Graphical abstract graphic file with name fx1.jpg [73]Open in a new tab Highlights * • Macrophage-mediated lymphoma cell phagocytosis is increased by PPP inhibition * • PPP inhibition is an immune-regulatory switch for macrophage function * • PPP inhibition is linked to the modulation of the UDPG-Stat1-Irg1-itaconate axis * • PPP inhibition is tolerable in vivo and boosts therapeutic B cell lymphoma targeting __________________________________________________________________ Beielstein et al. have shown that the metabolic inhibition of the pentose phosphate pathway in macrophages leads to increased macrophage activation, less lymphoma cell support, and increased antibody-dependent lymphoma cell clearance by macrophages. A significantly improved overall survival in a therapeutic lymphoma mouse model is achieved. Introduction The tumor microenvironment (TME) represents a hallmark of cancer, and interactions between its transformed and non-transformed immune bystander cells determine disease progression and therapeutic response.[74]^1 Tumor cells generate a tumor-supportive environment by cytokine and metabolite secretion. These mediators alter occurrence of bystander cells and shift the activity of the infiltrating immune cells from an anti- to a pro-tumoral response.[75]^2 Tumor-associated macrophages (TAMs) play a critical role in promoting tumor growth, facilitating vascularization and metastasis and suppressing other immune cells.[76]^3^,[77]^4 However, we have shown that macrophages are central for tumor cell clearance in aggressive B cell lymphoma during immunotherapy, although their phagocytic capacity becomes impaired by lymphoma cells.[78]^5 Recent decades have seen the development of numerous new treatment strategies for B cell malignancies, which have extended patient survival but struggled to substantially increase cure rates. The front-line strategy is chemo-immunotherapy, combining therapeutic antibodies like rituximab or obinutuzumab with chemotherapy. We demonstrated that leukemia cells in therapy-refractory niches reduce the engulfment of antibody-targeted tumor cells by macrophages, diminishing therapy efficacy and leading to relapse.[79]^5 Macrophage function depends on their local environment, which impacts their differentiation and polarization. Macrophages are a heterogeneous population with different subtypes exerting pro-inflammatory and phagocytic (M1-like macrophages) and anti-inflammatory and tissue-regenerative (M2-like macrophages) activities. TAMs represent a blend of these characteristics tending toward the anti-inflammatory and phagocytic inactive phenotype.[80]^6^,[81]^7 The TME includes activation mediators such as cytokines, chemokines, and metabolites, which control the polarization of contained macrophages. Changes in the microenvironment can alter macrophage metabolism, which is crucial for polarization and the closely linked macrophage function.[82]^8^,[83]^9 Changes in the cellular metabolism have the ability to repolarize the macrophage phenotype by which pro-tumoral TAMs could acquire anti-tumoral activity.[84]^10^,[85]^11^,[86]^12 The interaction between macrophages and lymphoma cells, as well as macrophages metabolic sensitivity, opens up a promising strategy to optimize anti-cancer therapy. Modulating macrophage metabolism may improve their anti-tumor efficacy and diminish their tumor-supportive function. In the present study, we demonstrate that pentose phosphate pathway (PPP) inhibition in macrophages increases their activity and phagocytic capacity whereby pro-tumoral bystander function is diminished. As a driving mechanism, we discovered a connection between metabolism and immune regulation by modulation of the UDPG-Stat1-Irg1-itaconate axis. The effects of PPP inhibition were transmitted into human patient samples and also reproduced in vivo, where significantly increased survival in an aggressive lymphoma mouse model was achieved. These results open up a promising field of treatment strategy against B cell malignancies in clinical use. Results Metabolic inhibition of the PPP leads to increased phagocytic capacity of macrophages To investigate how metabolic modulation of TAMs in the context of immunotherapy affects phagocytic capacity, we performed a metabolism-focused screening approach for antibody-dependent cellular phagocytosis (ADCP). Key metabolic pathways were blocked using representative inhibitors in a macrophage and humanized aggressive B cell lymphoma (hMB; cell line information see [87]STAR Methods) co-culture-assay system, and phagocytosis was assessed through specific antibody targeting (alemtuzumab; anti-CD52) ([88]Figure 1A). The antibody alemtuzumab was used as a tool compound for the first screening approach as several types of lymphoma downregulate CD20 expression but not CD52 expression, also seen in hMB cells. Several other antibodies, currently in clinical use, were investigated in further analysis. Figure 1. [89]Figure 1 [90]Open in a new tab Metabolic inhibition of the pentose phosphate pathway leads to increased phagocytic rate of macrophages (A) Scheme of ADCP-based metabolic screening approach. (B–D) Summary of ADCP change compared to basal phagocytosis rate of J774A.1 macrophages under inhibition of respective metabolic pathways. (B) Inhibition of only macrophages. (C) Inhibition of only hMB cells. (D) Inhibition of all co-culture components. Technical replicates (B) n = 15–22, (C) n = 15–58, (D) n = 15–28; biological replicates (B) n = 3–5, (C) n = 3–12, (D) n = 3–6. Data are shown as mean ± SEM. p values were calculated using unpaired t test. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [91]Figures S1 and [92]S2. Inhibition of glycolysis (via 2-deoxy-D-glucose), AMP-activated protein kinase (AMPK)-mediated cell energy regulation (via BML-275), mitochondrial ATP production (via oligomycin), and the PPP (via 6-aminonicotinamide and oxythiamine) was screened using non-toxic inhibitor concentrations ([93]Figure S1). The inhibition was conducted in co-culture and by pre-treatment of each cell type (macrophage or hMB cell), to infer specific macrophage vs. lymphoma cell phagocytic interactions. As the basal phagocytosis rate of macrophages is variable, the change in phagocytosis under treatment was calculated in comparison to the basal phagocytosis rate (=ADCP change, [94]Figures 1A and [95]S2). Glycolysis inhibition significantly increased ADCP rates in co-culture (+40%, p < 0.01) and by pre-treatment of lymphoma cells (+95%, p < 0.0001), while macrophage pre-treatment significantly diminished ADCP rate (−71%, p < 0.001) ([96]Figures 1B–1D). Similarly, AMPK inhibition increased ADCP rate significantly by lymphoma cell pre-treatment (+39%, p < 0.0001) and significantly diminished ADCP rate by pre-treatment of macrophages (−42%, p < 0.001) ([97]Figures 1C and 1D). Inhibition of mitochondrial ATP production also diminished ADCP rate significantly by macrophage pre-treatment (−21%, p < 0.0001) ([98]Figure 1D). Sole inhibition of the PPP induced significantly increased ADCP rates by co-culture treatment and macrophage pre-treatment. The increase was induced by both inhibition of the oxidative part of the PPP via 6-phosphogluconate dehydrogenase inhibition (6Pgd; inhibitor 6-aminonicotinamide) (co-culture +40% p < 0.01; macrophage pre-treatment +15%, p < 0.05) and inhibition of the non-oxidative part via transketolase inhibition (Tkt; inhibitor oxythiamine) (co-culture +51% p < 0.001; macrophage pre-treatment +28%, p < 0.01) ([99]Figures 1B and 1D). Moreover, lymphoma cell pre-treatment with oxythiamine increased phagocytic rate significantly (+19%, p < 0.0001) ([100]Figure 1C). Of note, Tkt inhibition induced the highest increase in phagocytic capacity in the co-culture and by pre-treatment of macrophages in the screening approach. Thus, inhibition of glycolysis, AMPK, and mitochondrial ATP production negatively affected macrophages’ phagocytic capacity, while blocking PPP favored lymphoma cell clearance by macrophages. Cross validation of PPP inhibition in macrophages confirms increased ADCP rates To further investigate the PPP in the context of macrophages’ function and as a target for improving immunotherapy, we applied alternative inhibitors (6Pgd: physcion, Tkt: p-hydroxyphenylpyruvate)[101]^13^,[102]^14 and confirmed significant increases in ADCP rates (physcion +26%, p < 0.0001; p-hydroxyphenylpyruvate +25%, p < 0.001) ([103]Figure 2A). Additionally, we recapitulated the phagocytosis assays with the human monocyte cell line THP1 using an alternative antibody (obinutuzumab; anti-CD20 type II), also identifying significant induction of ADCP (6-aminonicotinamide p < 0.05; oxythiamine p < 0.01) ([104]Figure 2B). Figure 2. [105]Figure 2 [106]Open in a new tab Cross validation of PPP inhibition in macrophages confirms increased ADCP rates (A–C) ADCP change compared to basal phagocytosis rate of J774A.1 macrophages under inhibition of PPP. (A) Alternative inhibitor physcion of 6-phosphogluconate dehydrogenase (6Pgd) in oxidative part of PPP (red) and p-hydroxyphenylpyruvate for inhibition of transketolase (Tkt) in non-oxidative part of PPP (blue). (B) Using human monocyte cell line THP1 and CD20 antibody obinutuzumab under inhibition of 6Pgd by 6-aminonicotinamide (red) and inhibition of Tkt by oxythiamine (blue). (C) ADCP assay performed under hypoxic conditions and inhibition of 6Pgd by physcion (red) or inhibition of Tkt by oxythiamine (blue). (D) Antibody-independent cellular phagocytosis of hMB cells by J774A.1 macrophages compared to control under inhibition of 6Pgd by 6-aminonicotinamide (left) and physcion (right). (E) ADCP change compared to basal phagocytosis rate of empty vector control J774A.1 macrophages under shRNA-mediated knockdown of 6Pgd (red) and Tkt (blue). (F) ADCP change compared to basal phagocytosis rate of J774A.1 macrophages under supplementation of metabolites of the PPP. Enzyme reactions in focus colored in violet (6Pgd) and blue (Tkt). E4P, erythrose-4-phosphate; F6P, fructose-6-phosphate; G3P, glyceraldehyde-3-phosphate; Glc6P, glucose-6-phosphate; R5P, ribose-5-phosphate; Ru5P, ribulose-5-phosphate; S7P, sedoheptulose-7-phosphate; X5P, xylulose-5-phosphate. Data are shown as mean ± SEM. Technical replicates (A) n = 30, (B) n = 17–25, (C) n = 25–28, (D) n = 30, (E) n = 20–23, (F) n = 13–20; biological replicates (A) n = 6, (B) n = 4–5, (C) n = 5–6, (D) n = 6, (E) n = 4–5, (F) n = 3–4. p values were calculated using one-way ANOVA. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [107]Figures S1 and [108]S3. Since hypoxia is a functional aspect of the TME in vivo, we also conducted ADCP assays under hypoxic conditions (O[2] 1.5%) and observed significantly increased ADCP rates (physcion +12%, p < 0.01; oxythiamine +20%, p < 0.01) ([109]Figure 2C). To evaluate if PPP inhibition also increases phagocytic capacity of macrophages without the targeting function of antibodies, we assessed antibody-independent cellular phagocytosis (AICP) ([110]Figure 2D) and observed significantly increased AICP rates only by inhibition of the oxidative part of the PPP (6-aminonicotinamide p < 0.01, physcion p < 0.01) ([111]Figure S3D). To abrogate off-target effects of the PPP inhibitors, we generated short hairpin RNA (shRNA) knockdown for the respective enzymes in macrophages ([112]Figures S3P–S3S). Silencing of 6Pgd and Tkt significantly increased macrophages’ ADCP rates (6Pgd +87%, p < 0.0001; Tkt +45%, p < 0.001) ([113]Figure 2E). Altogether, we demonstrate that PPP enzyme inhibition by metabolic inhibitors as well as 6Pgd and Tkt knockdown in macrophages promotes phagocytosis of lymphoma cells and warrants further investigation of the molecular function. Increased phagocytosis is driven by PPP enzyme inhibition and not by PPP metabolite shifting To identify which specific components and metabolites of the PPP directly affect phagocytic function, we performed ADCP assays with supplementation of single educts and products of the PPP ([114]Figure 2F). We observed unaltered ADCP rates using the non-exclusive PPP metabolites glucose-6-phosphate (Glc6P), ribose-5-phosphate (R5P), xylulose-5-phosphate (X5P), glyceraldehyde-3-phosphate (G3P), and fructose-6-phosphate (F6P). In contrast, supplementation of the PPP-exclusive glucose-6-phosphate dehydrogenase (G6pd) product 6-phosphogluconolactone significantly increased ADCP rate (+13%, p < 0.05), as well as the products of 6pgd (ribulose-5-phosphate [Ru5P]; +17%, p < 0.0001) and of Tkt (sedoheptulose-7-phosphate [S7P]; +20%, p < 0.05) ([115]Figure 2F; right panel). In contrast, supplementing the Tkt educt erythrose-4-phosphate (E4P) significantly reduced ADCP rate (−12%, p < 0.01) ([116]Figure 2F; right panel). In conclusion, products of Tkt and 6Pgd promote macrophages’ phagocytic activity while enzyme educts diminish it, indicating that inhibition of the enzymes itself and not a decrease in their products causes increased phagocytic capacity. PPP inhibition induces pro-inflammatory polarization and activation in macrophages To test whether PPP inhibition alters macrophage differentiation and activation, we assessed expression of markers delineating polarization by flow cytometry ([117]Figures 3A and 3B; [118]Table S1). We observed a trend of increased M1-like marker expression and decreased M2-like and TAM marker expression under PPP inhibition and knockdown. Figure 3. [119]Figure 3 [120]Open in a new tab PPP inhibition induces pro-inflammatory polarization and activation in macrophages (A and B) Radar plot of surface marker expression of J774A.1 macrophages. Expression of characteristic surface marker for different macrophage subtypes measured by immunofluorescent staining. Mean fluorescence intensity (MFI) is depicted. To improve readability, high MFI has been downscaled (factor named in brackets next to marker). (A) Compound mediated PPP inhibition. (B) shRNA-mediated PPP knockdown. (C) Immunofluorescent microscopy of J774A.1 macrophages under compound-mediated PPP inhibition and shRNA-mediated PPP knockdown. Blue, phalloidin staining of nucleus; green, actin staining of cytoskeleton. (D–G) Measurement of metabolic activity of J774A.1 macrophages under compound-mediated PPP inhibition by Seahorse analysis. Inhibition of non-oxidative part of PPP by oxythiamine, inhibition of oxidative part of PPP by physcion. (D) One representative example of XF Mito Stress test measurement of ECAR und OCR. (E) Respiratory basal rate and capacity. (F) Glycolytic basal rate and capacity. (G) ATP production. Data are shown in (A and B) as mean of four replicates, in (D) as mean of six replicates in one experiment ±SD, and in (E–G) as mean ± 5–95 percentile. Technical replicates (A and B) n = 4, (D) n = 6, (E–G) n = 18–27; biological replicates (A and B) n = 4, (D) n = 1, (E–G) n = 6–9. p values were calculated using one-way ANOVA, (D) using two-way ANOVA. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [121]Figure S4 and [122]Table S1. To evaluate macrophage morphology, we performed fluorescent microscopy ([123]Figure 3C). Under PPP inhibition the macrophages underwent a profound change in morphology from a round, centered appearance to a spread and outlaying phenotype with filopodia surrounding the cell body. As macrophages’ metabolic status greatly influences their activity and polarization, we assessed glycolytic and mitochondrial activity with the Seahorse XF Mito Stress test ([124]Figures 3D–3G, [125]Figure S4). We observed a significant increase of the oxygen consumption rate (OCR) (p < 0.0001) and extracellular acidification rate (ECAR) (p < 0.0001) ([126]Figure 3D) indicating an increased mitochondrial respiration and glycolytic activity and thus an increased metabolic activity of macrophages. Further analysis identified significant increase of mitochondrial basal activity (physcion p < 0.01; oxythiamine p < 0.001), mitochondrial maximal capacity (physcion p < 0.05; oxythiamine p < 0.01), glycolytic maximal capacity (physcion p < 0.05; oxythiamine p < 0.001), and ATP production (physcion p < 0.05; oxythiamine p < 0.0001) of macrophages ([127]Figures 3E–3G). Taken together, these data show an activation of macrophages by shifted polarization, cytoskeletal reorganization, and increased metabolic activity under PPP inhibition as possible functional basis of increased phagocytosis. PPP inhibition changes the proteomic profile of macrophages toward pro-inflammatory activity To investigate the mediators of increased phagocytic capacity in macrophages, we performed a multi-omics (proteomics, phosphoproteomics, and metabolomics) screening under chemical or shRNA-mediated PPP inhibition. A uniform regulation pattern of proteins involved in macrophage polarization and activation was observed by the use of independent inhibitors and PPP enzyme knockdowns ([128]Figures 4A and 4B, [129]Table S2). Under compound-mediated PPP inhibition, the anti-inflammatory proteins Ptgs1, Sqstm1, and Ybx3[130]^15^,[131]^16^,[132]^17 and the TAM- and M2-typical proteins Hagh and Ezr[133]^18^,[134]^19 were significantly downregulated, while the pro-inflammatory proteins Pam16 and Gosr1 were significantly upregulated ([135]Figure 4A). By using shRNA, an even more pronounced regulation was seen. Negative regulators of cytokine expression and pro-inflammatory signaling were significantly downregulated while promoters of pro-inflammatory activation were significantly upregulated (Atg16l1, Cast, Csf1r, Cybb, Inppl1, Oas3, Parp14, Fkbp5, Ilf2, Tlr7).[136]^20^,[137]^21 Moreover, there was a significant increase in protein expression needed for phagocytosis (Actn1, Actr1a, Iqgap3, Itgav, Lrp1, Myl12a, Necap1, Sh3bp1) ([138]Figure 4B; for phosphoproteomic analysis see [139]Table S3). Figure 4. [140]Figure 4 [141]Open in a new tab PPP inhibition changes the proteomic profile of macrophages towards pro-inflammatory activity (A and B) Volcano plots showing mean change of proteomic transcription under (A) compound-mediated PPP inhibition by 6-aminonicotinamide and oxythiamine compared to untreated J774A.1 macrophages and (B) shRNA-mediated PPP knockdown of 6Pgd and Tkt compared to empty vector control J774A.1 macrophages. Circle size represents number of significantly changed conditions. Red circles: significantly downregulated abundance; green circles: significantly upregulated abundance. Proteins known to participate in immune system are annotated in significant groups. (C and D) Pathway enrichment analysis of (C) proteomics and (D) phosphoproteomics of J774A.1 macrophages under compound-mediated PPP inhibition and shRNA-mediated PPP knockdown. Protein count in listed pathways represented in circle size, mean −log10 p value represented in heatmap analysis. (E and F) Analysis of significantly negative changed protein activity in normalized upstream kinase score (NUKS). (E) Top five most downregulated enzymes in NUKS analysis under shRNA-mediated PPP knockdown of 6Pgd and Tkt and (F) integrative analysis of compound-mediated PPP inhibition by physcion and oxythiamine. (G) Western blot analysis of Ptk2b expression in J774A.1 macrophages under shRNA-mediated PPP knockdown of 6Pgd and Tkt compared to empty vector control. (H) Scheme of hypothesized mechanism leading to pro-inflammatory phenotype of macrophages. In (G) data are shown as mean ± SEM. Technical replicates (A–F) n = 1, (G) n = 5; biological replicates (A–F) n = 3, (G) n = 5. p values in (G) were calculated using one-way ANOVA. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [142]Tables S2–S5. We performed pathway enrichment analysis for functional annotation ([143]Figures 4C and 4D), showing similar enrichment clusters for both compound-mediated and shRNA-mediated inhibition of the oxidative and the non-oxidative part of the PPP. A strong enrichment was seen for immune activity ([144]Figure 4C) including cytokine signaling, antigen processing, and antigen presentation with up to 124 involved proteins and significantly changed phosphorylation patterns ([145]Figure 4D). Moreover, enrichment in proteins relevant to phagocytosis and cytoskeletal organization was observed. In line with our metabolic flux analysis ([146]Figures 3D–3G), we observed great enrichment for proteins influencing mitochondrial and glycolytic activity ([147]Figure 4C). Particularly analysis of phosphoproteomics uncovered a significant enrichment in signaling pathways important for immune signaling (mitogen-activated protein kinase [Mapk]-Erk, Egf/Egfr, p53, Pi3k-Akt) and metabolic regulation (Pi3k-Akt, Hif1a) ([148]Figure 4D). To further analyze the impact of altered protein phosphorylation, we performed an adapted upstream kinase analysis on the basis of integrative inferred kinase activity (INKA) analysis ([149]Table S4).[150]^22 The five most inactivated kinases are displayed ([151]Figure 4E), highlighting the decrease of Hck in the normalized upstream kinase score (NUKS). Hck supports M2-like macrophage polarization, TAM activity, tumor growth, and tumor cell evasion[152]^23 and activates the Csf1 receptor (Csf1r).[153]^24 Csf1r signaling likewise induces M2-like macrophage polarization.[154]^25 Csf1r and its downstream kinase Mapk1 were also one of the five most inactivated kinases ([155]Figure 4E). In combined analysis of PPP inhibitors, the Csf1r downstream kinase Ptk2b (Pyk2) was the most negatively regulated kinase ([156]Figure 4F). Accordingly, a significant downregulation of Ptk2b in PPP knockdown macrophages was observed ([157]Figures 4G and [158]S5A). Furthermore, the most downregulated protein in both knockdown macrophages was Sema4d ([159]Figure 4B), which is an activator of the Ptk2b pathway.[160]^26 Following the Csf1r pathway further downstream ([161]Figure 4H), decreased immune-regulatory gene 1 (Irg1 = Acod1) expression, a major node in immunosuppressive regulation of macrophages, was seen in proteomic analysis ([162]Table S2). Changed Irg1 expression is one possible mechanism leading to altered macrophage activity and phagocytosis.[163]^27 With the exception of Hmox-1, all included signal molecules of the regarded Csf1r pathway were significantly downregulated in proteomic analysis ([164]Figure 4H) ([165]Table S2). PPP inhibition modulates glycogen metabolism and the immune response signaling axis UDPG-Stat1-Irg1-itaconate of macrophages Regarding the critical role of Irg1 on macrophage polarization, we aimed to explore the connection between metabolic modulation, Irg1 regulation, and the resulting macrophage phenotype. PPP and glycogenolysis activity are coupled causing suppression of both pathways if one is inhibited.[166]^28 We quantified glycogen levels identifying significant decreased glycogen amount under all conditions (p < 0.0001, [167]Figure 5A). Glycogen metabolism influences signaling regulating Stat1 activity.[168]^28 Thus, we hypothesized that inhibition of PPP would lead to suppression of glycogenolysis with subsequent decreased uridine diphosphate glucose (UDPG) production and thereby to an inhibition of P2y14 expression with following decreased Stat1 activity. Decreased Stat1 activity leads to less Irf1 and thereby to a decreased Irg1 expression, which possibly leads to functional increasing macrophage activity and phagocytosis.[169]^29^,[170]^30 Figure 5. [171]Figure 5 [172]Open in a new tab PPP inhibition modulates glycogen metabolism and the immune response signaling axis UDPG-Stat1-Irg1-itaconate of macrophages (A) Total glycogen amount in J774A.1 macrophages under compound-mediated PPP inhibition and shRNA-mediated knockdown of 6Pgd and Tkt. (B) Western blot analysis of protein expression of hypothesized connecting pathway in J774A.1 macrophages under shRNA-mediated knockdown of 6Pgd and Tkt compared to empty vector control. Mean expression displayed in bar graph analysis and one representative western blot example. (C) Scheme of working hypothesis of PPP metabolism modulating immune response. (D) Amount of 6pgd product ribulose-5-phosphate and Tkt product sedoheptulose-7-phosphate under shRNA-mediated inhibition of 6pgd and Tkt in J774A1 macrophages. (E) Metabolomic analysis of tricarboxylic acid cycle and citrate metabolism with display of enzyme expression of key enzymes under shRNA-mediated PPP knockdown of 6Pgd and Tkt compared to empty vector control J774A.1 macrophages. Amount of metabolites displayed in box and whiskers. Change in enzyme expression displayed in bar graphs. Genes: succinate dehydrogenase (Sdh), ATP citrate lyase (Acly), immunoregulatory gene 1 (Irg1 = Acod1). (F and G) Cytokine expression under 6Pgd inhibition by 6-aminonicotinamide or physcion and Tkt inhibition by oxythiamine in J774A.1 macrophages. (F) IL-6 expression compared to untreated control. (G) IL-10 expression compared to untreated control. (H) ADCP assay of bone marrow-derived macrophages of Irg1^+/+ wild-type mice and Irg1^−/− knockout mice. Macrophages differentiated out of femoral bone marrow with M-CSF. In (A), (B), and (F–H), bar plots are shown as mean ± SEM; in (D and E) metabolite amount is shown as minimum to maximum and protein expression is shown as calculated −Log2 fold change of control and knockdown macrophages. Technical replicates (A) n = 15, (B) n = 3–6, (D) n = 3, (E) n = 3, (F and G) n = 9–18, (H) n = 35; biological replicates (A) n = 3, (B) n = 3–6, (D) n = 3, (E) n = 3, (F and G) n = 3–6, (H) n = 7. p values were calculated in (A, B, D, and E) using one-way ANOVA, protein expression in (E) using student’s t test, and in (F–H) using unpaired t test. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [173]Figures S4 and [174]S5 and [175]Tables S6 and [176]S7. To validate this hypothesis, we performed western blot analysis of the hypothesized pathway-associated proteins and identified significant reductions in expression (p < 0.0001, [177]Figure 5B) with the highest decline of Irg1 amount (>80%, p < 0.0001, [178]Figure S5). The hypothesized pathway linking PPP activity and Irg1 expression is displayed in [179]Figure 5C. As Irg1 acts as a metabolic enzyme and changes in immune activity are driven by its product itaconate, we investigated the connection between metabolism and enzyme expression by metabolomic assessment ([180]Tables S5 and [181]S6) and compared it to changes in enzyme expression of interest detected in proteomics ([182]Table S2) ([183]Figures 5D and [184]S4C). The metabolomic screening[185]^31 confirmed a significantly decreased amount of exclusive 6Pgd and Tkt products ribulose-5-phosphate and sedoheptulose-7-phosphate (p < 0.0001, [186]Figure 5D) in 6pgd and Tkt knockdown macrophages, while drug-mediated inhibition did not show a significantly decreased amount ([187]Figure S4C). In line with the hypothesized pathway, a significant downregulation of Irg1 was observed under PPP inhibition (p < 0.01) with subsequent significantly decreased amount of itaconate (p < 0.0001) ([188]Figure 5E). Itaconate is an inhibitor of succinate dehydrogenase (Sdh). Accordingly, there was a significant decrease of the Sdh educt succinate (p < 0.001) and a significant increase of the Sdh product malate (p < 0.0001) ([189]Figure 5E). This indicates less suppression of Sdh due to decreased itaconate production by which mitochondrial oxidative activity is increased as observed in the metabolic flux analysis ([190]Figures 3F and 3G). Besides the use of citrate for itaconate production, citrate is also an educt of the ATP citrate lyase (Acly) for acetyl-coenzyme A production. It has been shown that Acly activity is an inducer of macrophage activation and supports pro-inflammatory cytokine production—for example interleukin (IL)-6—in macrophages.[191]^32 In proteomic analysis a significant increase of Acly expression under PPP inhibition was observed (p < 0.01, [192]Figure 5E). Itaconate is widely known as a regulatory immunosuppressive metabolite in macrophages, which promotes anti-inflammatory IL-10 secretion and inhibits pro-inflammatory IL-6 secretion.[193]^27 Moreover, we observed significantly increased nuclear factor κB1 expression under PPP inhibition and knockdown ([194]Table S2), an activator of IL-6 and inhibitor of IL-10 production. In line with these findings, we observed significantly increased IL-6 secretion (p < 0.05, [195]Figure 5F) and significantly decreased IL-10 secretion (p < 0.0001, [196]Figure 5G) by PPP inhibition. To further prove the functional role of the Irg1-itaconate pathway, we evaluated the phagocytic activity of macrophages of Irg1^−/− knockout mice. A significantly increased phagocytic activity of bone marrow-derived macrophages in ADCP assay ex vivo was observed in comparison to Irg1^+/+ wild-type mice (+34%, p < 0.05, [197]Figure 5H). In conclusion, we connected metabolic activity and immune regulation in macrophages via the UDPG-Stat1-Irg1-itaconate signaling axis provoked by PPP activity. Irg1 downregulation increases macrophage activation via itaconate reduction with subsequent metabolic activation and a pro-inflammatory shift in cytokine secretion. This also leads in vivo to an increased phagocytic capacity of macrophages. PPP inhibition in primary human cells increases phagocytic capacity of macrophages and decreases their bystander function To translate our findings into human context, we isolated primary human monocytes from healthy donors and differentiated them into macrophages by macrophage colony stimulating factor (M-CSF) under PPP inhibition. After testing cytotoxicity of the PPP inhibitors to primary human macrophages, ADCP assays with non-toxic concentrations of inhibitors were performed. Inhibition of both parts of the PPP significantly increased ADCP rates (physcion +64%; oxythiamine +92%, p < 0.0001, [198]Figures 6A and 6B). The human macrophages showed a similar switch toward pro-inflammatory cytokine secretion with significantly increased IL-6 (p < 0.05) and significantly decreased IL-10 secretion (p < 0.0001) ([199]Figures 6C and 6D). Figure 6. [200]Figure 6 [201]Open in a new tab PPP inhibition in primary human cells increases phagocytic capacity of macrophages and decreases their tumor-supportive bystander function (A and B) ADCP change compared to basal phagocytosis rate of human monocyte-derived macrophages. (A) ADCP change of monocyte-derived macrophages differentiated in the presence of physcion and M-CSF, (B) ADCP change of monocyte-derived macrophages differentiated in the presence of oxythiamine and M-CSF. (C and D) Cytokine expression of monocyte-derived macrophages differentiated in the presence of oxythiamine and M-CSF. (C) IL-6 expression, (D) IL-10 expression. (E and F) ADCP change of J774A.1 macrophages phagocyting primary CLL patient cells compared to basal phagocytosis rate. (E) ADCP change under compound-mediated PPP inhibition. (F) ADCP change under shRNA-mediated PPP knockdown. (G) ADCP change of monocyte-derived macrophages differentiated in the presence of oxythiamine and M-CSF phagocyting primary CLL patient cells compared to basal phagocytosis rate. (H) Viability of primary CLL patient cells after incubation with PPP inhibitors physcion or oxythiamine in mono-culture and in co-culture with J774A.1 macrophages. In co-culture setting, cells were treated in parallel or macrophages were pre-treated before onset of co-culture. (I–L) Half maximal inhibitory concentration (IC[50]) for individual primary CLL patient cell samples to bendamustine treatment compared to control. Cells were incubated with bendamustine after protective macrophage co-culture with untreated J774A.1 macrophages vs. PPP inhibition. (I and J) Inhibition of 6Pgd in oxidative part of PPP by physcion, (I) co-culture treatment, (J) macrophage pre-treatment. (K and L) Inhibition of Tkt in non-oxidative part of PPP by oxythiamine, (K) co-culture treatment, (L) macrophage pre-treatment. In (A–H) data are shown as mean ± SEM, in (I–L) as minimum to maximum. Technical replicates (A) n = 28, (B) n = 20, (C and D) n = 18, (E and F) n = 20, (G) n = 65, (H) n = 30, (I–L) n = 30; biological replicates (A) n = 6, (B) n = 4, (C and D) n = 6, (E and F) n = 5, (G) n = 12, (H) n = 10, (I–L) n = 10. p values were calculated in (A–G) using one-way ANOVA, in (H) using repeated measures (RM) one-way ANOVA, and in (I–L) using paired t test. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [202]Figure S6. To address effector function of macrophages in the context of primary human leukemia cells, primary chronic lymphocytic leukemia (CLL) cells of five individual patients were used for ADCP assays. A significant increase of phagocytosis was observed under PPP inhibition (+22%, p < 0.001, [203]Figure 6E) and by using knockdown macrophages (Tkt +60%; 6Pgd +92%, p < 0.0001, [204]Figure 6F) ([205]Figures S6C and S6D). To evaluate phagocytosis in a fully human setting, we performed ADCP assay with primary human monocyte-derived macrophages differentiated in the presence of PPP inhibitors and primary CLL patient cells (12 individual patients). A significantly increased phagocytic capacity was observed (+24%, p < 0.0001, [206]Figures 6G and [207]S6E). Thereby, we have demonstrated that primary indolent lymphoma and primary human macrophages are also affected by PPP modulation. Beyond the inefficient phagocytic function, TAMs exert direct supportive effects on tumor cells. CLL cells depend on macrophages as “nurse-like” bystander cells to survive.[208]^33 Macrophages in the microenvironment of CLL are polarized toward tumor-promoting TAMs and support CLL cells by chemokine secretion and immunosuppressive signaling. We therefore evaluated the effect of PPP-inhibited macrophages on primary CLL cells. Interestingly, PPP inhibition in mono-cultured primary CLL cells decreased their viability significantly (p < 0.0001, [209]Figure 6H, left), as well as inhibition of the non-oxidative part of the PPP in co-culture (p < 0.01, [210]Figure 6H, right). As TAMs are also important mediators in chemotherapy resistance, we evaluated if the co-cultivation under PPP inhibition affects the susceptibility of primary CLL cells toward apoptosis by chemotherapy. We observed significantly increased bendamustine-induced apoptosis among primary CLL cells under inhibition of both parts of the PPP (p < 0.001, [211]Figures 6I–6L) (for individual patient data see [212]Figures S6F–S6J). This boost in apoptosis was achieved by PPP inhibition in co-culture (p < 0.001, [213]Figures 6I and 6K) and by macrophages pre-treatment before exposing them to primary CLL cells (p < 0.01, [214]Figures 6J and 6L). These observations underline the role of altered macrophage support under PPP inhibition in the TME such as direct leukemia cell support or resistance to chemotherapy. PPP inhibition increases macrophages’ maturation and pro-inflammatory polarization in vivo To evaluate if PPP inhibition preserves its effect on macrophages in vivo, we treated C57BL/6J mice with the PPP inhibitor S3 (1-hydroxy-8-methoxy-anthraquinone). S3 is a more stable derivate of the 6Pgd inhibitor physcion.[215]^13 Investigating the myelopoiesis under PPP inhibition, a significant increase of cells in the LSK compartment (Lin^−, Sca-1^+, c-Kit^+) was seen (p < 0.0001, [216]Figure 7A). PPP inhibition significantly increased the amount of hematopoietic stem cells (HSCs; p < 0.05, [217]Figure 7B) and multipotent progenitor (MPP) cells ([218]Figure 7B), including MPP pools known to fuel the myeloid compartment (MPP2 p < 0.001; MPP3 p < 0.01; MPP5 p < 0.01, [219]Figure 7B).[220]^34^,[221]^35 This coincided with a significantly increased frequency of myeloid progenitor cells (p < 0.01, [222]Figure 7C) and macrophages (p < 0.001, [223]Figure 7C) in the bone marrow, indicating propelled myelopoiesis. Figure 7. [224]Figure 7 [225]Open in a new tab PPP inhibition increases myelopoiesis, macrophages’ maturation, and pro-inflammatory polarization in vivo and boosts anti-leukemic treatment response in an aggressive humanized lymphoma mouse model (A) Progenitor cell compartment LSK (Lin^−, Sca-1^+, c-Kit^+) in bone marrow of C57BL/6J mice treated with vehicle (control) or PPP inhibitor S3 i.p. for 7 days. (B) Multipotent progenitor (MPP) subsets in bone marrow of C57BL/6J mice treated with vehicle (control) or PPP inhibitor S3 intraperitoneally (i.p.) for 7 days. HSC (CD34^−, CD48^−, CD150^+, CD135^-), MPP1 (CD34^+, CD48^−, CD150^+, CD135^-), MPP2 (CD34^+, CD48^+, CD150^+, CD135^-), MPP3 (CD34^+, CD48^+, CD150^-, CD135^-), MPP4 (CD34^+, CD48^+, CD150^-, CD135^+), MPP5 (CD34^+, CD48^−, CD150^-, CD135^-). (C) Percentage of myeloid lineage cells of whole cell amount in bone marrow of NSG mice transfected with hMB cells, treated with vehicle or PPP inhibitor S3 i.p. for 12 days, and euthanized on day 15. Common myeloid progenitor cells (CD41^+, CD34^+), monocytes (Ly6c^+, CX3CR1^+), macrophages (F4/80^+, CD64^+). (D) Expression of characteristic surface marker for different macrophage subtypes on peritoneal macrophages measured by immunofluorescent staining. Mean fluorescence intensity (MFI) is depicted. To improve readability, high MFI has been downscaled (factor named in brackets next to marker). C57BL/6J mice treated with vehicle (control) or PPP inhibitor S3 i.p. for 7 days. (E) ADCP assay of bone marrow-derived macrophages. C57BL/6J mice treated with vehicle (control) or PPP inhibitor S3 i.p. for 7 days, macrophages differentiated out of femoral bone marrow with M-CSF. (F) Survival curve of aggressive lymphoma (hMB) bearing mice treated with PPP inhibitor S3 +/− therapeutic antibody alemtuzumab. (G) One representative example of immunohistochemical staining of hMB cells (CD19^+) and macrophages (CD68^+) in spleen of aggressive lymphoma (hMB) bearing mice treated with vehicle or alemtuzumab + S3. In (A–C and E) data are shown as mean ± SEM, in (D) data are shown as mean of ten replicates. Technical replicates (A and B) n = 12, (C) n = 3, (D) n = 9–10, (E) n = 70–75, (F) n = 21–25, (G) n = 4; biological replicates (A and B) n = 12, (C) n = 3, (D) n = 9–10, (E) n = 14–15, (F) n = 21–25, (G) n = 4. p values were calculated in (A–E) using unpaired t test and in (F) using Benjamini-Hochberg test. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See also [226]Figure S7 and [227]Table S8. We further investigated the polarization of in vivo macrophages ([228]Table S8) and detected a shift away from M2-like subtype with significantly decreased Arg1 expression (p < 0.05) and toward pro-phagocytic M1-like subtype with significantly increased CD38 expression (p < 0.05) in unstimulated peritoneal macrophages ([229]Figure 7D) (for other compartments see [230]Figure S7A). These findings are in line with our in vitro observations. Moreover, in vivo PPP inhibition significantly increased phagocytic activity of bone marrow-derived macrophages in ADCP assay ex vivo (+74%, p < 0.001, [231]Figure 7E). Altogether, we have shown that PPP inhibition in vivo activates macrophages’ inflammatory polarization and maturation as well as their phagocytic capacity, which increases their anti-tumor function in vivo. PPP inhibition boosts anti-leukemic treatment and thereby prolongs survival in an aggressive lymphoma mouse model To focus on the therapeutic effect of PPP inhibition in vivo, we evaluated treatment effects in an aggressive lymphoma mouse model. We used the humanized double-hit lymphoma mouse model (hMB),[232]^36 which is amenable for modeling human-specific antibody therapy. We treated the mice with the therapeutic antibody alemtuzumab and the PPP inhibitor S3. As the lymphoma reflects aggressive disease, untreated mice died rapidly after tumor cell injection (median overall survival [mOS] 22 days, [233]Figure 7F). By treatment with S3 only, this rapid tumor progression persisted. As shown in our previous work, treatment with alemtuzumab increases survival significantly in this aggressive lymphoma mouse model[234]^5 (mOS 25 days, p = 1.4e−05, [235]Figures 7F and [236]S7B). By adding the PPP inhibitor S3 to alemtuzumab, an additional significant prolongation of mouse survival was achieved in comparison to antibody treatment only (mOS 27 days, p = 0.0059, [237]Figures 7F and [238]S7B) with a stable increased number at risk up to day 25 (survival of 88%, [239]Figure 7F). Immunohistochemical analysis of spleens showed a marked reduction of CD19^+ lymphoma cell infiltration with concomitant increase of CD68^+ macrophage infiltration after treatment with alemtuzumab and S3 in comparison to vehicle control ([240]Figures 7G and [241]S7C). We finally demonstrated in vivo that PPP inhibition in the context of a highly aggressive lymphoma model increases the efficacy of antibody therapy to prolong overall survival significantly. Discussion TAMs are key drivers in various cancers associated with poor outcome and diminished efficacy of immunotherapies.[242]^1^,[243]^3^,[244]^5 The influence of glucose and mitochondrial metabolism on macrophages’ polarization and activity has been established.[245]^8^,[246]^9^,[247]^37 Activation of the PPP in macrophages has been implicated in immune tolerance and granuloma formation.[248]^38^,[249]^39 However, no functional implications of the PPP in TAM regulation have been established nor the effects of PPP inhibition on immune regulation have been identified. Here, we show that modulation of the PPP in TAMs serves as a robust regulator of phagocytic function and macrophage activity and prolongs survival in aggressive B cell lymphoma therapy. Metabolic inhibition screening in lymphoma-macrophage co-cultures emphasized detrimental effects on macrophage function for the majority of investigated pathways. Only inhibition of the PPP showed a significant increase of phagocytosis with a synergistic effect on effector and target cells. This was true across various in vitro and in vivo macrophage model systems of both murine and human descent by compound and genetic targeting. Even though the time-dependent effects of PPP inhibition differ between chemical compounds and genetic targeting, we observed similar phenotype under compound-mediated as well as shRNA-mediated PPP inhibition. The increased phagocytosis rate appeared also under hypoxic conditions as an approximation of physiological status of therapy-refractory niches of lymphoma—the lymph nodes and bone marrow[250]^40^,[251]^41—indicating therapeutic efficacy by metabolic inhibition in contrast to other therapy modalities in these niches. Previous reports demonstrated reduced cancer and leukemia cell growth in mice upon PPP inhibition.[252]^13^,[253]^42 Especially in CLL, macrophages play a pivotal role as supportive bystander cells in the TME, without which CLL cells would undergo spontaneous apoptosis.[254]^33 We have shown that PPP inhibition diminishes this pro-survival bystander function of macrophages and acts as a sensitizer to genotoxic regimens. Alterations of the PPP enzymes 6PGD and TKT have been previously described in many cancer types.[255]^43^,[256]^44^,[257]^45^,[258]^46^,[259]^47 Overexpression of TKT was closely associated with aggressive hepatocellular carcinoma features,[260]^48 and 6PGD was shown to promote metastasis,[261]^49 while suppression of 6Pgd attenuates cell proliferation and tumor growth[262]^42 and overcomes cisplatin resistance[263]^50 and Tkt inhibition sensitizes cancer cells to targeted therapy and reduces growth of metastatic lesions.[264]^48 PPP inhibition by physcion, S3, or 6-aminonicotinamide has demonstrated anti-tumorigenic effects in several solid tumor types and chemotherapeutic-resistant acute myeloid leukemia cells,[265]^47^,[266]^51^,[267]^52 without affecting non-malignant cells.[268]^42 Moreover, 6Pgd inhibition in CD8^+ T cells led to an increased effector function with higher tumoricidal activity.[269]^53 We have previously shown that macrophage effector polarization is crucial in therapeutic antibody-based regimens of B cell lymphoma and can be modulated.[270]^54^,[271]^55^,[272]^56 We now identified macrophage metabolism as an essential switch of macrophage effector function in lymphoma. We demonstrated that increase of phagocytosis is driven by PPP enzyme inhibition, rather than metabolite accumulation. Non-exclusive PPP metabolites did not influence phagocytosis, possibly due to degradation via glycolysis (G3P, F6P) or nucleotide synthesis (R5P) before entering PPP flux. In contrast, exclusive PPP metabolites altered phagocytosis activity: supplementation of the G6pd product 6-phosphogluconate, the 6Pgd product ribulose-5-phosphate, and the Tkt product sedoheptulose-7-phosphate increased phagocytosis, while supplementation of the Tkt educt erythrose-4-phosphate decreased phagocytic activity in macrophages. This points to a feedback inhibition of the respective PPP enzymes and emphasizes the enzyme inhibition as driving force for increased phagocytosis. PPP is a central linker between glucose metabolism, amino acid biosynthesis, fatty acid metabolism, and redox homeostasis.[273]^43 A gain in metabolic activity was observed under PPP inhibition with increased glycolytic and mitochondrial capacity causing enhanced ATP production, fueling macrophages’ activation. An increase of glycolysis is well described within the phenotypical switch to pro-inflammatory macrophages.[274]^57 We observed profound alteration of morphology and macrophage polarity, demonstrated by a decrease of markers associated with M2-like macrophages and TAMs, which represent immunosuppressive and tumor-promoting macrophage subtypes,[275]^3^,[276]^58^,[277]^59 while exclusive M1 marker, expressed on pro-inflammatory macrophages, was increased.[278]^60^,[279]^61^,[280]^62^,[281]^63 In total, the restriction of one metabolic pathway—the PPP—gives rise to numerous paths of activation, which renders a profound alteration of phenotype and particular phagocytic activity in macrophages. Thereby the anti-tumor function could be improved from independent directions. Our detailed multi-omics and functional analysis provides evidence that these effects are directly related to 6Pgd and Tkt enzyme activity loss, which polarizes macrophages to a pro-inflammatory phenotype through downregulation of Stat1 and Irg1. The functional switch between PPP enzyme activity and subsequent polarization program is Csf1r expression and activity of glycogen metabolism. Csf1r activation induces Hmox-1 expression,[282]^64^,[283]^65 which induces Irg1 expression,[284]^66 a central inhibitory regulator of macrophage activation.[285]^27 We demonstrated significant downregulation of Csf1r pathway proteins. Via Csf1r signaling, macrophages are polarized toward an M2-like or TAM phenotype by directly activating Erk1/2 (Mapk1/2) and Hck signaling.[286]^24^,[287]^25^,[288]^67^,[289]^68^,[290]^69 Both, Mapk1 and Hck activity was shown to be decreased under PPP inhibition in upstream kinase analysis. Considering the relevant role of Csf1r in macrophage ontogeny, activation, and polarization, reduced Csf1r expression might be responsible for macrophage activation under PPP inhibition. Therefore, Csf1r blockade might be a promising strategy to increase macrophage activity in the context of tumor therapy. Several CSF1R inhibitors are currently under clinical investigation.[291]^70 Nevertheless, as CSF1R is a macrophage-exclusive receptor, only a macrophage-exclusive effect could be achieved by using CSF1R inhibition, in contrast to the previously described multi-cellular effects of 6PGD and TKT inhibition. PPP inhibition has been functionally linked to inhibition of glycogenolysis causing decreased UDPG production.[292]^28 UDPG as a signaling molecule activates the P2y14 receptor (P2y14r). P2y14r activation in turn increases Stat1 expression and Mapk1 phosphorylation.[293]^71 Considering that Stat1 is a major regulator of Irf1 expression,[294]^29 which is the transcription factor of Irg1,[295]^72 the signaling cascade induced by PPP inhibition in macrophages is causing an altered Irg1 expression. Irg1 is almost exclusively expressed in activated immune cells and a key driver of immune inhibition via itaconate production.[296]^27 Itaconate inhibits glycolysis and mitochondrial activity by Sdh inhibition[297]^73 and promotes anti-inflammatory macrophage phenotype[298]^74 and tumor growth by increased reactive oxygen species secretion by TAMs.[299]^75 In contrast to the publication of Ma et al.,[300]^28 we observed a decreased amount of glycogen under PPP inhibition, but simultaneously increased glycolysis as a possible indicator for a shift of glucose processing causing decreased glycogen synthesis and glycogenolysis. Subsequently, we observed decreased expression of all UDPG-Stat1-Irg1-itaconate pathway proteins under PPP inhibition, particularly lower amounts of itaconate and an increased Sdh and Acly activity. Acly activity is known to induce macrophage activation and pro-inflammatory cytokine production.[301]^32 These changes subsequently resulted in a pro-inflammatory cytokine switch, phenotypic shift toward M1-like macrophages, and diminished primary leukemia cell support. The metabolic alterations observed here support the hypothesis of the connection between PPP inhibition, itaconate abundance, and immune regulation. Furthermore, the transcription factor Irf1 induces inducible nitric oxide synthase (iNos) expression,[302]^72 consistent with the observed decreased iNos expression under PPP inhibition. High iNos expression and activity have been correlated with malignancy and poor survival in several solid tumors and leukemia.[303]^76 Despite the pro-inflammatory transcriptional function of Stat1 in LPS-stimulated macrophages, we show an alternative mechanism of macrophage activation by metabolic depression of the anti-inflammatory properties of Stat1 via itaconate regulation leading to a pro-phagocytic phenotype of macrophages. In TAMs, Stat1 has been shown to be the generator of the blended M1/M2 phenotype and supporter of the anti-inflammatory and pro-tumorigenic properties.[304]^77^,[305]^78 As reduced expression of Irf1 also influences Stat1 and Csf1r expression,[306]^79 Irf1 appears as the junction between all recapitulated pathways, which are leading to the changed macrophage activity and polarization. Taken together, these signaling connections show a narrow network, which links PPP inhibition and immune regulation in macrophages and cancer cells. We demonstrated a highly significant increase of phagocytosis in primary human cells, indicating efficacy for potential clinical use. To model lymphoma in patients, we performed in vivo experiments with the PPP inhibitor S3, whose low toxicity and high effectiveness in treatment of other tumor entities were demonstrated before.[307]^13^,[308]^42 In the aggressive lymphoma mouse model,[309]^36 PPP inhibition has an amplification effect on antibody therapy leading to significant prolonged overall survival in comparison to antibody treatment only with associated increased macrophage lymphoma infiltration. PPP inhibition in vivo increased myelopoiesis and gave rise to progenitor cell expansion, indicating increased provision of a variety of immune cells. Macrophages displayed pro-inflammatory polarization and significantly increased phagocytic capacity after PPP inhibition in vivo. We have proven in vivo the efficacy of PPP inhibition leading to macrophage activation and improving therapy response by antibody-mediated phagocytic clearance of lymphoma with prolongation of overall survival. In conclusion, PPP inhibition may serve as immune-modulatory therapy repolarizing macrophages. We demonstrated metabolic modulation as a key mechanism of macrophage regulation. PPP inhibition causes diminished Irg1 expression leading to reduced anti-inflammatory itaconate production. Our work indicates PPP inhibition as a dual-principle therapy targeting cancer cells and their immune-microenvironment simultaneously, with implications for cancer treatment, especially in the context of antibody-based regimens. Limitations of the study This study has several important limitations to consider. The screening process was conducted primarily from the perspective of ADCP and emphasized a therapeutic angle, which may have limited the scope of the findings. While the PPP was a focus, its role in modulating lymphoma progression requires further investigation to fully understand its impact on the TME. The study concentrated on macrophages as targets for PPP inhibition. The effects of this inhibition on other immunotherapeutic approaches, such as bispecific antibodies and CAR-T cells, remain to be demonstrated. Crucially, the role of PPP inhibition in human patients has yet to be established through clinical trials. These limitations highlight areas for future research to build upon this study’s results and provide a more comprehensive understanding of PPP inhibition in lymphoma treatment. Resource availability Lead contact Further requests for information should be directed to and will be fulfilled by the lead contact, Christian P. Pallasch (christian.pallasch@uk-koeln.de). Materials availability This study did not generate new unique reagents. Data and code availability * • Proteomic and phosphoproteomic data have been deposited at PRIDE:PXD042428. Metabolomic data have been deposited at UCSD Metabolomics Workbench: ST003516. * • The data are publicly available as of the date of publication. Accession numbers are listed in the [310]key resources table. * • This paper does not report original code. * • Any additional information required to reanalyze the data reported in this paper is available from the [311]lead contact upon request. Acknowledgments