Citalopram exhibits immune-dependent anti-tumor effects by modulating C5aR1+ TAMs and CD8+ T cells

  1. Department of Gastroenterology, Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital, Fudan University, Shanghai, China
  2. State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
  3. Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
  4. Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
  5. Department of General Surgery, Hepato-biliary-pancreatic Center, Huadong Hospital, Fudan University, Shanghai, China
  6. Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
  7. State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;

Peer review process

Revised: This Reviewed Preprint has been revised by the authors in response to the previous round of peer review; the eLife assessment and the public reviews have been updated where necessary by the editors and peer reviewers.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Ping-Chih Ho
    Ludwig Institute for Cancer Research, Epalinges, Switzerland
  • Senior Editor
    Tadatsugu Taniguchi
    The University of Tokyo, Tokyo, Japan

Reviewer #1 (Public review):

Summary:

In their previous publication (Dong et al. Cell Reports 2024), the authors showed that citalopram treatment resulted in reduced tumor size by binding to the E380 site of GLUT1 and inhibiting the glycolytic metabolism of HCC cells, instead of the classical citalopram receptor. Given that C5aR1 was also identified as the potential receptors of citalopram in the previous report, the authors focused on exploring the potential of immune-dependent anti-tumor effect of citalopram via C5aR1. C5aR1 was found to be expressed on tumor-associated macrophages (TAMs) and citalopram administration showed potential to improve the stability of C5aR1 in vitro. Through macrophage depletion and adoptive transfer approaches in HCC mouse models, the data demonstrated the potential importance of C5aR1-expressing macrophage in the anti-tumor effect of citalopram in vivo. Mechanistically, their in vitro data suggested that citalopram may regulate the phagocytosis potential and polarization of macrophages through C5aR1. Next, they tried to investigate the direct link between citalopram and CD8+T cells by including an additional MASH-associated HCC mouse model. Their data suggest that citalopram may upregulate the glycolytic metabolism of CD8+T cells, probability via GLUT3 but not GLUT1-mediated glucose uptake. Lastly, as the systemic 5-HT level is down-regulated by citalopram, the authors analyzed the association between a low 5-HT and a superior CD8+T cell function against tumor. Although the data is informative, the rationale for working on additional mechanisms and logical link among different parts are not clear. In addition, some of the conclusion is also not fully supported by the current data.

Strengths:

The idea of repurposing clinical-in-used drugs showed great potential for immediate clinical translation. The data here suggested that the anti-depression drug, citalopram displayed immune regulatory role on TAM via a new target C5aR1 in HCC.

Comments on revised version:

The authors have addressed most of my concerns about the paper.

Reviewer #2 (Public review):

Summary:

Dong et al. present a thorough investigation into the potential of repurposing citalopram, an SSRI, for hepatocellular carcinoma (HCC) therapy. The study highlights the dual mechanisms by which citalopram exerts anti-tumor effects: reprogramming tumor-associated macrophages (TAMs) toward an anti-tumor phenotype via C5aR1 modulation and suppressing cancer cell metabolism through GLUT1 inhibition, while enhancing CD8+ T cell activation. The findings emphasize the potential of drug repurposing strategies and position C5aR1 as a promising immunotherapeutic target.

Strengths:

It provides detailed evidence of citalopram's non-canonical action on C5aR1, demonstrating its ability to modulate macrophage behavior and enhance CD8+ T cell cytotoxicity. The use of DARTS assays, in silico docking, and gene signature network analyses offers robust validation of drug-target interactions. Additionally, the dual focus on immune cell reprogramming and metabolic suppression presents a comprehensive strategy for HCC therapy. By highlighting the potential for existing drugs like citalopram to be repurposed, the study also emphasizes the feasibility of translational applications. During revision, the authors experimentally demonstrated that TAM has lower GLUT1, which further strengthens their claim of C5aR1 modulation-dependent TAM improvement for tumor therapy.

Weaknesses:

The authors proposed that CD8+ T cells have an TAM-independent role upon Citalropharm treatment. However, this claim requires further investigation to confirm that the effect is truly "TAM independent".

Author response:

The following is the authors’ response to the original reviews.

Reviewer #1 (Public review):

Summary:

In their previous publication (Dong et al. Cell Reports 2024), the authors showed that citalopram treatment resulted in reduced tumor size by binding to the E380 site of GLUT1 and inhibiting the glycolytic metabolism of HCC cells, instead of the classical citalopram receptor. Given that C5aR1 was also identified as the potential receptor of citalopram in the previous report, the authors focused on exploring the potential of the immune-dependent anti-tumor effect of citalopram via C5aR1. C5aR1 was found to be expressed on tumor-associated macrophages (TAMs) and citalopram administration showed potential to improve the stability of C5aR1 in vitro. Through macrophage depletion and adoptive transfer approaches in HCC mouse models, the data demonstrated the potential importance of C5aR1-expressing macrophage in the anti-tumor effect of citalopram in vivo. Mechanistically, their in vitro data suggested that citalopram may regulate the phagocytosis potential and polarization of macrophages through C5aR1. Next, they tried to investigate the direct link between citalopram and CD8+T cells by including an additional MASH-associated HCC mouse model. Their data suggest that citalopram may upregulate the glycolytic metabolism of CD8+T cells, probability via GLUT3 but not GLUT1-mediated glucose uptake. Lastly, as the systemic 5-HT level is down-regulated by citalopram, the authors analyzed the association between a low 5-HT and a superior CD8+T cell function against a tumor. Although the data is informative, the rationale for working on additional mechanisms and logical links among different parts is not clear. In addition, some of the conclusion is also not fully supported by the current data.

We thank the reviewer for their comprehensive summary of our study and appreciate the valuable feedback. We have made improvements based on these comments, and a detailed response addressing each point is presented below.

Strengths:

The idea of repurposing clinical-in-used drugs showed great potential for immediate clinical translation. The data here suggested that the anti-depression drug, citalopram displayed an immune regulatory role on TAM via a new target C5aR1 in HCC.

We thank the reviewer for recognizing the strengths of our study.

Weaknesses:

(1) The authors concluded that citalopram had a 'potential immune-dependent effect' based on the tumor weight difference between Rag-/- and C57 mice in Figure 1. However, tumor weight differences may also be attributed to a non-immune regulatory pathway. In addition, how do the authors calculate relative tumor weight? What is the rationale for using relative one but not absolute tumor weight to reflect the anti-tumor effect?

We appreciate your insights into the potential contributions of non-immune regulatory pathways to the observed tumor weight differences between Rag1-/- and wild type C57BL/6 mice. Indeed, the anti-tumor effects of citalopram involve non-immune mechanisms. Previously, we have demonstrated the direct effects of citalopram on cancer cell proliferation, apoptosis, and metabolic processes (PMID: 39388353). In this study, we focused on immune-dependent mechanisms, utilizing Rag1-/- mice to investigate a potential immune-mediated effect. The relative tumor weight was calculated by assigning an arbitrary value of 1 to the Rag1-/- mice in the DMSO treatment group, with all other tumor weights expressed relative to this baseline. As suggested, we have included absolute tumor weight data in the revised Figure 1B, 1E, 1F, and 3B.

(2) The authors used shSlc6a4 tumor cell lines to demonstrate that citalopram's effects are independent of the conventional SERT receptor (Figure 1C-F). However, this does not entirely exclude the possibility that SERT may still play a role in this context, as it can be expressed in other cells within the tumor microenvironment. What is the expression profiling of Slc6a4 in the HCC tumor microenvironment? In addition, in Figure 1F, the tumor growth of shSlc6a4 in C57 mice displayed a decreased trend, suggesting a possible role of Slc6a4.

As suggested, we probed the expression pattern of SERT in HCC and its tumor microenvironment. Using a single cell sequencing dataset of HCC (GSE125449), we revealed that SERT is also expressed by T cells, tumor-associated endothelial cells, and cancer-associated fibroblasts (see revised Figure S2G). Therefore, we cannot fully rule out the possibility that citalopram may influence these cellular components within the TME and contribute to its therapeutic effects. In the revised manuscript, we have included and discussed this result. In Figure 1F, SERT knockdown led to a 9% reduction in tumor growth, however, this difference was not statistically significant (0.619 ± 0.099 g vs. 0.594 ± 0.129 g; p = 0.75).

(3) Why did the authors choose to study phagocytosis in Figures 3G-H? As an important player, TAM regulates tumor growth via various mechanisms.

We choose to investigate phagocytosis because citalopram targets C5aR1-expressing TAM. C5aR1 is a receptor for the complement component C5a, which plays a crucial role in mediating the phagocytosis process in macrophages. In the revised manuscript, we have highlighted this rationale.

(4) The information on unchanged deposition of C5a has been mentioned in this manuscript (Figures 3D and 3F), the authors should explain further in the manuscript, for example, C5a could bind to receptors other than C5aR1 and/or C5a bind to C5aR1 by different docking anchors compared with citalopram.

Thank you for your insightful comment. In Figure 3D, tumor growth was attenuated in C5ar1-/- recipients compared with C5ar1-/- recipients, whereas C5a deposition remained unchanged. This suggests that while C5a is still present, its interaction with C5aR1 is critical for influencing tumor growth dynamics. In Figure 3F, C5a deposition was not affected by citalopram treatment. Indeed, docking analysis and DARTS assay revealed that citalopram binds to the D282 site of C5aR1. Previous report has shown that mutations on E199 and D282 reduce C5a binding affinity to C5aR1 (PMID: 37169960). Therefore, the impact of citalopram is primarily on C5a/C5aR1 interactions and downstream signaling pathways, rather than on altering C5a levels. In the revised manuscript, we have included this interpretation.

(5) Figure 3I-M - the flow cytometry data suggested that citalopram treatment altered the proportions of total TAM, M1 and M2 subsets, CD4+ and CD8+T cells, DCs, and B cells. Why does the author conclude that the enhanced phagocytosis of TAM was one of the major mechanisms of citalopram? As the overall TAM number was regulated, the contribution of phagocytosis to tumor growth may be limited.

We thank the reviewer’s valuable input. Indeed, recent studies have demonstrated that targeting C5aR1+ TAMs can induce many anti-tumor effects, such as macrophage polarization and CD8+ T cell infiltration (PMID: 30300579, PMID: 38331868, and PMID: 38098230). In the revised manuscript, we have clarified our conclusion to better articulate the relationship between citalopram treatment, TAM populations, and their phagocytic activity, with particular emphasis on the role of CD8+ T cells. For macrophage phagocytosis, one possible explanation is that citalopram targets C5aR1 to enhance macrophage phagocytosis and subsequent antigen presentation and/or cytokine production, which promotes T cell recruitment and activity as well as modulate other aspects of tumor immunity. Given that the anti-tumor effects of citalopram are largely dependent on CD8+ T cells, we conclude that CD8+ T cells are essential for the effector mechanisms of citalopram.

(6) Figure 4 - what is the rationale for using the MASH-associated HCC mouse model to study metabolic regulation in CD8+ T cells? The tumor microenvironment and tumor growth would be quite different. In addition, how does this part link up with the mechanisms related to C5aR1 and TAM? The authors also brought GLUT1 back in the last part and focused on CD8+ T cell metabolism, which was totally separated from previous data.

We chose the MASH-associated HCC mouse model because it closely mimics the etiology of metabolic-associated fatty liver disease (MAFLD), which is a significant contributor to the development of cirrhosis and HCC. In addition to the MASH-associated HCC mouse model, the study also incorporated the orthotopic Hepa1-6 tumor model. In our previous publication (Dong et al., Cell Reports 2024), we employed both of these HCC models. Therefore, we utilized the same two mouse models in this study. The inclusion of CD8+ T cells in our study is based on the understanding that citalopram targets GLUT1, which plays a crucial role in glucose uptake (PMID: 39388353). CD8+T cell function is heavily reliant on glycolytic metabolism, making it essential to investigate how citalopram’s effects on GLUT1 influence the metabolic pathways and functionality of these immune cells. In this study, we identified that the primary glucose transporter in CD8+ T cells is GLUT3, rather than GLUT1. The data presented in Figure 4 aim to illustrate the additional effect of citalopram on peripheral 5-HT levels, which, in turn, influences CD8+ T cell functionality. By linking these findings, we clarify how citalopram impacts both TAMs and CD8+ T cells. CD8+ T cells can be influenced by citalopram through various mechanisms, including TAM-dependent mechanisms, reduced systemic serum 5-HT concentrations, and unidentified direct effects. In the revised manuscript, we have enhanced the background information to avoid any gaps.

(7) Figure 5, the authors illustrated their mechanism that citalopram regulates CD8+ T cell anti-tumor immunity through proinflammatory TAM with no experimental evidence. Using only CD206 and MHCII to represent TAM subsets obviously is not sufficient.

Thank you for your valuable comments. As noted by the reviewer, TAMs can influence CD8+ T cell anti-tumor immunity through various mechanisms. In this study, we focused on elucidating the impact of citalopram on pro-inflammatory TAMs, which in turn affect CD8+ T cell anti-tumor immunity and ultimately influence tumor outcomes. Therefore, in the mechanistic diagram, we highlighted the effect of citalopram on pro-inflammatory TAMs, while the causal relationship between TAMs and CD8+ T cell anti-tumor immunity was indicated with a dotted line due to the limited evidence presented in this study. Additionally, we have expanded our discussion on how citalopram regulates CD8+ T cell anti-tumor immunity through pro-inflammatory TAMs.

For the analysis of TAMs, we initially sorted CD45+F4/80+CD11b+ cells and assessed M1/M2 polarization by measuring CD206 and MHCII expression. As an added strength, we isolated TAMs from the orthotopic GLUT1KD Hepa1-6 model using CD11b microbeads and conducted real-time qPCR analysis of M1-oriented (Il6, Ifnb1, and Nos2) and M2-oriented (Mrc1, Il10, and Arg1) markers. Consistent with our flow cytometry data, the qPCR results confirmed that citalopram induces a pro-inflammatory TAM phenotype (revised Figure S9A).

Reviewer #2 (Public review): Summary:

Dong et al. present a thorough investigation into the potential of repurposing citalopram, an SSRI, for hepatocellular carcinoma (HCC) therapy. The study highlights the dual mechanisms by which citalopram exerts anti-tumor effects: reprogramming tumor-associated macrophages (TAMs) toward an anti-tumor phenotype via C5aR1 modulation and suppressing cancer cell metabolism through GLUT1 inhibition while enhancing CD8+ T cell activation. The findings emphasize the potential of drug repurposing strategies and position C5aR1 as a promising immunotherapeutic target. However, certain aspects of experimental design and clinical relevance could be further developed to strengthen the study's impact.

We thank the reviewer’s thoughtful review and constructive feedback. As suggested, we have made improvements based on the feedback provided.

Strength:

It provides detailed evidence of citalopram's non-canonical action on C5aR1, demonstrating its ability to modulate macrophage behavior and enhance CD8+ T cell cytotoxicity. The use of DARTS assays, in silico docking, and gene signature network analyses offers robust validation of drug-target interactions. Additionally, the dual focus on immune cell reprogramming and metabolic suppression presents a thorough strategy for HCC therapy. By emphasizing the potential for existing drugs like citalopram to be repurposed, the study also underscores the feasibility of translational applications.

We sincerely appreciate the reviewer’s recognition of the detailed evidence supporting citalopram’s non-canonical action on C5aR1, along with the innovative methodologies employed and the promising potential for repurposing existing drugs in HCC therapy.

Major weaknesses/suggestions:

The dataset and signature database used for GSEA analyses are not clearly specified, limiting reproducibility. The manuscript does not fully explore the potential promiscuity of citalopram's interactions across GLUT1, C5aR1, and SERT1, which could provide a deeper understanding of binding selectivity. The absence of GLUT1 knockdown or knockout experiments in macrophages prevents a complete assessment of GLUT1's role in macrophage versus tumor cell metabolism. Furthermore, there is minimal discussion of clinical data on SSRI use in HCC patients. Incorporating survival outcomes based on SSRI treatment could strengthen the study's translational relevance.

By addressing these limitations, the manuscript could make an even stronger contribution to the fields of cancer immunotherapy and drug repurposing.

We appreciate the reviewer’s valuable suggestions. As suggested, we have included the following revisions:

(a) GSEA analyses: For GSEA analyses, we conducted RNA sequencing (RNA-seq) analysis on HCC-LM3 cells treated with citalopram or fluvoxamine, which led to the identification of 114 differentially expressed genes (DEGs; 80 co-upregulated and 34 co-downregulated), as reported previously (PMID: 39388353). These DEGs were then utilized to create an SSRI-related gene signature. Subsequently, we analyzed RNA-seq data from liver HCC (LIHC) samples in The Cancer Genome Atlas (TCGA) cohort, comprising 371 samples, categorizing them into high and low expression groups based on the median expression levels of each candidate target gene (such as C5AR1). Finally, we performed GSEA on the grouped samples (C5AR1-high versus C5AR1-low) using the SSRI-related gene signature. In the revised manuscript, we have included this information in the “Materials and Methods” section.

(b) Exploration of binding selectivity: We acknowledge the importance of exploring the potential promiscuity of citalopram’s interactions across GLUT1, C5aR1, and SERT1. While we cannot provide further experimental data to support this aspect, we have included the following points in the revised manuscript: 1) We emphasize the significance of exploring the relative binding affinities of citalopram to GLUT1, C5aR1, and SERT, as varying affinities could influence the drug’s overall efficacy. As highlighted in the current manuscript and our previous publication (PMID: 39388353), citalopram interacts with C5aR1 and GLUT1 through distinct binding sites and mechanisms, whereas its interaction with SERT is characterized by a more direct inhibition of serotonin binding (PMID: 27049939). To gain deeper insights into these interactions, employing techniques such as surface plasmon resonance or biolayer interferometry could provide valuable quantitative data on binding kinetics and affinities for each target. 2) We discuss how citalopram’s interactions with multiple targets may contribute to its therapeutic effects, particularly in the context of immune modulation and tumor progression. The potential for citalopram to exhibit diverse mechanisms of action through its interactions with these proteins warrants further investigation. A comprehensive understanding of these pathways could lead to the development of improved therapeutic strategies.

(c) GLUT1 knockdown in macrophages: In the revised manuscript, we revealed that TAMs predominantly express GLUT3 but not GLUT1 (Figures S8B and S8C). GLUT1 knockdown in THP-1 cells did not significantly impact their glycolytic metabolism (Figure S8D), whereas GLUT3 knockdown led to a marked reduction in glycolysis in THP-1 cells.

(d) Clinical data on SSRI use in HCC patients: Previously, we have reported that SSRIs use is associated with reduced disease progression in HCC patients (PMID: 39388353) (Cell Rep. 2024 Oct 22;43(10):114818.). As detailed below:

“We determined whether SSRIs for alleviating HCC are supported by real-world data. A total of 3061 patients with liver cancer were extracted from the Swedish Cancer Register. Among them, 695 patients had been administrated with post-diagnostic SSRIs. The Kaplan-Meier survival analysis suggested that patients who utilized SSRIs exhibited a significantly improved metastasis-free survival compared to those who did not use SSRIs, with a P value of log-rank test at 0.0002. Cox regression analysis showed that SSRI use was associated with a lower risk of metastasis (HR = 0.78; 95% CI, 0.62-0.99)”.

Reviewer #1 (Recommendations for the authors):

(1) Add experiments to address the questions listed in the weaknesses.

As suggested, related experiments are performed to strengthen the conclusions.

(2) It would be appreciated to show the expression profile of SERT or employ KO mouse models to eliminate the effect of SERT.

As suggested, analysis of a single-cell sequencing dataset of HCC (GSE125449) revealed that SERT is expressed not only in HCC cells but also in T cells, tumor-associated endothelial cells, and cancer-associated fibroblasts (Figure S2G). Consistently, SERT has been reported as an immune checkpoint restricting CD8 T cell antitumor immunity (PMID: 40403728). Furthermore, SERT KO mice (Cyagen Biosciences, S-KO-02549) was employed to investigate the effects of citalopram. However, the Slc6a4 gene knockout in mice resulted in a significant decrease in 5-HT levels in the brain and a lack of cortical columnar structures. Importantly, the mice exhibited an intolerance to citalopram treatment. Therefore, we did not pursue further investigation into the effects of citalopram in SERT KO mice.

(3) Due to the concern of specificity and animal health, it would be more direct if the authors could use, for example, C5ar1-fl/fl x Adgre1-Cre mouse models.

Thank you for your valuable suggestion. We fully agree with your comment regarding the value of introducing C5ar1-fl/fl and Adgre1-Cre mouse models, along with the necessary experimental setups, to substantiate this point. However, in our study, the C5ar1 KO mice exhibited normal overall appearance and viability, indicating that the model is generally healthy. Furthermore, we have validated the specific role of C5aR1 in macrophages through bone marrow reconstitution experiments, reinforcing the importance of C5aR1 in these cells. Therefore, we chose the current model to balance experimental effectiveness with considerations for animal health.

(4) For example, a GSEA or GO analysis of comparison of macrophages from C5ar1-/- or C5ar1+/- mice may point to the enriched pathway of phagocytosis in macrophages derived from C5ar1-/- rather than C5ar1+/- mice, and this information is helpful for the integrity of this work. Besides, it would be more reliable if a nucleus staining is included in Figures 3G and 3H.

As suggested, macrophages were isolated from tumor-bearing C5ar1-/- and C5ar1+/- mice and subsequently analyzed using RNA sequencing. The Gene Set Enrichment Analysis (GSEA) revealed a significant enrichment of the phagocytosis pathway in macrophages derived from C5ar1-/- mice compared to those from C5ar1+/- mice (see revised Figure S6A). While we acknowledge that the addition of a nucleus staining would enhance reliability, we would like to point out that this style of presentation is also commonly found in articles related to phagocytosis. Furthermore, this experiment involved a significant number of experimental mice, and in accordance with the 3Rs principle for animal experiments, we did not obtain additional sorted TAMs to perform the phagocytosis assay. Thank you for your understanding.

(5) In line 122, there is a typo, and it should be 'analysis'.

Thank you for pointing out the typo. It has been corrected to "analysis" in the revised manuscript.

(6) In line 217, there is no causal relationship between the contexts, and using 'as a result' may lead to misunderstanding.

As suggested, ‘as a result’ has been removed to avoid any misunderstanding.

(7) In line 322, please make sure if it should be HBS or PBS.

It is PBS, and revisions have been made.

(8) Figure S7, the calculation of cell proportions needs to use a consistent denominator.

As suggested, we calculated cell proportions using a consistent denominator (CD45+ cells).

(9) Figure 4C, label error.

Thanks for your careful review. It has been corrected to "MASH".

Reviewer #2 (Recommendations for the authors):

Dong et al. present compelling evidence for repurposing citalopram, a selective serotonin reuptake inhibitor (SSRI), as a potential therapeutic for hepatocellular carcinoma (HCC). While the concept of SSRI repurposing is not novel, this manuscript provides valuable insights into the drug's dual mechanisms: targeting tumor-associated macrophages (TAMs) via C5aR1 modulation and enhancing CD8+ T cell activity, alongside inhibiting cancer cell metabolism through GLUT1 suppression. The findings underscore the promise of drug repurposing strategies and identify C5aR1 as a noteworthy immunotherapeutic target. Addressing the following points will enhance the manuscript's impact and relevance to cancer immunotherapy.

Specific Comments:

(1) The authors identify C5aR1 on TAMs as a direct target of citalopram, independent of its classical SERT target, using drug-induced gene signature network analysis and co-immunofluorescence of CD163+ macrophages with C5aR1. The DARTS assay further supports the binding of C5aR1 to citalopram, complemented by in silico docking analysis adapted from their previous GLUT1 study. Since GLUT1 and SERT1 are transporter proteins while C5aR1 is a GPCR, these heterogeneous binding interactions suggest potential promiscuity in SSRI-target engagement.

(a) Figure 2A: The authors identify C5aR1 as a target using GSEA but do not specify the dataset used (e.g., cancer or immune cells) or the signature database consulted. Providing this context would enhance reproducibility.

For GSEA, we performed RNA sequencing (RNA-seq) on HCC-LM3 cells treated with citalopram or fluvoxamine and identified 114 differentially expressed genes (DEGs), which included 80 genes that were co-upregulated and 34 that were co-downregulated, as previously documented (PMID: 39388353). These DEGs were subsequently used to develop an SSRI-related gene signature. We then employed the RNA-seq data from liver hepatocellular carcinoma (LIHC) samples within The Cancer Genome Atlas (TCGA) cohort, which included 371 samples. HCC samples in the TCGA cohort were categorized into high and low expression groups based on the median expression levels of each candidate target gene, such as C5AR1. Finally, we conducted GSEA on the grouped samples (such as C5AR1-high versus C5AR1-low) using the SSRI-related gene signature. For reproducibility, detailed information has been added to the “Materials and Methods” section of the revised manuscript.

(b) Figure 2F: Given citalopram's reported role in inhibiting GLUT1, a comparative discussion on the relative contributions of GLUT1 inhibition versus C5aR1 modulation in tumor suppression is warranted. Performing a DARTS assay for GLUT1 in THP-1 cells, which express high GLUT1 levels and exhibit upregulation in M1 macrophages (https://doi.org/10.1038/s41467-022-33526-z), would clarify SSRI interactions with macrophage metabolism.

As suggested, we first investigated citalopram treatment in THP-1 cells. The result showed the glycolytic metabolism of THP-1 cells remained largely unaffected following citalopram treatment, as evidenced by glucose uptake, lactate release, and extracellular acidification rate (ECAR) (Figure S8A). Next, we mined a single cell sequencing datasets of HCC and revealed that TAMs predominantly express GLUT3 but not GLUT1 (Figure S8B). Consistently, Western blotting analysis showed a higher expression of GLUT3 and minimal levels of GLUT1 in THP-1 cells (Figure S8C). Consistently, it has been well documented that GLUT1 expression increased after M1 polarization stimuli an GLUT3 expression increased after M2 stimulation in macrophages (PMID: 37721853, PMID: 36216803). GLUT1 knockdown in THP-1 cells did not significantly impact their glycolytic metabolism (Figure S8D), whereas GLUT3 knockdown led to a marked reduction in glycolysis in THP-1 cells. Based on these findings, we conclude that the effects of citalopram on macrophages are primarily mediated through targeting C5aR1 rather than GLUT1.

(c) Figures 2H-I: A comparison of drug-protein interactions across GLUT1, C5aR1, and SERT1 would be valuable to identify potential shared or distinct binding features.

Citalopram exhibits distinct binding characteristics across its various targets, including GLUT1, C5aR1, and its classical target, SERT. In the case of C5aR1, our in silico docking analysis identified two key binding conformations at the orthosteric site. The interactions involved significant electrostatic contacts between citalopram’s amino group and negatively charged residues like E199 and D282. Notably, D282’s accessibility and orientation towards the binding cavity suggest it plays a crucial role in citalopram binding, highlighting the importance of specific amino acid interactions at this site. For GLUT1 (PMID: 39388353), citalopram’s interaction also demonstrated notable hydrophobic contacts, particularly through the fluorophenyl group with residues V328, P385, and L325. The cyanophtalane group penetrated the substrate-binding cavity, indicating that citalopram could occupy a similar binding site as glucose, which is distinct from the binding mechanism observed in C5aR1. The involvement of E380 in both poses for GLUT1 further emphasizes the role of electrostatic interactions in mediating citalopram’s binding to this transporter. In contrast, for SERT (PMID: 27049939), citalopram locks the transporter in an outward-open conformation by occupying the central binding site, which is located between transmembrane helices 1, 3, 6, 8 and 10. This binding directly obstructs serotonin from accessing its binding site, illustrating a more definitive blockade mechanism. Additionally, the allosteric site at SERT, positioned between extracellular loops 4 and 6 and transmembrane helices 1, 6, 10, and 11, enhances this blockade by sterically hindering ligand unbinding, thus providing a clear explanation for the allosteric modulation of serotonin transport. In summary, while citalopram interacts with C5aR1 and GLUT1 through distinct binding sites and mechanisms, its interaction with SERT is characterized by a more straightforward blockade of serotonin binding. The unique structural and functional attributes of each target highlight the versatility of citalopram and suggest that its pharmacological effects may vary significantly depending on the specific protein being targeted. In the revised manuscript, we have included detailed information in the revised manuscript.

(2) The manuscript presents evidence that citalopram reprograms TAMs to an anti-tumor phenotype, enhancing their phagocytic capacity.

(a) Bone Marrow Reconstitution Experiments (Figure 3): The use of donor (dC5aR1) and recipient (rC5aR1) mice is significant but requires clarification. Explicitly defining donor and recipient terminology and including a schematic of the experimental design would improve reader comprehension.

We appreciate your valuable feedback. As suggested, the terminology for donor (dC5aR1) and recipient (rC5aR1) mice was defined: “we injected GLUT1KD Hepa1-6 cells into syngeneic recipient C5ar1-/- (rC5ar1-/- ) mice that had been reconstituted with donor C5ar1+/- (dC5ar1+/-) or C5ar1-/- (dC5ar1-/-) bone marrow (BM) cells to analyze the therapeutic effect of citalopram”. Additionally, we have included a schematic of the experimental design to enhance reader comprehension (see revised Figure 3E).

(b) GLUT1 Knockdown (KD) Tumor Cells: While GLUT1 KD tumor cells are utilized, the authors do not assess GLUT1 KD or knockout (KO) in macrophages. Testing the effect of citalopram on macrophages with GLUT1 KO/KD would help determine the relative importance of C5aR1 versus GLUT1 in mediating SSRI effects.

As responded above, GLUT1 knockdown in THP-1 cells did not significantly alter their glycolytic metabolism (Figure S8D). This observation can be explained by the predominant expression of GLUT3 in TAMs rather than GLUT1 (Figures S8B and S8C). Indeed, knockdown of GLUT3 led to a significant reduction in glycolysis in THP-1 cells (Figure S8C).

(c) C5aR1's Pro-Tumoral Role: The authors state that C5aR1 fosters an immunosuppressive microenvironment but omit a discussion of current literature on C5aR1's pro-tumoral role (e.g., https://doi.org/10.1038/s41467-024-48637-y, https://www.nature.com/articles/s41419-024-06500-4, https://doi.org/10.1016/j.ymthe.2023.12.010). Including this background in both the introduction and discussion would contextualize their findings.

Thanks for your valuable feedback. As suggested, we have revised the manuscript to include discussions on C5aR1’s pro-tumoral role, referencing the suggested studies in both the introduction and discussion sections for better context. As detailed below:

(1) Targeting C5aR1+ TAMs effectively reverses tumor progression and enhances anti-tumor response;

(2) Targeting C5aR1 reprograms TAMs from a protumor state to an antitumor state, promoting the secretion of CXCL9 and CXCL10 while facilitating the recruitment of cytotoxic CD8+ T cells;

(3) Moreover, citalopram induces TAM phenotypic polarization towards to a M1 proinflammatory state, which supports anti-tumor immune response within the TME.

(d) C5aR1 Expression in TAMs: Is C5aR1 expression constitutive in TAMs? Further details on C5aR1 expression dynamics in TAMs under different conditions could strengthen the discussion. Public datasets on TAMs in various states (e.g., https://www.nature.com/articles/s41586-023-06682-5, https://www.cell.com/cell/abstract/S0092-8674(19)31119-5, https://pubmed.ncbi.nlm.nih.gov/36657444/) may offer useful insights.

Thank you for your valuable suggestions. As suggested, we investigated the expression patterns of C5aR1 in TAMs using a HCC cohort (http://cancer-pku.cn:3838/HCC/). In the study conducted by Qiming Zhang et al. (PMID: 31675496), six distinct macrophage subclusters were identified, with M4-c1-THBS1 and M4-c2-C1QA showing significant enrichment in tumor tissues. M4-c1-THBS1 was enriched with signatures indicative of myeloid-derived suppressor cells (MDSCs), while M4-c2-C1QA exhibited characteristics that resembled those of TAMs as well as M1 and M2 macrophages. Our subsequent analysis revealed that C5aR1 is highly expressed in these two clusters, while expression levels in the other macrophage clusters were notably lower (see revised Figure S3).

(3) The manuscript shows that citalopram-induced reductions in systemic serotonin levels enhance CD8+ T cell activation and cytotoxicity, as evidenced by increased glycolytic metabolism and elevated IFN-γ, TNF-α, and GZMB expression.

(a) How CD8+ T cell activation is done in serotonin-deficient environments?

As reported (PMID: 34524861), one possible explanation is that serotonin may enhance PD-L1 expression on cancer cells, thereby impairing CD8+ T cell function. A deficiency of serotonin in the tumor microenvironment can delay tumor growth by promoting the accumulation and effector functions of CD8+ T cells while reducing PD-L1 expression. In addition to the SERT-mediated transport and 5-HT receptor signaling, CD8+ T cells can express TPH1 (PMID: 38215751, PMID: 40403728), enabling them to synthesize endogenous 5-HT, which activates their activity through serotonylation-dependent mechanisms (PMID: 38215751). In the revised manuscript, we have incorporated these interpretations.

(4) Suggestions for the model figure revision-C5aR1 in TAMs without Citalopram (Figure 5).

(a) Including a control scenario depicting receptor status and function in TAMs without citalopram treatment would provide a clearer baseline for understanding citalopram's effects.

Thank you for your valuable input regarding the model figure revision. We have included a revised mechanism model that depicts the receptor status and function of C5aR1 in TAMs without citalopram treatment, as you suggested.

(5) Suggestions for addressing clinical relevance.

The study predominantly uses preclinical mouse models, although some human HCC data is analyzed (Figures 2B and 3O). However, there is no discussion of clinical data on SSRI use in HCC patients.

Incorporating an analysis of patient survival outcomes based on SSRI treatment (e.g., https://pmc.ncbi.nlm.nih.gov/articles/PMC5444756/, https://pmc.ncbi.nlm.nih.gov/articles/PMC10483320/) would enhance the translational relevance of the findings.

Previously, we reported that the use of SSRIs is associated with reduced disease progression in HCC patients, based on real-world data from the Swedish Cancer Register (PMID: 39388353). As suggested, we have further discussed the clinical relevance of SSRIs in the revised manuscript. As detailed below:

“In a study involving 308,938 participants with HCC, findings indicated that the use of antidepressants following an HCC diagnosis was linked to a decreased risk of both overall mortality and cancer-specific mortality (PMID: 37672269). These associations were consistently observed across various subgroups, including different classes of antidepressants and patients with comorbidities such as hepatitis B or C infections, liver cirrhosis, and alcohol use disorders. Similarly, our analysis of real-world data from the Swedish Cancer Register demonstrated that SSRIs are correlated with slower disease progression in HCC patients (PMID: 39388353). Given these insights, antidepressants, especially SSRIs, show significant potential as anticancer therapies for individuals diagnosed with HCC”.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation