeloma. Clin. Cancer Res. 2015, 21, 2730–2736. [Google Scholar] [CrossRef] [Green Version]
  273. Kaur, S.; Cicalese, K.V.; Banerjee, R.; Roberts, D.D. Preclinical and clinical development of therapeutic antibodies targeting functions of CD47 in the tumor microenvironment. Antibody Ther. 2020, 3, 179–192. [Google Scholar] [CrossRef] [PubMed]
  274. Xie, Y.; Yang, H.; Yang, C.; He, L.; Zhang, X.; Peng, L.; Zhu, H.; Gao, L. Role and Mechanisms of Tumor-Associated Macrophages in Hematological Malignancies. Front. Oncol. 2022, 12, 933666. [Google Scholar] [CrossRef] [PubMed]
  275. Song, J.X.; Wen, Y.; Li, R.W.; Dong, T.; Tang, Y.F.; Zhang, J.J.; Sa, Y.L. Phenotypic characterization of macrophages in the BMB sample of human acute leukemia. Ann. Hematol. 2020, 99, 539–547. [Google Scholar] [CrossRef] [PubMed]
  276. Komohara, Y.; Niino, D.; Saito, Y.; Ohnishi, K.; Horlad, H.; Ohshima, K.; Takeya, M. Clinical significance of CD 163+ tumor-associated macrophages in patients with adult T-cell leukemia/lymphoma. Cancer Sci. 2013, 104, 945–951. [Google Scholar] [CrossRef]
  277. Hohtari, H.; Brück, O.; Blom, S.; Turkki, R.; Sinisalo, M.; Kovanen, P.E.; Kallioniemi, O.; Pellinen, T.; Porkka, K.; Mustjoki, S. Immune cell constitution in bone marrow microenvironment predicts outcome in adult ALL. Leukemia 2019, 33, 1570–1582. [Google Scholar] [CrossRef] [Green Version]
  278. Chen, S.-Y.; Yang, X.; Feng, W.-L.; Liao, J.-F.; Wang, L.-N.; Feng, L.; Lin, Y.-M.; Ren, Q.; Zheng, G.-G. Organ-specific microenvironment modifies diverse functional and phenotypic characteristics of leukemia-associated macrophages in mouse T cell acute lymphoblastic leukemia. J. Immunol. 2015, 194, 2919–2929. [Google Scholar] [CrossRef] [Green Version]
  279. Saint Fleur-Lominy, S.; Maus, M.; Vaeth, M.; Lange, I.; Zee, I.; Suh, D.; Liu, C.; Wu, X.; Tikhonova, A.; Aifantis, I. STIM1 and STIM2 mediate cancer-induced inflammation in T cell acute lymphoblastic leukemia. Cell Rep. 2018, 24, 3045–3060. [Google Scholar] [CrossRef] [Green Version]
  280. Watts, J.; Nimer, S. Recent advances in the understanding and treatment of acute myeloid leukemia. F1000Research 2018, 7, F1000 Faculty Rev-1196. [Google Scholar] [CrossRef] [Green Version]
  281. Heuser, M.; Ofran, Y.; Boissel, N.; Mauri, S.B.; Craddock, C.; Janssen, J.; Wierzbowska, A.; Buske, C. Acute myeloid leukaemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2020, 31, 697–712. [Google Scholar] [CrossRef] [PubMed]
  282. Sallman, D.; Asch, A.; Kambhampati, S.; Al Malki, M.; Zeidner, J.; Donnellan, W.; Lee, D.; Vyas, P.; Jeyakumar, D.; Mannis, G. AML-196: The first-in-class anti-CD47 antibody magrolimab in combination with azacitidine is well tolerated and effective in AML patients: Phase 1b results. Clin. Lymphoma Myeloma Leuk. 2021, 21, S290. [Google Scholar] [CrossRef]
  283. Moore, J.A.; Mistry, J.J.; Hellmich, C.; Horton, R.H.; Wojtowicz, E.E.; Jibril, A.; Jefferson, M.; Wileman, T.; Beraza, N.; Bowles, K.M.; et al. LC3-associated phagocytosis in bone marrow macrophages suppresses acute myeloid leukemia progression through STING activation. J. Clin. Investig. 2022, 132, e153157. [Google Scholar] [CrossRef]
  284. Dalton, W.B.; Ghiaur, G.; Resar, L.M. Taking the STING out of acute myeloid leukemia through macrophage-mediated phagocytosis. J. Clin. Investig. 2022, 132, e157434. [Google Scholar] [CrossRef] [PubMed]
  285. Alves da Silva, P.H.; Xing, S.; Kotini, A.G.; Papapetrou, E.P.; Song, X.; Wucherpfennig, K.W.; Mascarenhas, J.; Ferrari de Andrade, L. MICA/B antibody induces macrophage-mediated immunity against acute myeloid leukemia. Blood 2022, 139, 205–216. [Google Scholar] [CrossRef] [PubMed]
  286. Herbrich, S.; Baran, N.; Cai, T.; Weng, C.; Aitken, M.J.L.; Post, S.M.; Henderson, J.; Shi, C.; Richard-Carpentier, G.; Sauvageau, G.; et al. Overexpression of CD200 is a Stem Cell-Specific Mechanism of Immune Evasion in AML. J. Immunother. Cancer 2021, 9, e002968corr1. [Google Scholar] [CrossRef] [PubMed]
  287. Que, Y.; Li, H.; Lin, L.; Zhu, X.; Xiao, M.; Wang, Y.; Zhu, L.; Li, D. Study on the Immune Escape Mechanism of Acute Myeloid Leukemia with DNMT3A Mutation. Front. Immunol. 2021, 12, 653030. [Google Scholar] [CrossRef] [PubMed]
  288. Peña-Martínez, P.; Ramakrishnan, R.; Högberg, C.; Jansson, C.; Nord, D.G.; Järås, M. Interleukin 4 promotes phagocytosis of murine leukemia cells counteracted by CD47 upregulation. Haematologica 2022, 107, 816–824. [Google Scholar] [CrossRef]
  289. Liu, J.; Wei, Y.; Jia, W.; Can, C.; Wang, R.; Yang, X.; Gu, C.; Liu, F.; Ji, C.; Ma, D. Chenodeoxycholic acid suppresses AML progression through promoting lipid peroxidation via ROS/p38 MAPK/DGAT1 pathway and inhibiting M2 macrophage polarization. Redox Biol. 2022, 56, 102452. [Google Scholar] [CrossRef]
  290. Li, Q.; Liang, C.; Xu, X.; Zhang, C.; Cao, W.; Wang, M.; Jiang, Z.; Xing, H.; Yu, J. CLEC12A plays an important role in immunomodulatory function and prognostic significance of patients with acute myeloid leukemia. Leuk. Lymphoma 2022, 63, 2136–2148. [Google Scholar] [CrossRef]
  291. Hu, S.; Wang, J.; Zhang, Y.; Bai, H.; Wang, C.; Wang, N.; He, L. Three salvianolic acids inhibit 2019-nCoV spike pseudovirus viropexis by binding to both its RBD and receptor ACE2. J. Med. Virol. 2021, 93, 3143–3151. [Google Scholar] [CrossRef]
  292. Li, C.; Menoret, A.; Farragher, C.; Ouyang, Z.; Bonin, C.; Holvoet, P.; Vella, A.T.; Zhou, B. Single-cell transcriptomics–based MacSpectrum reveals macrophage activation signatures in diseases. JCI Insight 2019, 4, e126453. [Google Scholar] [CrossRef]
  293. Schürch, C.M.; Bhate, S.S.; Barlow, G.L.; Phillips, D.J.; Noti, L.; Zlobec, I.; Chu, P.; Black, S.; Demeter, J.; McIlwain, D.R. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 2020, 182, 1341–1359.e19. [Google Scholar] [CrossRef]
  294. Miari, K.E.; Guzman, M.L.; Wheadon, H.; Williams, M.T. Macrophages in Acute Myeloid Leukaemia: Significant Players in Therapy Resistance and Patient Outcomes. Front. Cell Dev. Biol. 2021, 9, 692800. [Google Scholar] [CrossRef] [PubMed]
  295. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood J. Am. Soc. Hematol. 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Burger, J.A.; Tsukada, N.; Burger, M.; Zvaifler, N.J.; Dell’Aquila, M.; Kipps, T.J. Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell–derived factor-1. Blood J. Am. Soc. Hematol. 2000, 96, 2655–2663. [Google Scholar]
  297. Mesaros, O.; Jimbu, L.; Neaga, A.; Popescu, C.; Berceanu, I.; Tomuleasa, C.; Fetica, B.; Zdrenghea, M. Macrophage polarization in chronic lymphocytic leukemia: Nurse-like cells are the caretakers of leukemic cells. Biomedicines 2020, 8, 516. [Google Scholar] [CrossRef]
  298. Cols, M.; Barra, C.M.; He, B.; Puga, I.; Xu, W.; Chiu, A.; Tam, W.; Knowles, D.M.; Dillon, S.R.; Leonard, J.P. Stromal endothelial cells establish a bidirectional crosstalk with chronic lymphocytic leukemia cells through the TNF-related factors BAFF, APRIL, and CD40L. J. Immunol. 2012, 188, 6071–6083. [Google Scholar] [CrossRef] [Green Version]
  299. Boissard, F.; Fournie, J.; Quillet-Mary, A.; Ysebaert, L.; Poupot, M. Nurse-like cells mediate ibrutinib resistance in chronic lymphocytic leukemia patients. Blood Cancer J. 2015, 5, e355. [Google Scholar] [CrossRef] [Green Version]
  300. Van Attekum, M.; Terpstra, S.; Reinen, E.; Kater, A.; Eldering, E. Macrophage-mediated chronic lymphocytic leukemia cell survival is independent of APRIL signaling. Cell Death Discov. 2016, 2, 16020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  301. Boissard, F.; Fournié, J.J.; Laurent, C.; Poupot, M.; Ysebaert, L. Nurse like cells: Chronic lymphocytic leukemia associated macrophages. Leuk. Lymphoma 2015, 56, 1570–1572. [Google Scholar] [CrossRef] [PubMed]
  302. Domagala, M.; Ysebaert, L.; Ligat, L.; Lopez, F.; Fournié, J.J.; Laurent, C.; Poupot, M. IL-10 Rescues CLL Survival through Repolarization of Inflammatory Nurse-like Cells. Cancers 2021, 14, 16. [Google Scholar] [CrossRef]
  303. Hančić, S.; Gršković, P.; Gašparov, S.; Ostojić Kolonić, S.; Dominis, M.; Korać, P. Macrophage Infiltration Correlates with Genomic Instability in Classic Hodgkin Lymphoma. Biomedicines 2022, 10, 579. [Google Scholar] [CrossRef]
  304. Gusak, A.; Fedorova, L.; Lepik, K.; Volkov, N.; Popova, M.; Moiseev, I.; Mikhailova, N.; Baykov, V.; Kulagin, A. Immunosuppressive Microenvironment and Efficacy of PD-1 Inhibitors in Relapsed/Refractory Classic Hodgkin Lymphoma: Checkpoint Molecules Landscape and Macrophage Populations. Cancers 2021, 13, 5676. [Google Scholar] [CrossRef]
  305. Karihtala, K.; Leivonen, S.K.; Brück, O.; Karjalainen-Lindsberg, M.L.; Mustjoki, S.; Pellinen, T.; Leppä, S. Prognostic Impact of Tumor-Associated Macrophages on Survival Is Checkpoint Dependent in Classical Hodgkin Lymphoma. Cancers 2020, 12, 877. [Google Scholar] [CrossRef] [Green Version]
  306. Rashed, R.A.; Zaki, M.A.M.; Mohamed, N.A.W.; Mansou, O.M.; Refaey, F. Prognostic Value of Tumor Associated Macrophage Markers CD163 and CD68 Immunohistochemistry in Classical Hodgkin Lymphoma. Clin. Lab. 2021, 67, 200920. [Google Scholar] [CrossRef]
  307. Abd Allah, M.Y.Y.; Fahmi, M.W.; El-Ashwah, S. Clinico-pathological significance of immunohistochemically marked tumor-associated macrophage in classic Hodgkin lymphoma. J. Egypt. Natl. Cancer Inst. 2020, 32, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  308. Procházka, V.; Papajík, T.; Dýšková, T.; Dihel, M.; Brychtová, S.; Prouzová, Z.; Kriegová, E.; Lukášová, M.; Hanáčková, V. The Lymphoma-Associated Macrophage to Hodgkin-Reed-Sternberg Cell Ratio Is a Poor Prognostic Factor in Classic Hodgkin Lymphoma Patients. Clin. Lymphoma Myeloma Leuk. 2019, 19, e573–e580. [Google Scholar] [CrossRef] [PubMed]
  309. Tamma, R.; Ingravallo, G.; Annese, T.; Gaudio, F.; Perrone, T.; Musto, P.; Specchia, G.; Ribatti, D. Tumor Microenvironment and Microvascular Density in Follicular Lymphoma. J. Clin. Med. 2022, 11, 1257. [Google Scholar] [CrossRef] [PubMed]
  310. Ruan, J.; Ouyang, M.; Zhang, W.; Luo, Y.; Zhou, D. The effect of PD-1 expression on tumor-associated macrophage in T cell lymphoma. Clin. Transl. Oncol. 2021, 23, 1134–1141. [Google Scholar] [CrossRef] [PubMed]
  311. Cencini, E.; Fabbri, A.; Schiattone, L.; Sicuranza, A.; Mecacci, B.; Granai, M.; Mancini, V.; Lazzi, S.; Bocchia, M.; Leoncini, L. Prognostic impact of tumor-associated macrophages, lymphocyte-to-monocyte and neutrophil-to-lymphocyte ratio in diffuse large B-cell lymphoma. Am. J. Blood Res. 2020, 10, 97–108. [Google Scholar] [CrossRef] [Green Version]
  312. Zhang, D.; Hamdoun, S.; Chen, R.; Yang, L.; Ip, C.K.; Qu, Y.; Li, R.; Jiang, H.; Yang, Z.; Chung, S.K. Identification of natural compounds as SARS-CoV-2 entry inhibitors by molecular docking-based virtual screening with bio-layer interferometry. Pharmacol. Res. 2021, 172, 105820. [Google Scholar] [CrossRef]
  313. Liu, M.K.; Cheng, L.L.; Yi, H.M.; He, Y.; Li, X.; Fu, D.; Dai, Y.T.; Fang, H.; Cheng, S.; Xu, P.P.; et al. Enhanced lipid metabolism confers the immunosuppressive tumor microenvironment in CD5-positive non-MYC/BCL2 double expressor lymphoma. Front. Oncol. 2022, 12, 885011. [Google Scholar] [CrossRef] [PubMed]
  314. Vegliante, M.C.; Mazzara, S.; Zaccaria, G.M.; De Summa, S.; Esposito, F.; Melle, F.; Motta, G.; Sapienza, M.R.; Opinto, G.; Volpe, G.; et al. NR1H3 (LXRα) is associated with pro-inflammatory macrophages, predicts survival and suggests potential therapeutic rationales in diffuse large b-cell lymphoma. Hematol. Oncol. 2022, 40, 864–875. [Google Scholar] [CrossRef] [PubMed]
  315. Beider, K.; Voevoda-Dimenshtein, V.; Zoabi, A.; Rosenberg, E.; Magen, H.; Ostrovsky, O.; Shimoni, A.; Weiss, L.; Abraham, M.; Peled, A.; et al. CXCL13 chemokine is a novel player in multiple myeloma osteolytic microenvironment, M2 macrophage polarization, and tumor progression. J. Hematol. Oncol. 2022, 15, 144. [Google Scholar] [CrossRef] [PubMed]
  316. Yan, H.; He, D.; Qu, J.; Liu, Y.; Xu, R.; Gu, H.; Chen, J.; Li, Y.; Zhang, E.; Zhao, Y.; et al. Interleukin-32γ promotes macrophage-mediated chemoresistance by inducing CSF1-dependent M2 macrophage polarization in multiple myeloma. Cancer Immunol. Immunother. 2022, 72, 327–338. [Google Scholar] [CrossRef] [PubMed]
  317. Zhang, J.; Liu, Z.; Cao, P.; Wang, H.; Liu, H.; Hua, L.; Xue, H.; Fu, R. Tumor-associated macrophages regulate the function of cytotoxic T lymphocyte through PD-1/PD-L1 pathway in multiple myeloma. Cancer Med. 2022, 11, 4838–4848. [Google Scholar] [CrossRef]
  318. Liu, Y.; Yan, H.; Gu, H.; Zhang, E.; He, J.; Cao, W.; Qu, J.; Xu, R.; Cao, L.; He, D.; et al. Myeloma-derived IL-32γ induced PD-L1 expression in macrophages facilitates immune escape via the PFKFB3-JAK1 axis. Oncoimmunology 2022, 11, 2057837. [Google Scholar] [CrossRef]
  319. Gao, Y.; Li, L.; Zheng, Y.; Zhang, W.; Niu, B.; Li, Y. Monoclonal antibody Daratumumab promotes macrophage-mediated anti-myeloma phagocytic activity via engaging FC gamma receptor and activation of macrophages. Mol. Cell. Biochem. 2022, 477, 2015–2024. [Google Scholar] [CrossRef]
  320. Yuan, G.; Huang, Y.; Yang, S.T.; Ng, A.; Yang, S. RGS12 inhibits the progression and metastasis of multiple myeloma by driving M1 macrophage polarization and activation in the bone marrow microenvironment. Cancer Commun. 2022, 42, 60–64. [Google Scholar] [CrossRef]
  321. Feng, X.; Szulzewsky, F.; Yerevanian, A.; Chen, Z.; Heinzmann, D.; Rasmussen, R.D.; Alvarez-Garcia, V.; Kim, Y.; Wang, B.; Tamagno, I. Loss of CX3CR1 increases accumulation of inflammatory monocytes and promotes gliomagenesis. Oncotarget 2015, 6, 15077. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The released DAMPS due to cell death within the tumors can activate macrophages and result in immunosuppression via the elevated production of IL-10, which suppresses the expression of pro-inflammatory cytokines and promotes the formation of Tregs. Moreover, macrophages can contribute to the secretion of IL-6 and TNFα, leading to cancerous cell survival. In addition, TNFα activates the NF-κB pathway upon binding to the TNFR1/2 receptor, controls the expression of VEGF, IL-6, and the stimulation of neo-angiogenesis. IL-6 could then promote cell proliferation and differentiation via the JAK/STAT3 pathway. Phagocytosis, the propagation of TNFα, and interleukin-1 beta (IL1β) can induce macrophage recruitment.
Figure 1. The released DAMPS due to cell death within the tumors can activate macrophages and result in immunosuppression via the elevated production of IL-10, which suppresses the expression of pro-inflammatory cytokines and promotes the formation of Tregs. Moreover, macrophages can contribute to the secretion of IL-6 and TNFα, leading to cancerous cell survival. In addition, TNFα activates the NF-κB pathway upon binding to the TNFR1/2 receptor, controls the expression of VEGF, IL-6, and the stimulation of neo-angiogenesis. IL-6 could then promote cell proliferation and differentiation via the JAK/STAT3 pathway. Phagocytosis, the propagation of TNFα, and interleukin-1 beta (IL1β) can induce macrophage recruitment.
Cancers 15 03722 g001
Figure 2. Role of macrophages in angiogenesis. Macrophages are considered to play a critical role in inflammatory and tumor angiogenesis due to their widespread existence in healthy and inflamed tissues, their ability to become activated in response to certain stimuli, and their production of various secretory agents. Alternatively activated macrophages could release proteases, various growth factors, and monokines, including TGF-alpha, bFGF, GM-CSF, VEGF/VPF, IGF-I, PDGF, TGF-beta, IL-1, IL-6, substance P, IL-8, TNF-alpha, interferons, prostaglandins, and thrombospondin 1. These factors can influence each phase of angiogenesis, such as the induction of endothelial cell migration or proliferation, changes to the local extracellular matrix, and inhibition of vascular growth via the formation of differentiated capillaries.
Figure 2. Role of macrophages in angiogenesis. Macrophages are considered to play a critical role in inflammatory and tumor angiogenesis due to their widespread existence in healthy and inflamed tissues, their ability to become activated in response to certain stimuli, and their production of various secretory agents. Alternatively activated macrophages could release proteases, various growth factors, and monokines, including TGF-alpha, bFGF, GM-CSF, VEGF/VPF, IGF-I, PDGF, TGF-beta, IL-1, IL-6, substance P, IL-8, TNF-alpha, interferons, prostaglandins, and thrombospondin 1. These factors can influence each phase of angiogenesis, such as the induction of endothelial cell migration or proliferation, changes to the local extracellular matrix, and inhibition of vascular growth via the formation of differentiated capillaries.
Cancers 15 03722 g002
Figure 3. TAMs in immunotherapy.
Figure 3. TAMs in immunotherapy.
Cancers 15 03722 g003
Figure 4. Macrophage and hematologic cancers. Interactions between macrophages residing in the TME and tumor cells finally result in polarization toward M2 macrophages which can provide insights regarding disease progression. Various small peptides belonging to the CXC chemokine family (including chemokine (C-X-C motif) ligand 10 (CXCL 10), CXCL11, and CXCL 13 and 16), the CC chemokine family (including chemokine ligand 1 (CCL1), CCL5, 17, 18, 22, and CCL24) are produced during this process. In addition, transforming growth factor beta (TGF-β), vascular endothelial growth factor (VEGF), tumor necrosis factor alpha (TNFα), interleukin-1α (IL-1α), 1 β, 10, 12, and IL-23 can also contribute to different functions, as shown.
Figure 4. Macrophage and hematologic cancers. Interactions between macrophages residing in the TME and tumor cells finally result in polarization toward M2 macrophages which can provide insights regarding disease progression. Various small peptides belonging to the CXC chemokine family (including chemokine (C-X-C motif) ligand 10 (CXCL 10), CXCL11, and CXCL 13 and 16), the CC chemokine family (including chemokine ligand 1 (CCL1), CCL5, 17, 18, 22, and CCL24) are produced during this process. In addition, transforming growth factor beta (TGF-β), vascular endothelial growth factor (VEGF), tumor necrosis factor alpha (TNFα), interleukin-1α (IL-1α), 1 β, 10, 12, and IL-23 can also contribute to different functions, as shown.
Cancers 15 03722 g004
Table 1. Different strategies in targeting macrophages for treatment.
Table 1. Different strategies in targeting macrophages for treatment.
StrategyPathwayTargetAgent/Drug(s)Type of TumorResult(s)Ref. or Trial No.
ConventionalTAM depletionCSF-1RBLZ945Solid tumorsEnhancing the level of CD8+ cytotoxic T cells leading to the prevention of tumor growth[94]
CSF-1RPLX3397 (Pexidartinib)Sarcoma, breast cancer, prostate cancer, and solid tumorsInfiltration of T cells in the TME[95,96]
IL10/VEGF/TGFβZoledronic acidBreast cancerInfiltration of CD8+ T cells and improving immune responses[97]
Pan-macrophagesTrabectedin Soft tissue sarcomas and recurrent ovarian cancerCausing selective cytotoxicity to TAM populations by triggering the extrinsic TRAIL apoptotic pathway[98,99,100,101]
Pan-macrophagesLurbinectedinOvarian cancer and solid tumorsEliminating tumor cells directly through the TRAIL-dependent apoptosis pathway and a reduction in angiogenesis[102,103,104,105]
CSF1-RARRY-382Solid tumorsNot determined[106]
CSF1-RAMG820CRC and solid tumorsNot determined[106,107]
CSF1-REmactuzumab Solid tumors Inhibiting the activation of CSF1R [108]
MMP-2Doxorubicin Melanoma, Breast cancerReduction in Treg infiltration to the TME[109]
MMP-2Clodronate Bone
metastatic cancers
Suppressing tumor growth and angiogenesis[110]
TargetDrug(s)Type of tumorResult(s)Ref. or Trial no.
Blocking recruitmentCCL2Carlumab Prostate cancerBlocking CCL2 signaling leading to tumor growth prevention[111]
CCL2CNTO 888Solid tumorsCCL2 inhibition[112]
CCR2PropagermaniumBreast cancerCCL2 inhibition[113]
CCR2PF-04136309Pancreatic cancerReducing the circulatory CCR2+ monocytes and an increase in bone marrow CCR2+ monocytes [114]
CCR2BMS-813160CRC and pancreatic cancerInhibition of inflammatory monocytes and macrophages migration[115]
CCR5LeronlimabBreast cancerInhibition of tumor development, adhesion, and invasion[116,117]
CCL5Maraviroc Metastatic colorectal cancerInducing M1 like TAMs polarization, which mediated antitumor responses [118]
CCR5VicrivirocMetastatic colorectal cancerCCR5 inhibition[119]
CCR5TAK-779Colorectal cancer CCR5 inhibition[120]
CCR5AnibamineOvarian cancer cells CCR5 inhibition[121]
CCR5GSK706769Colorectal cancer CCR5 inhibition[122]
CX3CL1DC101/anti-Ly6G antibodyColon cancerInhibition of macrophage recruitment in the TME[123]
CSFR-1RPexiddartinib (PLX-3397)Tenosynovial giant cell tumors and other solid tumorsCSF-1R inhibition[124]
CSFR-1RChiauranib (CS2164)Solid tumorsInhibition of CSF-1R and angiogenesis-related kinases (VEGFR, PDGFRa, and c-Kit)[125]
CSFR-1RRG7155Solid tumorsReducing CSF-1R, CD163, TAMs, and peripheral blood CCR2Monocyte[126]
CSFR-1RCabiralizumabSolid tumorsCSF-1R inhibition[127]
CSFR-1RAZD7507 Pancreatic cancerCSF-1R inhibition[128]
CX3CL1JMS-17-2Breast cancer cellsMetastatic seeding and colonization
of breast cancer cells
[129]
TAM repolarizationTargetDrug(s)Type of tumorResult(s)Ref. or Trial no.
CD40CP-870 and 893Melanoma, pancreatic cancer, and solid tumorsStimulation of adaptive immune responses and M1 macrophage activation and cancer cell apoptosis [130,131,132,133]
NCT02225002
CD40APX005M (Sotigalimab)Melanoma and pancreatic cancerInducing T cell-dependent tumor regression and improving survival[134]
NCT02706353
CD40CDX-1140Melanoma and breast cancerActivating DCs and B cells and leading to NFkB stimulation in CD40-expressing cells[135]
NCT04616248
CD40SEA-CD40 Solid and hematological tumorsBinding with increased affinity to FcγRIIIa resulting in an enhanced effector function and CD40 agonism[136]
NCT02376699
CD47/SIRPαTTI-621Hematological malignanciesIncreasing cancer cells phagocytosis by macrophages and antigen presentation which activate T cells[137,138]
CD47/SIRPαMagrolimabSolid and hematological tumorsCD47 inhibition[139]
CD47/SIRPαHu5F9-G4Solid tumorsCD47 inhibition[140]
CD47/SIRPαIBI188Solid tumorsCD47 inhibition[141]
CD47/SIRPαZL1201Solid tumorsCD47 inhibition[141]
CD47/SIRPαBI 765063Solid tumorsSIRPα inhibitors[141]
CD47/SIRPαCC-9525Solid tumorsSIRPα inhibitors[141]
CD47/SIRPαChiLob7/4Various tumorsCD40 agonists [142]
CSFR-1RBLZ945Solid tumorsReducing M2 associated gene expression (Adm, Arg, Mrc1, and F13a1) in TAMs [143]
CSFR-1RPLX3397GliomaTAM repolarization and consequent tumor suppression [144]
-Membrane-coated Fe3O4 nanoparticleMelanoma Re-educating M2-macrophages to M1, decreasing cancer’s metabolic function, and induction of immunologic cell death[145]
TLR7/8 agonistResiquimod Melanoma M2 repolarization into M1 and elevating the level of antibody-dependent cellular phagocytosis[146,147]
TLR7/8 agonistTransConSolid tumorsEnhancing tumor growth inhibition[148]
TLR3 agonistBO-112CRC, gastric cancer, and melanomaRe-education of M2 macrophages towards M1 and inhibition of tumor growth[149,150]
NCT04508140
TLR9 agonistCMP-001 (vidutolimod)MelanomaUpregulating IFN-responsive genes [151]
TLR7 agonistSHR2150Solid tumorsImmunostimulating and antineoplastic activitiesNCT04588324
PI3KIPI-549Solid tumorsEnhancing NFκB activation preventing tumor growth and elevated cytotoxic T cell activity[152,153]
MARCOMARCO mAbMelanoma, colon, and breast cancerUpregulating the level of regulatory T cells and anti-inflammatory cytokine IL-37, decreasing tumor growth [154,155]
Ferumoxytol nanoparticles Carboxy-dextran coated super paramagnetic ironoxide nanoparticles (SPIONs) -Inducing TAMs phenotypic shift towards tumor-suppressive phenotype and activation of the MAPK pathway [156]
Polystyrene nanoparticles functionalized with carboxyl or amino groups poly(styrene-co-maleic anhydride) (PSMA) nanoparticles conjugated with polymer poly [2-methoxy-5-(2-ethylhexyloxy)-1,4-phenylenevinylene, PPV] -Impairing CD163 and CD200R expression and IL-10 production in M2 macrophages [157]
Cationic polymers Cationic dextran and polyethyleneimine (PEI) SarcomaChanging TAM phenotype via TLR4 signaling [158]
TLR7/8 agonist R848 (TLR7/8 agonist)-loaded β-cyclodextrin nanoparticlesColorectal cancerRe-education of M2-macrophages to M1,
enhancing response rates to immunotherapy when combined with the immune checkpoint inhibitor anti-PD-1
[159]
Macrophage cell therapy (CAR-M)TargetDrug(s)Type of tumorResult(s)Ref. or Trial No.
HER2CT-0508HER2+ solid tumorsTrafficking into the tumor, phagocytosing and killing cancer cellsNCT04660929
-TEMFERONGlioblastomaTemferon is well tolerated by patients[160]
Immune checkpoint blockade (ICB) immunotherapyPDL-1-NSCLC and other tumorsEnhancing the cytotoxic function of T cells[161]
VISTA-Myeloid cellsInteracting with P-selectin glycoprotein ligand 1 (PSGL1), functioning as a T cell checkpoint inhibitory ligand[162]
TIM4-Renal cell carcinoma (RCC)Suppressing CD8+ T cell responses, blocking TIM4 with antibodies, and enhancing the efficacy of ICB at these sites[163]
Vaccine -Exosomes derived from M1- but not M2-polarized macrophages-Boosting the antitumor vaccine by eliciting a release of Th1 cytokines and a stronger antigen-specific cytotoxic T cell response[164]
Indoleamine 2,3-dioxygenase (IDO)-Non-small cell lung cancerEliciting CD8+ and CD4+ T cell-mediation[165]
Sipuleucel-T-Prostate cancerInducing antigen-specific T cells with a fusion protein combining a targeting tumor antigen prostate acid phosphatase with GM-CSF, prolonging the survival of patients in a few clinical trials[166]
STING agonist-Multiple established tumors-[167]
Apoptotic peptides CD206 M2pepColon cancerMurine TAMs (CD45+F4/80+CD301+)[168]
CD206 UNOSolid tumorsCD206 TAMs
binding to CD206+ (M2) macrophages
[169]
CD206 MelittinSolid tumorsCD206 TAMs[170]
CD206 RP-182Solid tumorsCD206 TAMs[171]
IL-4RIL4RPep-1Breast cancerIL-4R-expressing macrophages[172]
Tyrosine-protein kinase receptor (Tie2)T4
Peptide
Breast cancerTyrosine-protein kinase receptor (Tie2) expressing macrophages (TEMs)[173]
CD-47Pep-20Wilde rangeCD-47[174]
Retinoid X receptor betaCRVBreast tumorsTAMs retinoid X receptor beta, a receptor found to be expressed predominantly by TAMs[175]
Table 2. Macrophage and hematologic malignancies.
Table 2. Macrophage and hematologic malignancies.
Hematological MalignanciesDiseaseDrug/AgentStudy Status/StageMechanism/Observation(s)References
LeukemiaCLLTrabectedinPreclinical evaluationAntiangiogenic and macrophage killing due to CCL2-CCR2 signaling axis inhibition[233]
CSF-1R signaling inhibitorPreclinical evaluationCSF-1R signaling inhibition[234]
GW-2580, ARRY-382Preclinical evaluationCSF-1R signaling inhibition[235]
LenalidomidePreclinical evaluationModifying the TME via promotion of T and NK cell functions and downregulating anti-inflammatory and proangiogenic cytokines [236]
TG-1801 (NI-1701)Clinical/phase ICD47/SIRPα-targeted bispecific antibodies[237]
SRF231Clinical/phase IA/IBCD47 inhibition[238]
ALLBLZ-945Preclinical evaluationCSF-1R signaling inhibition[239]
PLX3397Preclinical evaluationCSF-1R signaling inhibition[240]
CXCR4 inhibitor plerixaforPreclinical evaluationCXCR4/CXCL12 axis inhibition[241]
Preemptive IFN-αPreclinical evaluationTAM reprogramming[242]
Anti-CD47 mAbPreclinical evaluationEnabling phagocytosis of tumor cells by TAM[243]
CD204-positive TAMPreclinical evaluationCD204-positive TAM was associated with malignant cells proliferation, measured according to the Ki-67 labeling index[244]
BMP4Preclinical evaluationInducing immunosuppressive dendritic cells and favoring the generation of M2-like macrophages with pro-tumoral features[245]
Exposure to myeloid differentiation promoting cytokinesPreclinical evaluationB-ALL blasts reprogramming into Macrophage[246]
AMLArtesunatePreclinical evaluationTAM reprogramming JAK2/STAT3 Downregulation[247]
PacritinibPreclinical evaluationCSF1R inhibition with a JAK2/FLT3 inhibitor, depletion of TAMs, and, consequently, inhibited leukemic cell survival[248]
Hu5F9-G4Clinical/phase IAnti-CD47 led to hemoglobin decline and increased transfusion requirements[249]
AMLHu5F9-G4 + AtezolizumabClinical/phase ICD47 inhibition[250]
ALX148Clinical/phase I/IISIRPα fusion protein that blocks CD47[251]
AK117 Clinical/phase I/IICD47 inhibition[252]
IBI188 Clinical/phase IBCD47 inhibition[253]
AML/MDSHu5F9-G4Clinical/phase IICD47 inhibition[254]
TJC4Clinical/phase IBCD47 inhibition[255]
IMM-01Clinical/phase I/IISIRPα fusion protein that blocks CD47[256]
CC-90002Clinical/phase ICD47 inhibition[257]
DSP107Clinical/phase IICD47/SIRPα-targeted bispecific antibodies[258]
TP53 Mutant AMLHu5F9-G4Clinical/phage IIICD47 inhibition[259]
LymphomaHodgkin lymphoma (HL)PLX3397Clinical/phase IIHighly selective inhibitor of CSF1R and Kit receptor tyrosine kinases[260]
Brentuximab VedotinClinical/phase IVAn anti-CD30 antibody–drug conjugate[261]
Mocetinostat Clinical/phase IIAn oral isotype-selective histone deacetylase inhibitor[262]
Non-Hodgkin lymphoma (NHL)Hu5F9-G4 + RituximabClinical/phase IICD47 inhibition[263]
Hu5F9-G4 + Rituximab + AcalabrutinibClinical/phase ICD47 inhibition[264]
IMM0306Clinical/phase ICD47/SIRPα-targeted bispecific antibodies[265]
ALX148Clinical/phase I/IIInhibiting CD47-SIRPα checkpoint[266]
GentulizumabClinical/phase ICD47 inhibition[267]
Anti-CD47 mAb Hu5F9-G4Clinical/phase IIEnabling phagocytosis of tumor cells by TAM[263]
DacetuzumabClinical phase IIAnti-CD40 mAb[268]
MyelomaMMTrabectedinTrabectedinAntiangiogenic and macrophage killing due to CCL2-CCR2 signaling axis
TTI-621Phase IbSIRPα-IgG1 Fc fusion protein inhibiting CD47-SIRPα Checkpoint[137]
TTI-622Phase Ia/IbSIRPα-IgG1 Fc fusion protein inhibiting CD47-SIRPα Checkpoint[269]
AO-176Phase I/IIHumanized IgG2 anti-CD47 mAb inhibiting CD47-SIRPα Checkpoint[270]
SRF231Phase Ia/IbFully human anti-CD47 mAb inhibiting CD47-SIRPα Checkpoint[271]
BI-505Phase IFully human anti-ICAM-1 mAb overcoming immunosuppression[272]
DacetuzumabClinical phase IIAnti-CD40 mAb[268]
IBI-322Clinical/phase ICD47/SIRPα-targeted bispecific antibodies[273]
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

Khalili, S.; Zeinali, F.; Moghadam Fard, A.; Taha, S.R.; Fazlollahpour Naghibi, A.; Bagheri, K.; Shariat Zadeh, M.; Eslami, Y.; Fattah, K.; Asadimanesh, N.; et al. Macrophage-Based Therapeutic Strategies in Hematologic Malignancies. Cancers 2023, 15, 3722. https://doi.org/10.3390/cancers15143722

AMA Style

Khalili S, Zeinali F, Moghadam Fard A, Taha SR, Fazlollahpour Naghibi A, Bagheri K, Shariat Zadeh M, Eslami Y, Fattah K, Asadimanesh N, et al. Macrophage-Based Therapeutic Strategies in Hematologic Malignancies. Cancers. 2023; 15(14):3722. https://doi.org/10.3390/cancers15143722

Chicago/Turabian Style

Khalili, Saeed, Fatemeh Zeinali, Atousa Moghadam Fard, Seyed Reza Taha, Andarz Fazlollahpour Naghibi, Kimia Bagheri, Mahdieh Shariat Zadeh, Yeghaneh Eslami, Khashayar Fattah, Naghmeh Asadimanesh, and et al. 2023. "Macrophage-Based Therapeutic Strategies in Hematologic Malignancies" Cancers 15, no. 14: 3722. https://doi.org/10.3390/cancers15143722

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