Abstract
Cancer immunotherapy has made great progress in effectively attacking or eliminating cancer. However, the challenges posed by the low reactivity of some solid tumors still remain. Macrophages, as a key component of the tumor microenvironment (TME), play an important role in determining the progression of solid tumors due to their plasticity and heterogeneity. Targeting and reprogramming macrophages in TME to desired phenotypes offers an innovative and promising approach for cancer immunotherapy. Meanwhile, the rapid development of in vivo molecular imaging techniques provides us with powerful tools to study macrophages. In this review, we summarize the current progress in macrophage reprogramming from conceptual roadmaps to therapeutic approaches, including monoclonal antibody drugs, small molecule drugs, gene therapy, and chimeric antigen receptor-engineered macrophages (CAR-M). More importantly, we highlight the significance of molecular imaging in observing and understanding the process of macrophage reprogramming during cancer immunotherapy. Finally, we introduce the therapeutic applications of imaging and reprogramming macrophages in three solid tumors. In the future, the integration of molecular imaging into the development of novel macrophage reprogramming strategies holds great promise for precise clinical cancer immunotherapy.
Keywords: Macrophage reprogramming, Imaging techniques, Immunotherapy, Polarized phenotypes, Solid tumors
Introduction
As a large group of diseases, cancers caused 10 million deaths globally in 2020 (Siegel et al. 2021). Up to date, cancer immunotherapy has become a highly debated topic in the field of cancer treatment (Wang et al. 2021). The main focus of immunotherapy is to enhance the immune responses of various immune cells within the tumor microenvironment (TME) to effectively eliminate cancer cells (Li et al. 2021a). Among the immune cells present in TME, macrophages account for over one-third of the total population (Duan and Luo 2021). Therefore, targeting macrophages has shown promising potential in improving the efficacy of cancer immunotherapy.
In general, macrophages are often divided into two types: classically activated M1-type and alternatively activated M2-type. This classic dichotomy is based on various stimuli, surface molecules, cytokine secretion patterns, metabolic signals, and cell–cell interactions (Fig. 1). Macrophages exhibit notable characteristics, primarily their remarkable plasticity and heterogeneity, enabling them to transition between distinct phenotypes and execute a range of functions (Loke and Lin 2022). Although the classical dichotomy of macrophage phenotypes has faced challenges from advanced technologies such as single-cell RNA-seq (scRNA-seq) (Xue et al. 2022), cellular indexing of transcriptomes and epitopes by sequencing (Ma et al. 2022b), mass cytometry (Ding et al. 2023), and hyperspectral image (Strack 2023). The M1/M2 classification remains crucial in many inflammation-related diseases, especially in cancer, which always reduces the M1/M2 ratio to achieve immune evasion (Gharavi et al. 2022; Noonepalle et al. 2023). During early-stage cancer, a strong immune system with powerful adaptive and innate immunity exhibits a higher proportion of M1 type macrophages to surveil and eliminate abnormal cells. This corresponds to a Th1 immune response by secreting higher levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-2, IL-6, IL-12, and IL-23 (Chen et al. 2023).
Fig. 1.
The typical markers and morphology of polarized macrophages. The polarization states of macrophages exhibit various markers and morphologies, represented by the M1 and M2 phenotypes. The representative markers of M1/M2 macrophages consist of cluster of differentiation (CD), interleukin (IL), CC chemokine ligand (CCL), CXC chemokine ligand (CXCL), toll-like receptor (TLR), transcription factors (TFs), and other effectors. In terms of morphology, M1 macrophages typically have a round and flattened appearance. This distinct shape contributes to their slower movement speeds and denser cell-to-cell contacts, which allow them to exert pro-inflammatory effects, such as killing tumors and other infectious pathogens. In contrast, M2 macrophages exhibit more elongated and distinct synapses, facilitating faster movement for tissue repair, angiogenesis, and other anti-inflammatory effects
However, with the development of immune selection, the proliferation of abnormal cells gradually loses its major histocompatibility complex class I (MHC-I) and major histocompatibility complex class II (MHC-II) antigens and decreases the amount of tumor antigens in the equilibrium phase (Lerner et al. 2023). By this time, tumor-derived soluble factors switch M1 type macrophages to M2 type macrophages and shape a tumor-promoting TME, in which M2 type macrophages protect cancer cells from immune cell attack, allowing for their progression and metastasis (Fig. 2) (Qian et al. 2023). Therefore, although the majority of cancer immunotherapies harness T lymphocytes to destroy tumors, it is believed that targeting macrophages to switch their dysfunctional phenotypes also holds promising therapeutic potential in cancer immunotherapy.
Fig. 2.
The intervention of polarized macrophages with cancer cells. During early-stage cancer, a strong immune system with powerful adaptive and innate immunity exhibits a higher percentage of M1 type macrophages to surveil and clear abnormal cells, which correspond to Th1 immune responses by secreting higher levels of pro-inflammatory cytokines, such as TNF-α and IL-1β, IL-2, IL-6, IL-12, and IL-23. However, with the development of immune selection, the proliferation of abnormal cells gradually loses their major histocompatibility complex class I (MHC-I) and major histocompatibility complex class II (MHC-II) antigens and decreases the amount of tumor antigens in the equilibrium phase. By this time, tumor-derived soluble factors switch M1 type macrophages to M2 type macrophages and shape a tumor-promoting TME, in which M2 type macrophages assist cancer cells in evading immune attack, thereby promoting tumor progression and metastasis
At present, various strategies have been adopted to develop macrophage-targeted therapies, including macrophage depletion (Cassetta and Pollard 2023), macrophage phagocytosis enhancement (Hu et al. 2023), and modifying macrophage recruitment (Bied et al. 2023). Though these strategies have been moved forward in several preclinical trial phases and partly showed promising preclinical results, the off-tumor toxicity and other side effects limit their applications in cancer immunotherapy. In recent years, a novel targeting macrophage strategy called “macrophage reprogramming” has represented a more viable option to address these issues. Macrophage reprogramming, also known as macrophage repolarization, is defined as a continuous dynamic phenotype switch between the pro-inflammatory M1 phenotype and the anti-inflammatory M2 phenotype. Macrophage reprogramming aims to modulate the activation dysregulation of macrophages and get controllable phenotypic alterations in diseases. The destination of macrophage reprogramming is the reconstruction of a strong and well-balanced immune environment to eradicate diseases. Importantly, this strategy modulates the macrophages themselves to maintain the healthy homeostasis of tissues in diseases.
The heterogeneity and plasticity of macrophages increase the difficulty of targeting macrophage therapies. One difficulty lies in accurately tracking and evaluating the effects of targeted therapies. Several conventional biochemical techniques mainly focus on a certain moment in macrophage-targeted therapies, often overlooking the global dynamic structural and functional changes of polarized macrophages. Molecular imaging supplements this missing spatiotemporal dynamic information by providing long-term tracking, high sensitivity and specificity imaging (Hurt et al. 2023). Moreover, molecular imaging provides a variety of powerful non-invasive tools for dynamically monitoring biological processes, including optical imaging (Pahlevaninezhad et al. 2022), photothermal imaging (Fu et al. 2023), photoacoustic imaging (Lin and Wang 2022), raman imaging (Shi et al. 2022), and so on. Based on a series of advanced and highly sensitive fluorescent dyes or proteins, imaging techniques have been applied in cancer imaging, neuroscience detection, metabolism measurement, response monitoring of clinical treatment, and other preclinical and clinical applications (Rowe and Pomper 2022). Nowadays, molecular imaging has been widely used in real-time monitoring of polarized macrophages during various biological events.
In this review, we comprehensively summarize recent advances in the field of macrophage reprogramming, including distinct roadmaps, approaches, and applications from preclinical experiments to clinical trials. Moreover, we discuss various molecular imaging approaches that can be used to monitor these biological processes in real time. Overall, this review provides an extensive overview of modulating and imaging macrophage reprogramming for cancer immunotherapy.
The Main Roadmaps of Macrophage Reprogramming
Generally, the imbalanced phenotypes of macrophages frequently occur in various diseases and can further worsen the progression of these diseases through molecular cascades and the immune network (Mass et al. 2023). To address these excessively polarized macrophages, macrophage reprogramming has emerged as a superior strategy for correcting inappropriate deviations. At present, this developing reprogramming strategy aims to obtain controllable promotion of the desired pro-inflammatory M1 or anti-inflammatory M2 phenotypes in specific diseases (Ma et al. 2023). The core roadmaps of macrophage reprogramming include: (1) targeting and modulating genomic DNA and epigenetics; (2) targeting cell surface and intracellular molecules to activate or block them; and (3) regenerating desired cellular microenvironments with specific physicochemical properties.
Targeting Genomic DNA and Epigenetics
The activation and polarization of macrophages involve a complex interplay of signaling and effector molecules. The gene regulatory network of polarized macrophages plays a central role in these biological processes, which includes transcriptional, post-transcriptional regulation, translation, post-translational modifications, and correlative signal transduction of activation and polarization-associated genes (Revel et al. 2022). Epigenetic modifications are increasingly recognized as an important mechanism for regulating gene expression. In the TME, cancer cells extend their survival advantages through epigenetic modifications (Sun et al. 2022). Correspondingly, immune cells also take advantage of this mechanism to boost antitumor immune responses. Considering the strong heterogeneity of macrophages, more and more research has been directed toward exploring epigenetic pathways for macrophage reprogramming, such as DNA methylation, histone modifications, and noncoding RNAs.
Targeting Cell Surface and Intracellular Molecules
The heterogeneity and plasticity of macrophages largely depend on cell surface molecules and cell signaling, which include cell surface receptors, ligand-receptor pairs, and signaling pathway modulators (Zhang et al. 2023). Considering the complexity, dynamics, and universality of macrophages throughout the body, targeting specific cell populations with appropriate cell surface molecules, especially clinically relevant surface receptors, will enable the development of more precise reprogramming strategies to treat diseases without off-target-related adverse effects. The major cell surface receptors involved in the pro-inflammatory M1 phenotype include CD16, CD32, CD64, CD68, CD80, CD86, CD369, MHC-II, and so on (Cheng et al. 2022). M2 macrophages are characterized by up-regulation of several surface molecules, such as the mannose receptor (Fig. 1). Targeting these receptors and their ligands have advanced to preclinical trials and shown promising effects.
Regenerating Cellular Microenvironment
More and more evidence proves that the discriminative activation and polarization of macrophages largely depend on the different physiological and pathological microenvironments, especially in the TME (Christofides et al. 2022). The microenvironment of macrophages consists of extracellular matrix, distinct cellular populations in terms of phenotype and spatial distribution, soluble proteins such as growth factors, and physicochemical signal molecules. These factors influence cellular phenotypes and functions through various physical, mechanical, and biochemical mechanisms like pH, oxygen, and shear stress (Zhao et al. 2022). Additionally, macrophages secrete a series of cytokines to influence the microenvironment, and ultimately stimulate cell proliferation. Therefore, manipulating the cellular microenvironment will be more conducive to macrophage activation and polarization.
The Therapeutic Approaches of Macrophage Reprogramming
As stated in the previous paragraph, it summarizes the main directions of macrophage reprogramming. Based on these promising roadmaps, some therapeutic approaches have been developed to reprogram macrophages, which include monoclonal antibody drugs, small molecule drugs, gene therapy, and chimeric antigen receptor engineered macrophages (CAR-M). Some of these methods have been adopted in preclinical trials and achieved certain promising outcomes.
Monoclonal Antibody Drugs
Since the approval of the first monoclonal antibody drug Orthoclone OKT3 (muromonab-CD3) by the Food and Drug Administration (FDA) in 1986, over 100 antibody drugs have been approved for disease treatment (Attwood et al. 2020). These advancements have greatly accelerated the development of monoclonal antibody drugs for modifying macrophage reprogramming. In most common cancers, the presence of anti-inflammatory M2 phenotype tumor-associated macrophages (TAMs) always leads to the failure of conventional cancer therapies (Zhang et al. 2021b). Therefore, a series of monoclonal antibody drugs with high specificity have been developed to target and reprogram TAMs from the anti-inflammatory M2 phenotype to the pro-inflammatory M1 phenotype in cancer immunotherapies.
The general approach to developing monoclonal antibody drugs involves targeting the cell surface signaling molecules on macrophages. Liu et al. (2023) reported that CD40 monoclonal antibodies promoted M1 polarization through the modulation of glutamine metabolism and fatty acid oxidation. Additionally, the key mechanism of therapeutic antibodies is antibody-dependent cellular phagocytosis (ADCP) (Kamber et al. 2021). Cao et al. (2022) found that paclitaxel efficiently reprogrammed macrophages into the M1 phenotype, and this reprogramming enhanced ADCP when combined with cetuximab and rituximab. Interestingly, this phenomenon differed from the efferocytosis of apoptotic cancer cells induced by M2 macrophages. These findings underscore the significance of macrophage reprogramming in determining the long-term efficacy of anticancer monoclonal antibodies.
Small Molecule Drugs
Small molecule drugs, characterized by low molecular weights and simple structures, possess the capability to effectively permeate cells, which exerting a wide range of biological effects (Childs-Disney et al. 2022). The common applications of small molecule drugs are developed to target signaling molecules. Among these, colony-stimulating factor 1 (CSF1) has been recognized as one of the most important molecule targets in macrophage reprogramming. The CSF1/CSF1R signaling inhibitor pexidartinib (PLX3397) has been approved by the FDA to reprogram TAMs in tenosynovial giant cell tumors. Fujiwara et al. (2021) found PLX3397 significantly exhibited versatile immune modulating effects to reprogram TAMs by suppressing the phosphorylation of extracellular signal-regulated protein kinases 1/2 (pERK1/2) activation and the polarization of M2 TAMs. Moreover, other CSF1/CSF1R signaling inhibitors, such as BLZ945, PLX7486, ARRY382, DCC3014, and JNJ40346527 (Edicotinib), have also achieved significant progress in various cancer treatments (Kosti et al. 2022).
In addition, internal signaling molecules in macrophages are also potential targets for reprogramming. For example, vorinostat, a small molecule drug, was developed to inhibit histone deacetylases to switch macrophages from an M2 phenotype to an M1 phenotype (Li et al. 2021b). Similarly, TG100-115, a PI3Kγ/δ pathway inhibitor, has been identified to suppress tumor growth and metastasis by reconstructing TME and reprogramming macrophages (Sullivan et al. 2018). Based on similar design principles, a series of internal signaling pathways, such as the nuclear factor kappa B (NF-κB) pathway, the mitogen-activated protein kinase (MAPK) signalling pathway, the janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway, the adenosine monophosphate-activated kinase (AMPK)-peroxisome proliferatoractivated receptor (PPAR)-dependent pathway, and the nuclear factor erythroid-2 related factor 2 (Nrf2)/heme oxygenase (HO-1) signaling pathway, have been targeted for the design of small molecule drugs.
Gene Therapy
With the development of genetics and molecular biology, gene therapy has become an attractive therapeutic approach from bench to bedside. Nowadays, there are several developing gene therapy methods to reprogram macrophages, including delivery of therapeutic nucleic acids (TNAs), direct gene editing, and epigenetic regulation (Gupta et al. 2021).
The TNAs have been explored for their role in regulating ILs that are associated with polarized macrophages. Li et al. (2022) designed an engineered exosome that encapsulated IL-10 pDNA to reprogram macrophages. This approach aimed to reduce the levels of pro-inflammatory cytokines (such as IL-1β and TNF-α) and increase the expression of IL-10 cytokines. In addition to pDNA, miRNA and mRNA are also widely adopted for inducing reprogramming in macrophages (Song et al. 2022; Zhang et al. 2019). Furthermore, the advent of gene-editing technology has accelerated the development of human gene therapy (Zou et al. 2022). For example, Lin et al. (2022) developed a gene delivery system to reprogram macrophages through clustered regularly interspaced short palindromic repeats (CRISPR)-mediated CD47 blockade and IL-12 overexpression in situ. This combination gene therapy successfully remolded the TME, promoting the production of IL-12 and synergistically reprogramming TAMs to the M1 phenotype, thereby enhancing the macrophage-mediated anti-tumor immunotherapy effects.
Some researchers also pay attention to regulating the epigenetics of macrophages. Lauterbach et al. (2019) found that targeting the ATP-citrate lyase, which was associated with histone acetylation, could treat certain cancers by reprogramming macrophages. Moreover, Zhang et al. (2021a) discovered that glycoprotein A repetitions predominant and DNA methylation-mediated mechanisms metabolically reprogrammed TAMs in pancreatic cancer cells. Furthermore, Qian et al. (2017) developed the M2-like TAM dual-targeting nanoparticles (M2NPs) loaded with CSF-1R small interfering RNA and scavenger receptor B type 1 peptides to eliminate M2-like TAMs. As expected, this strategy specifically targeted M2-like TAMs and elicited more powerful anti-cancer immune responses compared to other tissue-resident macrophages in the liver, spleen, and lung.
CAR-M
Chimeric antigen receptor-engineered T cells (CAR-T) therapy has been developed to treat hematological malignancy and has achieved revolutionary clinical responses (Lu and Jiang 2022). The success of CAR-T has accelerated the development of similar immune cell therapies, such as chimeric antigen receptor-engineered natural killer cells (CAR-NK), chimeric antigen receptor-engineered natural killer T cells (CAR-NKT), CAR-M, CAR-Treg, and CAR-γδT (Lin et al. 2021). Considering the extensive distribution of macrophages in most solid tumors, CAR-M, as a unique therapeutic approach, has achieved promising preclinical and clinical effects.
Klichinsky et al. (2020) developed anti-human epidermal growth factor receptor 2 (HER2) CAR-M (CT-0508), which genetically manipulated human macrophages by CAR technology to acquire constant pro-inflammatory (M1) phenotype macrophages against tumors. This work has achieved encouraging progress in first-in-human phase 1 clinical trials (NCT04660929) and acquired the fast-track designation of the FDA in 2021 (Mukhopadhyay 2020). Based on nanobiotechnology, Kang et al. (2021) designed polymer nanocarriers delivering plasmids encoding CAR and interferon-γ to manipulate macrophages in vivo. This delicate design effectively reprogrammed the TAMs from the M2 phenotype to the M1 phenotype and successfully accomplished the CAR-M in situ. Meanwhile, the CAR-M remodeled the TME and suppressed tumor growth by augmenting the immune functions of M1 TAMs and tumor-specific cytotoxic T lymphocytes. Moreover, Pierini et al. (2022) developed a combination therapy that mixed CAR-M and PD-1/PD-L1 T cell checkpoint axis blockade. This combination therapy obtained much better results in reprogramming macrophages to enhance the anti-tumor effects.
The Molecular Imaging of Macrophage Reprogramming
It is difficult for traditional biochemical methods to obtain accurate and long-term research data that contain spatiotemporal dynamics information in health and disease states, especially in the occurrence and development of tumors. With the continuous development of molecular imaging techniques, represented by intravital imaging (IVI) (Entenberg et al. 2023), imaging macrophage reprogramming provides a more intuitive and visual method to explore the regulation of macrophage activation and polarization in situ.
It is crucial for imaging macrophage reprogramming to characterize different activation states of macrophages in real-time. Surface markers related to macrophages and some metabolic molecules are commonly adopted as markers for distinguishing polarized macrophages (Jiang et al. 2022; Lee and Bensinger 2022). However, few studies have reported the application of visualizing gene expression to differentiate macrophage polarization in vivo. The weak expression of these proteins makes it difficult to distinguish the real molecule signals from the background. Some signal amplification strategies have been employed to improve the signal intensity, such as antibody labeling. Ramos et al. (2022) identified the folate receptor beta positive (FOLR2+) tissue-resident macrophages in healthy tissue and cancers. Furthermore, they used confocal living imaging to image their cytokinetics connections with CD8+ T cells by labeling fluorescently coupled antibodies. Zhang et al. (2017) developed Dylight755-N-hydroxysuccinimide-conjugated anti-CD206 monoclonal antibodies and 85 MBq Na125I-conjugated anti-CD206 monoclonal antibodies, which were used for imaging M2 macrophages in vivo through near-infrared fluorescence imaging or single photon emission computed tomography.
Molecular Imaging of Macrophage Metabolism
An alternative strategy for monitoring macrophage reprogramming is metabolic imaging, which involves detecting various metabolic products, such as the activity of cellular enzymes (Lung et al. 2022). During macrophage reprogramming, nitric oxide (NO) produced by inducible nitric oxide synthase has been widely used as a signature molecule for M1 polarization (Fig. 3). A series of exogenous synthetic or endogenous gene-coding probes have been developed to characterize polarized macrophages by detecting NO. For exogenous probes, Zhao et al. (2021) developed a FRET-NO probe and Hoe-Rh-NO for endogenous NO imaging, and Liang et al. (2022) developed a two-photon ratiometric fluorescent probe for NO imaging. Li et al. (2020a) developed a highly sensitive NO probe for imaging inflammation reactions in the stroke process, and Zhou et al. (2020) developed a smart NO probe, PYSNO, for NO imaging in myocardial fibrosis tissue. Currently, fluorescence probes with endogenous NO gene encoding have lower detective sensitivity compared to exogenous NO probes. However, their non-invasive nature enables them to more accurately reflect the real-time status of macrophage reprogramming.
Fig. 3.
The representative methods of targeted imaging polarized macrophages. a Representative structure of exogenous and endogenous probes for the detection of nitric oxide (NO) and reactive oxygen species (ROS) as well as matrix metalloproteinase (MMP) activity associated with macrophage activity. The different fluorescent probes are classified according to their targets: NO, ROS, and MMP. Reproduced with permission from reference (Dickinson et al. 2010). b Fluorescent probes for imaging the phagocytosis of macrophages. The probes are named according to their targets: phagosome pH: PhagoGreen, CD11b: CDnir7, Slc18b1: CDg16, GLUT1: CDr17. c Imaging of the mitochondrial organization is correlated with the activation status of macrophages. There are some representative structured illumination microscopy images of mitochondria (red color, MitoTracker Red CMXRos) in bone marrow-derived macrophages (BMDMs) (scale bar in upper image, 10 µm; scale bar in lower image, 2 µm). Reproduced with permission from reference (Li et al. 2020b)
Despite releasing NO, activated macrophages also release reactive oxygen species (ROS) and matrix metalloproteinases (MMPs) (Fig. 3). Deguchi et al. (2006) utilized an activated near-infrared fluorescent probe with high sensitivity and high-resolution to detect gelatinase enzyme activity (MMP-2 and MMP-9) in polarized macrophages in vivo. In addition to detecting protein expression, the expression of intracellular RNA also exhibits significant differences between M1 and M2 activation. Yang et al. (2019) have developed a new method that is suitable for real-time detection of RNA in living cells. In the future, RNA-based molecular probes might also be gradually applied to distinguish macrophages with different activation states.
Molecular Imaging of Macrophage Morphology
It has been reported that macrophages, due to their strong heterogeneity and plasticity, always exhibit irregular morphological changes associated with various polarization states (Ackermann et al. 2022). In recent years, the development of novel technologies, represented by machine learning and deep learning techniques, has greatly improved the ability to distinguish macrophage polarization states through imaging their morphology. For example, Kröger et al. (2022) developed a two-photon excited lifetime imaging method that efficiently distinguished polarized macrophages from dermal cells in vivo using specific machine learning algorithms. Li et al. (2020b) analyzed the morphology of mitochondrial organization in bone marrow-derived macrophages (BMDMs) and polarized macrophages by extracting characteristic parameters, such as mean length of branches, mean network size (the mean number of branches per network), and mitochondrial footprints (mitochondrial coverage area). These parameters were successfully used to recognize and distinguish macrophages with different activation states in vivo. Pavillon et al. (2018) obtained morphological parameters of macrophages through non-invasive and labeling-free methods such as quantitative phase microscopy, spontaneous fluorescence imaging, and molecular indicators from Raman spectroscopy. They then constructed a multivariate statistical model based on logistic regression to visualize macrophage reprogramming.
Molecular Imaging of Macrophage Phagocytosis
In addition to the irregular cell morphology of polarized macrophages, recent research has reported that various polarized macrophages have significant differences in the phagocytosis of fluorescent compounds. Lin et al. (2019) developed a novel nanoparticle named “Nanopomegranate” with fast self-assembly, dye aggregates, and dual-modality “off/on” capability. In particular, this nanopomegranate showed better photostability, extinction coefficient, and labeling efficiency (98.8%) for visualizing kupffer cells (KCs) in the liver compared to commercial FluoSpheres. Further, Deng et al. (2021) designed an innovative drawer-type abdominal window and combined it with nanopomegranate to achieve more than 10 days of IVI in KCs of the liver using intravital fluorescence and photoacoustic imaging. Kang et al. (2014) developed a near-infrared probe named CDnir7 for visualizing polarized macrophages in vivo using multiple imaging methods, such as in vivo imaging system spectrum, fluorescence molecular tomography, and multi-spectral optoacoustic tomography. Vazquez-Romero et al. (2013) synthesized a series of BODIPY derivatives with high fluorescence quantum yield and excellent cell permeability using a multi-component reaction method. Among them, a fluorescent probe called PhagoGreen, characterized by high sensitivity to low pH, selectively imaged the phagocytosis of reprogramming macrophages through responding to the acidification of macrophage phagosomes. To image activated macrophages, Park et al. (2019) screened CDg16 from more than 8000 compounds and further identified Slc18b1/SLC18B1 molecules that interacted with CDg16, regulating the phagocytosis of activated macrophages. Although the CDg16 probe provided a valuable tool for IVI of macrophage activation, it could not distinguish the specific polarized phenotype of macrophages. Based on the CDg16 probe, Cho et al. (2022) constructed a luminescent-carbohydrate library and screened the CDr17 probe, which distinguished M1 polarized macrophages through glucose transporter 1 translocation. These fluorescent probes, which are stable in cells and independent of molecular binding, mainly rely on the Gating Oriented-Live Cell Distinction of ion channels of cell-surface membranes and provide a novel strategy for IVI of reprogramming macrophages (Choi et al. 2019).
The Therapeutic Applications of Macrophage Reprogramming
Hepatic Carcinoma (HCC)
The microenvironment of HCC is abundantly infiltrated by various macrophage populations, such as resident macrophages, KCs, and monocyte-derived macrophages (Ohshiro et al. 2023). The activation and polarization states of these various macrophage populations directly affect the development of HCC, especially the anti-inflammatory M2 phenotype TAMs. Targeting and reprogramming liver macrophages, converting them from the M2 to M1 phenotype has been regarded as a valuable approach in liver cancer immunotherapy.
Li et al. (2017) tested a novel CC chemokine receptor 2 (CCR2) antagonist in murine hepatocellular carcinoma cells and found that the blockade or knockout of CCR2 significantly reconstructed TME by reprogramming M2 TAMs and activating the CD8+ cytotoxic T cells. Moreover, Wang et al. (2022) designed a nanodrug to reprogram TAMs with an NF-κB inhibitor and PD-L1 antibody in liver cancer. Interestingly, some traditional drugs are now being used to reprogram macrophages in HCC. De Oliveira et al. (2019) used IVI to visualize the disease progression of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis-HCC in zebrafish models. Their data showed that metformin specifically reprogrammed macrophages and increased T-cell density to restore tumor surveillance. Nowadays, a variety of novel preclinical and clinical methods have been developed to reprogram macrophages in the treatment of HCC, which pay more attention to interdisciplinary approaches and new technologies. These studies further highlight the important role of reprogramming macrophages in HCC.
Lung Carcinoma
Lung carcinoma accounts for almost 25% of all cancer deaths in 2022 (Giaquinto et al. 2022). Thorsson et al. (2019) performed immunogenomic analysis from The Cancer Genome Atlas, which included 33 diverse cancer types and found that macrophage phenotypes, especially M2 TAMs, were closely correlated with poor prognosis in lung cancer patients. Therefore, targeting and reprogramming macrophages may provide a new powerful therapy for treating lung cancer.
Zhou et al. (2022) performed a high-throughput screening and found that carfilzomib, a selective proteasome inhibitor, reprogrammed the M2 macrophages into the M1 phenotype by inducing endoplasmic reticulum stress and activating NF-κB. In addition to conventional anticancer drugs, some novel nanodrugs have been developed for lung cancer treatment in preclinical trials. Su et al. (2022) designed Au@PG nanoparticles to reprogram macrophages by transforming anti-inflammatory M2 macrophages into pro-inflammatory M1 macrophages in lung cancer models. As mentioned above, monoclonal antibodies serve as potential therapeutic agents. Yu et al. (2022) developed an anti-Chi3L1-humanized antibody and evaluated its antitumor and antimetastatic effects. They found that this monoclonal antibody inhibited M2 TAMs through signal transducer and activator of transcription 6, while having no effect on M1-polarized macrophages in lung cancer. La Fleur et al. (2021) identified tumor-derived IL-37 and macrophage receptors with collagenous structure (MARCO) as potential therapeutic targets in lung cancer. The blocking antibodies to IL-37 and MARCO remodeled the TME and reprogrammed the TAMs to kill lung cancer cells. In conclusion, these various studies prove that reprogramming macrophages is beneficial for treating lung cancer patients.
Breast Carcinoma
Breast carcinoma almost accounts for 12.5% of all new annual cancer cases globally (Dolatkhah et al. 2020). Nearly 30% of patients develop recurrent disease due to therapy resistance, which eventually metastasizes in distant organs and becomes the main cause of death (Pan et al. 2017). Moreover, a large number of studies, especially the widely applied scRNA-seq, have identified the versatile role of macrophages connected with the progression to malignancy of breast cancer. These macrophages mediate tumor cell migration, invasion, and intravasation by suppressing anti-tumor immunity (Zhang et al. 2021b). Therefore, developing novel immunotherapy to target and reprogram macrophages may effectively inhibit resistance, recurrence, and metastasis of breast cancer, thus improving treatment outcomes and the results of combination therapies.
At present, the prerequisite for targeting and reprogramming macrophages is to accurately identify the function of macrophages in breast cancer. Wang et al. (2023) developed a visible mesoporous silica nanoparticle with apolipoprotein A-1 (ApoA-1) mimetic peptide R4F and indocyanine green. This multifunctional nanoparticle was not only efficiently taken up to label 4T1 cells and macrophages but also decreased M2-like TAMs and killed cancer cells by photoacoustic imaging and photothermal therapy. Nishida-Aoki and Gujral (2022) developed an in vitro TAM polarization system and screened phenotypic kinase inhibitors that significantly decreased the populations of M2 TAMs in breast cancer. Additionally, using epigenetic regulation is also common in reprogramming TAMs for treating breast cancer. Ma et al. (2022a) proved that knockout of miR-182 suppressed the growth of breast cancer by reducing M2 TAM populations, which involved suppressing toll-like receptor (TLR)-4, and inactivated NF-κB pathways. Furthermore, antagomiR-182 effectively elicited macrophage reprogramming and tumor suppression in breast cancer. Mehta et al. (2021) demonstrated that macrophage populations were the most abundant immune cell type in breast cancer through scRNA-seq. Then, combined with multi-omics profiling, they found that poly (ADP-ribose) polymerase inhibitors indistinguishably reprogrammed both macrophage populations by metabolic pathways. Additionally, they evaluated the therapeutic effects of combination therapy with poly (ADP-ribose) polymerase inhibitors and CSF1R-blocking antibodies. The results showed that the combination therapy induced a durable reprogramming of TME, providing a promising therapeutic strategy for breast cancer. Overall, targeting and reprogramming macrophages in breast cancer is a novel therapeutic method with great potential for clinical treatment.
Conclusions and Perspectives
The development of cancer involves multistep biological processes, including proliferation, survival, invasion, and metastasis. Immunotherapy is committed to developing specific cancer therapies for each of these processes. As the most abundant immune cell population in TME, macrophages are the most potential treatment targets for cancers. Among various targeting macrophage immunotherapies, macrophage reprogramming has been considered the most promising next-generation immunotherapy due to its precision and low side effects. In this review, we summarize the current progress in macrophage reprogramming from conceptual roadmaps to therapeutic approaches and applications. Specifically, we focus on molecular imaging of macrophage reprogramming. Molecular imaging allows us to directly observe various molecular events during the macrophage reprogramming process in cancer immunotherapy (Table 1). In the future, integrating molecular imaging to develop new macrophage reprogramming methods will become a promising direction for precise clinical cancer immunotherapy (Fig. 4).
Table 1.
A schematic summary of modulating and imaging macrophage reprogramming
| Polaration | Targets | Cancers | Events | Methods | Imaging | Models | References |
|---|---|---|---|---|---|---|---|
| M1 | CSF-1R, SRB1 | Melanoma tumors | Dual-targeting reprogramming TAMs | Gene therapy | Confocal intravital microscopy | Mice | Qian et al. (2017) |
| Mesothelin, HER2 | Ovarian cancer | Phagocytic activity | CAR-M | Bioluminescence, imaging flow cytometry | Mice, Human | Klichinsky et al. (2020) | |
| CD20, EGFR, HER2 | Lymphoma, epidermoid carcinoma, breast adenocarcinoma | ADCP | Monoclonal antibody drugs | Confocal microscopy, IVIS | Mice, BMDM | Li et al. (2021a) | |
| PD-1,TLR7/8 | Colorectal cancer | High-throughput and low-cost phenotypic screening | Small molecule drugs | Fluorescence reflectance imaging, intravital imaging | Mice | Rodell et al. (2018) | |
| ASF1A, PD-1 | Lung cancer | In vivo epigenetic CRISPR screen | Gene therapy | MRI | Mice | Li et al. (2020b) | |
| M2 | CD206, NO | Breast cancer | Early metastasis | Small molecule drugs | The NIR-II FL and bioluminescence | Mice, RAW264.7 | Yuan et al. (2023) |
| SIRPα, CD47 | Breast cancer, melanoma | TAM repolarization | Gene therapy | Confocal imaging | Mice | Rao et al. (2020b) | |
| m6A modification |
Melanoma, lung cancer |
Epigenetic regulation | Gene therapy | Bioluminescence | Mice | Yin et al. (2021) | |
| CD24, Siglec-10 | Ovarian cancer, breast cancer | Anti-phagocytic | Monoclonal antibody drugs | Bioluminescence | Mice, Human | Barkal et al. (2019) | |
| MUC1, CD47 | Pancreatic cancer | Phagocytic activity | CAR-M | Bioluminescence, TEM, IVIS | Human, Mice, BMDMs, RAW 264.7 cells | Liu et al. (2023) |
CSF1R colony-stimulating factor 1 receptor, HER2 human epidermal growth factor receptor 2, CD20 cluster of differentiate 20, EGFR epidermal growth factor receptor, PD-1 programmed cell death protein 1, TLR7/8 toll-like receptor 7/8, ASF1A anti-silencing function 1A histone chaperone, CD206 the mannose receptor, NO nitric oxide, SIRPα signal regulatory protein α, CD47 cluster of differentiation 47, M6A N6-methyladenosine, CD24 heat stable antigen (HSA) receptor, Siglec-10 sialic acid binding Ig like lectin 10, MUC1 mucin 1, ADCP antibody-dependent cellular phagocytosis, IVIS in vivo imaging system, MRI magnetic resonance imaging, TEM transmission electron microscopy, BMDM bone marrow-derived macrophage
Fig. 4.

Targeted modulation and imaging of macrophage reprogramming
Modulating and imaging macrophage reprogramming provides an innovative solution to visually understand this specific process. However, there remains a long way to go for good clinical practice. Firstly, due to the great plasticity and heterogeneity of macrophages, existing macrophage-based cancer immunotherapies exhibit a lack of specificity, potentially disrupting the body's immune homeostasis and increasing the risk of treatment failure in cancer patients. Besides, the individual differences between tumors and patients limit the therapeutic effects of macrophage reprogramming as a universal method. To overcome this obstacle, identifying new single-specific target molecules in macrophages via advanced technologies such as scRNA-seq is a potential direction for precise macrophage targeting. Secondly, the safety of reprogramming macrophages by various methods should be thoroughly tracked and evaluated. This requires joint efforts by regulatory agencies and scientists to ensure that patients around the world have early access to safe and high-quality cancer immunotherapies. Finally, the in vivo efficiency of inducing macrophage reprogramming (such as gene-editing efficacy, expansion efficiency, and immune efficacy) remains a challenge, limiting their applications in preclinical and clinical trials. Emerging gene-editing techniques, such as optimized CRISPR-Cas9 technology, base editing methods, and prime editing, might offer new strategies for solving these problems.
On the other hand, there are still some challenges in molecular imaging technologies to image macrophage reprogramming, such as non-invasion, real-time, precise labeling, long-term tracking, and penetration depth. To address these questions, some strategies have been adopted, including: (1) designing novel multimodality probes that combine diverse imaging techniques, such as fluorescence imaging integrated with magnetic resonance imaging and positron emission tomography. (2) Developing advanced imaging instruments and contrast agents. (3) Cooperating with new computer image processing technologies, such as deep learning and machine learning. In the future, these enhanced macrophage reprogramming imaging methods will achieve more precise and individualized treatment of therapeutic drugs by dynamically visualizing and guiding in real-time.
To sum up, with the development of molecular imaging and molecular biology, imaging macrophage reprogramming will significantly enhance our understanding to this specific biological event. Based on these findings, more and more innovative probe sensors and cancer immunotherapies will be developed to detect and eliminate solid tumors in the future.
Acknowledgements
This work was supported by the State Key Program of National Natural Science Foundation of China (32330048), the National Natural Science Foundation of China (82371857), and the Hainan University Scientific Research Foundation (KJRC2023B09).
Abbreviations
- TME
Tumor microenvironment
- TAMs
Tumor-associated macrophages
- CAR-M
Chimeric antigen receptor-engineered macrophages
- CD
Cluster of differentiation
- IL
Interleukin
- TNF
Tumor necrosis factor
- TLR
Toll-like receptor
- MHC-I
Major histocompatibility complex class I
- MHC-II
Major histocompatibility complex class II
- ADCP
Antibody-dependent cellular phagocytosis
- CSF1
Colony-stimulating factor 1
- CAR-T
Chimeric antigen receptor-engineered T cells
- IVI
Intravital imaging
- NO
Nitric oxide
- ROS
Reactive oxygen species
- MMP
Matrix metalloproteinase
- KCs
Kupffer cells
- BMDMs
Bone marrow-derived macrophages
- NF-κB
Nuclear factor kappa B
- HCC
Hepatic carcinoma
Authors' Contributions
ZHZ and ZL provided the concept, designed the outline, and directed the writing of the paper. JLW performed the literature survey, drafted the manuscript, and finished the figures and tables. YFL, RZ, and ZZC participated in the literature collection. ZF and YLX offered crucial content revisions and language polishing. All authors contributed to the article and approved the submitted version and final version.
Data Availability
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
Declarations
Conflict of Interest
The authors have no relevant financial or non-financial interests to disclose.
Ethical Approval
Not applicable.
Consent to Participate
Not applicable.
Consent for Publication
Not applicable.
Contributor Information
Zheng Liu, Email: liu-zheng@hainanu.edu.cn.
Zhihong Zhang, Email: czyzzh@mail.hust.edu.cn.
References
- Ackermann M, Rafiei Hashtchin A, Manstein F et al (2022) Continuous human iPSC-macrophage mass production by suspension culture in stirred tank bioreactors. Nat Protoc 17(2):513–539. 10.1038/s41596-021-00654-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Attwood MM, Jonsson J, Rask-Andersen M et al (2020) Soluble ligands as drug targets. Nat Rev Drug Discov 19(10):695–710. 10.1038/s41573-020-0078-4 [DOI] [PubMed] [Google Scholar]
- Bied M, Ho WW, Ginhoux F et al (2023) Roles of macrophages in tumor development: a spatiotemporal perspective. Cell Mol Immunol 20:983–992. 10.1038/s41423-023-01061-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao X, Chen J, Li B et al (2022) Promoting antibody-dependent cellular phagocytosis for effective macrophage-based cancer immunotherapy. Sci Adv 8(11):eabl9171. 10.1126/sciadv.abl9171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cassetta L, Pollard JW (2023) A timeline of tumour-associated macrophage biology. Nat Rev Cancer 23(4):238–257. 10.1038/s41568-022-00547-1 [DOI] [PubMed] [Google Scholar]
- Chen S, Saeed AF, Liu Q et al (2023) Macrophages in immunoregulation and therapeutics. Sig Transduct Target Ther 8(1):207. 10.1038/s41392-023-01452-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng K, Cai N, Zhu J et al (2022) Tumor-associated macrophages in liver cancer: from mechanisms to therapy. Cancer Commun 42(11):1112–1140. 10.1002/cac2.12345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Childs-Disney JL, Yang X, Gibaut QMR et al (2022) Targeting RNA structures with small molecules. Nat Rev Drug Discov 21(10):736–762. 10.1038/s41573-022-00521-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cho H, Kwon H-Y, Sharma A et al (2022) Visualizing inflammation with an M1 macrophage selective probe via GLUT1 as the gating target. Nat Commun 13(1):5974. 10.1038/s41467-022-33526-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi Y-K, Kim J-J, Chang Y-T (2019) Holding-oriented versus gating-oriented live-cell distinction: highlighting the role of transporters in cell imaging probe development. Acc Chem Res 52(11):3097–3107. 10.1021/acs.accounts.9b00253 [DOI] [PubMed] [Google Scholar]
- Christofides A, Strauss L, Yeo A et al (2022) The complex role of tumor-infiltrating macrophages. Nat Immunol 23(8):1148–1156. 10.1038/s41590-022-01267-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Oliveira S, Houseright RA, Graves AL et al (2019) Metformin modulates innate immune-mediated inflammation and early progression of NAFLD-associated hepatocellular carcinoma in zebrafish. J Hepatol 70(4):710–721. 10.1016/j.jhep.2018.11.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deguchi J-o, Aikawa M, Tung C-H et al (2006) Inflammation in atherosclerosis: visualizing matrix metalloproteinase action in macrophages in vivo. Circulation 114(1):55–62. 10.1161/CIRCULATIONAHA.106.619056 [DOI] [PubMed] [Google Scholar]
- Deng DQ, Dai BL, Wei JS et al (2021) A drawer-type abdominal window with an acrylic/resin coverslip enables long-term intravital fluorescence/photoacoustic imaging of the liver. Nanophotonics 10(12):3369–3381. 10.1515/nanoph-2021-0281 [Google Scholar]
- Dickinson BC, Huynh C, Chang CJ (2010) A palette of fluorescent probes with varying emission colors for imaging hydrogen peroxide signaling in living cells. J Am Chem Soc 132(16):5906–5915. 10.1021/ja1014103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding C, Shrestha R, Zhu X et al (2023) Inducing trained immunity in pro-metastatic macrophages to control tumor metastasis. Nat Immunol 24(2):239–254. 10.1038/s41590-022-01388-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dolatkhah R, Somi MH, Jafarabadi MA et al (2020) Breast cancer survival and incidence: 10 years cancer registry data in the Northwest, Iran. Int J Breast Cancer 2020:6. 10.1155/2020/1963814 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duan Z, Luo Y (2021) Targeting macrophages in cancer immunotherapy. Sig Transduct Target Ther 6(1):127. 10.1038/s41392-021-00506-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Entenberg D, Oktay MH, Condeelis JS (2023) Intravital imaging to study cancer progression and metastasis. Nat Rev Cancer 23(1):25–42. 10.1038/s41568-022-00527-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fu P, Cao W, Chen T et al (2023) Super-resolution imaging of non-fluorescent molecules by photothermal relaxation localization microscopy. Nat Photon 17(4):330–337. 10.1038/s41566-022-01143-3 [Google Scholar]
- Fujiwara T, Yakoub MA, Chandler A et al (2021) CSF1/CSF1R signaling inhibitor pexidartinib (PLX3397) reprograms tumor-associated macrophages and stimulates t-cell infiltration in the sarcoma microenvironment. Mol Cancer Ther 20(8):1388–1399. 10.1158/1535-7163.MCT-20-0591 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gharavi AT, Hanjani NA, Movahed E et al (2022) The role of macrophage subtypes and exosomes in immunomodulation. Cell Mol Biol Lett 27(1):83. 10.1186/s11658-022-00384-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giaquinto AN, Miller KD, Tossas KY et al (2022) Cancer statistics for African American/black people 2022. CA Cancer J Clin 72(3):202–229. 10.3322/caac.21718 [DOI] [PubMed] [Google Scholar]
- Gupta A, Andresen JL, Manan RS et al (2021) Nucleic acid delivery for therapeutic applications. Adv Drug Deliv Rev 178:113834. 10.1016/j.addr.2021.113834 [DOI] [PubMed] [Google Scholar]
- Hu Z, Li WQ, Chen SM et al (2023) Design of a novel chimeric peptide via dual blockade of CD47/SIRP alpha and PD-1/PD-L1 for cancer immunotherapy. Sci China Life Sci 66:2310–2328. 10.1007/s11427-022-2285-6 [DOI] [PubMed] [Google Scholar]
- Hurt RC, Buss MT, Duan M et al (2023) Genomically mined acoustic reporter genes for real-time in vivo monitoring of tumors and tumor-homing bacteria. Nat Biotechnol 41:919–931. 10.1038/s41587-022-01581-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang S, Chan CN, Rovira-Clavé X et al (2022) Combined protein and nucleic acid imaging reveals virus-dependent B cell and macrophage immunosuppression of tissue microenvironments. Immunity 55(6):1118–1134. 10.1016/j.immuni.2022.03.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamber RA, Nishiga Y, Morton B et al (2021) Inter-cellular CRISPR screens reveal regulators of cancer cell phagocytosis. Nature 597(7877):549–554. 10.1038/s41586-021-03879-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang N-Y, Park S-J, Ang XWE et al (2014) A macrophage uptaking near-infrared chemical probe CDnir7 for in vivo imaging of inflammation. Chem Commun 50(50):6589–6591. 10.1039/c4cc02038c [DOI] [PubMed] [Google Scholar]
- Kang M, Lee SH, Kwon M et al (2021) Nanocomplex-mediated in vivo programming to chimeric antigen receptor-M1 macrophages for cancer therapy. Adv Mater 33(43):2103258. 10.1002/adma.202103258 [DOI] [PubMed] [Google Scholar]
- Klichinsky M, Ruella M, Shestova O et al (2020) Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol 38(8):947–953. 10.1038/s41587-020-0462-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kosti CN, Vaitsi PC, Pappas AG et al (2022) CSF1/CSF1R signaling mediates malignant pleural effusion formation. JCI Insight 7(6):e155300. 10.1172/jci.insight.155300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kröger M, Scheffel J, Shirshin EA et al (2022) Label-free imaging of M1 and M2 macrophage phenotypes in the human dermis in vivo using two-photon excited FLIM. Elife 11:e72819. 10.7554/eLife.72819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- La Fleur L, Botling J, He F et al (2021) Targeting MARCO and IL37R on immunosuppressive macrophages in lung cancer blocks regulatory T cells and supports cytotoxic lymphocyte function. Cancer Res 81(4):956–967. 10.1158/0008-5472.CAN-20-1885 [DOI] [PubMed] [Google Scholar]
- Lauterbach MA, Hanke JE, Serefidou M et al (2019) Toll-like receptor signaling rewires macrophage metabolism and promotes histone acetylation via ATP-citrate lyase. Immunity 51(6):997–1011. 10.1016/j.immuni.2019.11.009 [DOI] [PubMed] [Google Scholar]
- Lee M-S, Bensinger SJ (2022) Reprogramming cholesterol metabolism in macrophages and its role in host defense against cholesterol-dependent cytolysins. Cell Mol Immunol 19(3):327–336. 10.1038/s41423-021-00827-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lerner EC, Woroniecka KI, D’Anniballe VM et al (2023) CD8+ T cells maintain killing of MHC-I-negative tumor cells through the NKG2D-NKG2DL axis. Nat Cancer 4:1258–1127. 10.1038/s43018-023-00600-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Yao W, Yuan Y et al (2017) Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut 66(1):157–167. 10.1136/gutjnl-2015-310514 [DOI] [PubMed] [Google Scholar]
- Li C, Hu W, Wang J et al (2020a) A highly specific probe for the imaging of inflammation-induced endogenous nitric oxide produced during the stroke process. Analyst 145(18):6125–6129. 10.1039/D0AN00824A [DOI] [PubMed] [Google Scholar]
- Li Y, He Y, Miao K et al (2020b) Imaging of macrophage mitochondria dynamics in vivo reveals cellular activation phenotype for diagnosis. Theranostics 10(7):2897. 10.7150/thno.40495 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Dai H, Wang H et al (2021a) Exploring innate immunity in cancer immunotherapy: opportunities and challenges. Cell Mol Immunol 18(6):1607–1609. 10.1038/s41423-021-00679-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Su X, Liu R et al (2021b) HDAC inhibition potentiates anti-tumor activity of macrophages and enhances anti-PD-L1-mediated tumor suppression. Oncogene 40(10):1836–1850. 10.1038/s41388-020-01636-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H, Feng Y, Zheng X et al (2022) M2-type exosomes nanoparticles for rheumatoid arthritis therapy via macrophage re-polarization. J Control Release 341:16–30. 10.1016/j.jconrel.2021.11.019 [DOI] [PubMed] [Google Scholar]
- Liang M, Liu Z, Zhang Z et al (2022) A two-photon ratiometric fluorescent probe for real-time imaging and quantification of NO in neural stem cells during activation regulation. Chem Sci 13(15):4303–4312. 10.1039/D2SC00326K [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin L, Wang LV (2022) The emerging role of photoacoustic imaging in clinical oncology. Nat Rev Clin Oncol 19(6):365–384. 10.1038/s41571-022-00615-3 [DOI] [PubMed] [Google Scholar]
- Lin Q, Deng D, Song X et al (2019) Self-assembled “Off/On” nanopomegranate for in vivo photoacoustic and fluorescence imaging: strategic arrangement of Kupffer cells in mouse hepatic lobules. ACS Nano 13(2):1526–1537. 10.1021/acsnano.8b07283 [DOI] [PubMed] [Google Scholar]
- Lin H, Cheng J, Mu W et al (2021) Advances in universal CAR-T cell therapy. Front Immunol 12:744823. 10.3389/fimmu.2021.744823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin M, Yang Z, Yang Y et al (2022) CRISPR-based in situ engineering tumor cells to reprogram macrophages for effective cancer immunotherapy. Nano Today 42:101359. 10.1016/j.nantod.2021.101359 [Google Scholar]
- Liu P-S, Chen Y-T, Li X et al (2023) CD40 signal rewires fatty acid and glutamine metabolism for stimulating macrophage anti-tumorigenic functions. Nat Immunol 24(3):452–462. 10.1038/s41590-023-01430-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loke P, Lin JD (2022) Redefining inflammatory macrophage phenotypes across stages and tissues by single-cell transcriptomics. Sci Immunol 7(70):eabo4652. 10.1126/sciimmunol.abo4652 [DOI] [PubMed] [Google Scholar]
- Lu J, Jiang G (2022) The journey of CAR-T therapy in hematological malignancies. Mol Cancer 21(1):1–15. 10.1186/s12943-022-01663-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lung TWF, Charytonowicz D, Beaumont KG et al (2022) Klebsiella pneumoniae induces host metabolic stress that promotes tolerance to pulmonary infection. Cell Metab 34(5):761-774.e769. 10.1016/j.cmet.2022.03.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma C, He D, Tian P et al (2022a) miR-182 targeting reprograms tumor-associated macrophages and limits breast cancer progression. Proc Natl Acad Sci USA 119(6):e2114006119. 10.1073/pnas.2114006119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma RY, Black A, Qian BZ (2022b) Macrophage diversity in cancer revisited in the era of single-cell omics. Trends Immunol 43(7):546–563. 10.1016/j.it.2022.04.008 [DOI] [PubMed] [Google Scholar]
- Ma S, Sun B, Duan S et al (2023) YTHDF2 orchestrates tumor-associated macrophage reprogramming and controls antitumor immunity through CD8+ T cells. Nat Immunol 24(2):255–266. 10.1038/s41590-022-01398-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mass E, Nimmerjahn F, Kierdorf K et al (2023) Tissue-specific macrophages: how they develop and choreograph tissue biology. Nat Rev Immunol. 10.1038/s41577-023-00848-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mehta AK, Cheney EM, Hartl CA et al (2021) Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer. Nat Cancer 2(1):66–82. 10.1038/s43018-020-00148-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mukhopadhyay M (2020) Macrophages enter CAR immunotherapy. Nat Methods 17(6):561–561. 10.1038/s41592-020-0862-4 [DOI] [PubMed] [Google Scholar]
- Nishida-Aoki N, Gujral TS (2022) Polypharmacologic reprogramming of tumor-associated macrophages toward an inflammatory phenotype. Cancer Res 82(3):433–446. 10.1158/0008-5472.CAN-21-1428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noonepalle SKR, Hernandez MG, Delgado CZ et al (2023) Reprogramming macrophages with HDAC6 inhibitors for anti-cancer macrophage-based cell therapy. Cancer Res 83(7):900–900. 10.1158/1538-7445.AM2023-900 [Google Scholar]
- Ohshiro K, Bhowmick K, Yang X et al (2023) PKM2 Modulates hepatic macrophage regulation of NASH, ferroptosis and HCC through TGF-β signaling. Cancer Res 83(7):4854–4854. 10.1158/1538-7445.AM2023-4854 [Google Scholar]
- Pahlevaninezhad M, Huang Y-W, Pahlevani M et al (2022) Metasurface-based bijective illumination collection imaging provides high-resolution tomography in three dimensions. Nat Photon 16(3):203–211. 10.1038/s41566-022-00956-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan H, Gray R, Braybrooke J et al (2017) 20-year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years. N Engl J Med 377(19):1836–1846. 10.1056/NEJMoa1701830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park S-J, Kim B, Choi S et al (2019) Imaging inflammation using an activated macrophage probe with Slc18b1 as the activation-selective gating target. Nat Commun 10(1):1111. 10.1038/s41467-019-08990-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pavillon N, Hobro AJ, Akira S et al (2018) Noninvasive detection of macrophage activation with single-cell resolution through machine learning. Proc Natl Acad Sci USA 115(12):E2676–E2685. 10.1073/pnas.1711872115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pierini S, Gabbasov R, Worth A et al (2022) Chimeric antigen receptor macrophages (CAR-M) sensitize solid tumors to anti-PD1 immunotherapy. Cancer Res 82(12):2112–2112. 10.1158/1538-7445.AM2022-2112 [Google Scholar]
- Qian Y, Qiao S, Dai YF et al (2017) Molecular-targeted immunotherapeutic strategy for melanoma via dual-targeting nanoparticles delivering small interfering RNA to tumor-associated macrophages. ACS Nano 11(9):9536–9549. 10.1021/acsnano.7b05465 [DOI] [PubMed] [Google Scholar]
- Qian Y, Galan-Cobo A, Guijarro I et al (2023) MCT4-dependent lactate secretion suppresses antitumor immunity in LKB1-deficient lung adenocarcinoma. Cancer Cell 41(7):1363–1380. 10.1016/j.ccell.2023.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramos RN, Missolo-Koussou Y, Gerber-Ferder Y et al (2022) Tissue-resident FOLR2+ macrophages associate with CD8+ T cell infiltration in human breast cancer. Cell 185(7):1189–1207. 10.1016/j.cell.2022.02.021 [DOI] [PubMed] [Google Scholar]
- Revel M, Sautès-Fridman C, Fridman W-H et al (2022) C1q+ macrophages: passengers or drivers of cancer progression. Trends Cancer 8(7):517–526. 10.1016/j.trecan.2022.02.006 [DOI] [PubMed] [Google Scholar]
- Rowe SP, Pomper MG (2022) Molecular imaging in oncology: current impact and future directions. CA Cancer J Clin 72(4):333–352. 10.3322/caac.21713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi L, Wei M, Miao Y et al (2022) Highly-multiplexed volumetric mapping with Raman dye imaging and tissue clearing. Nat Biotechnol 40(3):364–373. 10.1038/s41587-021-01041-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegel R, Miller K, Fuchs H et al (2021) Cancer statistics, 2021. CA Cancer J Clin 71(4):359–359. 10.3322/caac.21654 [DOI] [PubMed] [Google Scholar]
- Song H, Yang Y, Sun Y et al (2022) Circular RNA Cdyl promotes abdominal aortic aneurysm formation by inducing M1 macrophage polarization and M1-type inflammation. Mol Ther 30(2):915–931. 10.1016/j.ymthe.2021.09.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strack R (2023) Capturing hyperspectral images. Nat Methods 20(6):783–783. 10.1038/s41592-023-01921-z [DOI] [PubMed] [Google Scholar]
- Su W-P, Chang L-C, Song W-H et al (2022) Polyaniline-based glyco-condensation on Au nanoparticles enhances immunotherapy in lung cancer. ACS Appl Mater Interfaces 14(21):24144–24159. 10.1021/acsami.2c03839 [DOI] [PubMed] [Google Scholar]
- Sullivan RJ, Hong DS, Tolcher AW et al (2018) 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 36(15):3013–3013. 10.1200/JCO.2018.36.15_suppl.3013 [Google Scholar]
- Sun L, Zhang H, Gao P (2022) Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell 13(12):877–919. 10.1007/s13238-021-00846-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thorsson V, Gibbs DL, Brown SD et al (2019) The immune landscape of cancer. Immunity 51(2):411–412. 10.1016/j.immuni.2018.03.023 [DOI] [PubMed] [Google Scholar]
- Vazquez-Romero A, Kielland N, Arevalo MJ et al (2013) Multicomponent reactions for de novo synthesis of BODIPY probes: in vivo imaging of phagocytic macrophages. J Am Chem Soc 135(43):16018–16021. 10.1021/ja408093p [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y, Wang M, Wu HX et al (2021) Advancing to the era of cancer immunotherapy. Cancer Commun 41(9):803–829. 10.1002/cac2.12178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang T, Lin M, Mao J et al (2022) Inflammation-regulated nanodrug sensitizes hepatocellular carcinoma to checkpoint blockade therapy by reprogramming the tumor microenvironment. ACS Appl Mater Interfaces 14(44):49542–49554. 10.1021/acsami.2c14448 [DOI] [PubMed] [Google Scholar]
- Wang J, Peng X, Wei J et al (2023) In situ phagocyte-mediated deep tumor penetration assisted by ApoA-1 mimetic peptide-modified silicasome. Nano Today 50:101864. 10.1016/j.nantod.2023.101864 [Google Scholar]
- Xue R, Zhang Q, Cao Q et al (2022) Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature 612(7938):141–147. 10.1038/s41586-022-05400-x [DOI] [PubMed] [Google Scholar]
- Yang L-Z, Wang Y, Li S-Q et al (2019) Dynamic imaging of RNA in living cells by CRISPR-Cas13 systems. Mol Cell 76(6):981–997. 10.1016/j.molcel.2019.10.024 [DOI] [PubMed] [Google Scholar]
- Yu JE, Yeo IJ, Son DJ et al (2022) Anti-Chi3L1 antibody suppresses lung tumor growth and metastasis through inhibition of M2 polarization. Mol Oncol 16(11):2214–2234. 10.1002/1878-0261.13152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C, Yu X, Gao L et al (2017) Noninvasive imaging of CD206-positive M2 macrophages as an early biomarker for post-chemotherapy tumor relapse and lymph node metastasis. Theranostics 7(17):4276. 10.7150/thno.20999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Du X, Liu M et al (2019) Hijacking antibody-induced CTLA-4 lysosomal degradation for safer and more effective cancer immunotherapy. Cell Res 29(8):609–627. 10.1038/s41422-019-0184-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang M, Pan X, Fujiwara K et al (2021a) Pancreatic cancer cells render tumor-associated macrophages metabolically reprogrammed by a GARP and DNA methylation-mediated mechanism. Sig Transduct Target Ther 6(1):366. 10.1038/s41392-021-00769-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y, Chen H, Mo H et al (2021b) Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. Cancer Cell 39(12):1578–1593. 10.1016/j.ccell.2021.09.010 [DOI] [PubMed] [Google Scholar]
- Zhang X, Ji L, Li MO (2023) Control of tumor-associated macrophage responses by nutrient acquisition and metabolism. Immunity 56(1):14–31. 10.1016/j.immuni.2022.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao L, Huang Z, Ma D et al (2021) A nucleus targetable fluorescent probe for ratiometric imaging of endogenous NO in living cells and zebrafishes. Analyst 146(13):4130–4134. 10.1039/D1AN00426C [DOI] [PubMed] [Google Scholar]
- Zhao H, Ming T, Tang S et al (2022) Wnt signaling in colorectal cancer: pathogenic role and therapeutic target. Mol Cancer 21(1):144. 10.1186/s12943-022-01616-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou T, Wang J, Xu J et al (2020) A smart fluorescent probe for NO detection and application in myocardial fibrosis imaging. Anal Chem 92(7):5064–5072. 10.1021/acs.analchem.9b05435 [DOI] [PubMed] [Google Scholar]
- Zhou Q, Liang J, Yang T et al (2022) Carfilzomib modulates tumor microenvironment to potentiate immune checkpoint therapy for cancer. EMBO Mol Med 14(1):e14502. 10.15252/emmm.202114502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou Y, Sun X, Yang Q et al (2022) Blood-brain barrier–penetrating single CRISPR-Cas9 nanocapsules for effective and safe glioblastoma gene therapy. Sci Adv 8(16):eabm8011. 10.1126/sciadv.abm8011 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.



