Fig 5 - uploaded by Heiko Bruns
Content may be subject to copyright.
Lenalidomide restores the vitamin D pathway in vivo. a Monocytes of MM patients (untreated: black bars, treated with IMiD: red bars, treated with proteasome inhibitors: white bars) were isolated from PBMC by positive selection of CD14+ cells using a MACS system and mRNA expression of VDR, CYP27B1, and 

Lenalidomide restores the vitamin D pathway in vivo. a Monocytes of MM patients (untreated: black bars, treated with IMiD: red bars, treated with proteasome inhibitors: white bars) were isolated from PBMC by positive selection of CD14+ cells using a MACS system and mRNA expression of VDR, CYP27B1, and 

Source publication
Article
Full-text available
Macrophages are key mediators of the therapeutic effects exerted by monoclonal antibodies, such as the anti-CD38 antibody MOR202, currently introduced in multiple myeloma (MM) therapy. Therefore, it is important to understand how antibody-mediated effector functions of myeloma-associated macrophages (MAMs) are regulated. Here, we focused on the eff...

Contexts in source publication

Context 1
... vitamin D acts not only on effector cells, but also on target cells, we reasoned that vitamin D treatment of myeloma cells might increase their vulnerability against monoclonal antibodies. VDR functions as a ligand- inducible transcription factor. Upon binding to its ligand 1,25D3, the VDR-1,25D3 complex binds to DNA sequences called vitamin D response elements located in regulatory regions of target genes leading to either increased or decreased transcription. We ana- lyzed chromatin immunoprecipitation sequencing (CHIP-Seq) data from human transformed B-cells (cellline GM10861) [20] stimulated with 1,25D3 and found high levels of VDR bound to the promotor region of genes coding for SDC1 (CD138), CD38, SLAMF7, and CD47 after treatment with 1,25D3 (Fig. 3a). These results were confirmed by ChIP with a VDR antibody (Supplemental Fig. 3). Therefore, we speculated that vitamin D modulates the protein expression of these molecules on MM cells. We treated MM cell lines with the active form of vitamin D and analyzed the protein expression by flow cytometry. In fact, we found sig- nificantly higher expression of CD38 on the surface of MM cell lines after 48 h (RPMI8226: p = 0.0005; OPM2: p = 0.0064; MM1S: p = 0.001) ( Fig. 3b and Supplemental Fig. 4a), but no differences in the expression of CD138, SLAMF7, and CD47. These results were confirmed on the mRNA level by qPCR (Supplemental Fig. 4b). Of note, we did not observe any direct cytotoxic effects of 1,25D3 in our experimental settings (Supplemental Fig. 5a, b). Next, we examined whether vitamin D increases the expression of CD38 on primary MM cells of treatment naïve patients. There- fore, we treated BM samples of untreated MM (n = 6) patients for 48 h with 1,25D3 and analyzed the CD38 expression on MM cells (CD45 low , CD19 low , and CD138 high ) by flow cytometry. Similar to the MM cell lines, 1,25D3 exposure resulted in significant (p = 0.007) induction of CD38 expression on the surface of primary MM cells (Fig. 3c). However, we found no significant effect of 1,25D3 on the CD38 expression on normal hematopoietic cells (Supplemental Fig. 6). Since efficacy of monoclonal antibodies is partly dependent on target antigen expression, we wondered if MOR202 has a higher propensity to bind to the surface of MM cells after treatment with 1,25D3. Therefore, we labeled MOR202 with a fluorescence dye (Alexa-Fluor-647) and analyzed the binding capacity of the antibody to myeloma cells by confocal microscopy. In line with the previously observed increased CD38 expression, we found enhanced binding of MOR202 to the surface of MM cells after incubation with 1,25D3 (Fig. 3d). These results indicate that the active form of vitamin D not only acts on effector cells but also increases the vul- nerability toward anti-CD38 antibodies by enhancing CD38 expression on MM cells. Fig. 2 Lenalidomide restores the vitamin D pathway in human macro- phages. a Generated macrophages were treated for 72 h with several anti- myeloma drugs at the following concentrations: dexamethasone (25 µM), thalidomide (1 µM), lenalidomide (1 µM), pomalidomide (1 µM), velcade (10 nM), carfilzomib (10 nM), melphalan (10 nM), and prednisolone (50 µM). mRNA expression of VDR, CYP27B1, and CYP24A1 were ana- lyzed by qPCR. b Immunofluorescence analysis of VDR (left panel, green), CYP27B1 (middle panel, green), and CYP24A1 (right panel, green) protein expression of untreated (upper panel) or lenalidomide (1 µM, 72 h)-treated macrophages (lower panel) (630×). Scale bar: 20 µm. c Macrophages (n = 5) were treated with (black) or without lenalidomide (green) for 72 h and incubated with vitamin D-sufficient serum (25D3 = 100 nM) for 24 h. 1,25D3 conversion was measured in cell lysates by ultra-high-performance liquid chromatography tandem mass spectro- metry (UHPLC-MS/MS). d Graph shows the relative intensity (%) of 25D3 (black bars) and 1,25D3 (red bars) measured in lenalidomide- treated macrophages (n = 6) by LC-MS/MS Fig. 3 Vitamin D regulates CD38 expression on MM cells. a ChIP-Seq analysis of VDR binding to the promotor sequence of the genes encoding CD138, CD38, SLAMF7, and CD47 in human transformed B-cells (cellline GM10861) treated for 36 h with and without 1,25D3 (100 nM). ChIP-Seq traces were generated from GSE22484 (Rama- gopalan et al. [20]). Black bars represent the VDR binding side. Exons are presented as vertical bars, the untranslated region is represented by a black line, and arrows indicate the direction of transcription. b Myeloma cell lines (as indicated) were treated with 1,25D3 (10 nM; red bars) or left untreated (black bars) and the protein expression of CD138, CD38, SLAMF7, and CD47 were measured by flow cyto- metry. Presented in the graph are the x-fold-changes of the MFI of the test condition (+1,25D3) to the corresponding MFI of the medium control ± SEM. c Bone marrow of untreated myeloma patients were treated for 48 h with 1,25D3 (10 nM) and CD38 expression as MFI was analyzed on MM cells (CD19−/CD138+) by flow cytometry (n = 6). d Myeloma cell lines (as indicated) were treated with and without 1,25D3 (10 nM) for 48 h and incubated with Alexa-647- labeled MOR202 (red), and analyzed by confocal laser microscopy. Scale bar: 10 ...
Context 2
... mentioned previously lenalidomide increases the expression of CYP27B1 in macrophages. In fact, this step is required for lenalidomide to spark the beneficial effects of vitamin D in terms of improving the antibody-dependent activity of macrophages against MM cells. To assess whe- ther lenalidomide therapy impacted the vitamin D pathway in vivo, we initially analyzed isolated monocytes from untreated MM patients and from IMiD-treated patients (for patient characteristics, please refer to Supplemental Fig. 4 MOR202-dependent elimination of MM cells is enhanced by lenalidomide and 25D3. a Generated macrophages were coincubated with VPD-labeled myeloma cell lines (as indicated) in different effector target ratios (1:1, 5:1, and 10:1) in the presence or absence of MOR202 (10 µg/ml) for 24 h. The number and the viability of mye- loma cells were measured by flow cytometry in the presence of 123count eBeads to determine absolute numbers of surviving MM cells. The percentage of MOR202-mediated killing was then calcu- lated using the following formula: MOR202-mediated killing = 100- ((absolute number of surviving MM cells in the presence of MOR202/ absolute number of surviving MM cells in the presence of IgG1) × 100%). b VPD-labeled MM target cells (as indicated) were added to lenalidomide-treated macrophage effectors (1 µM, 72 h) at E/T ratios of 1:1 with or without 25D3 (≈10 nM) in the presence or absence of MOR202 (10 µg/ml). After 24 h, surviving MM cells were enumerated by flow cytometric analysis of CD11b−/VPD+ cells in the presence of cell count beads. Percentage of MOR202-mediated killing was then calculated as described above. c CPD-labeled MM target cells (as indicated, red) were added to lenalidomide-treated macrophage (CFSE stained, green) effectors at E/T ratios of 1:1 with or without 25D3 in the presence or absence of MOR202. After 2 h phagocytosis was analyzed by confocal microscope at (630×). Scale bar: 20 µm. d Macrophages were treated with lenalidomide (1 µM) for 48 h with or without 25D3 (≈10 nM). After 24 h of incubation, macrophages were transfected with control siRNA or siRNA against CYP27B1. Trans- fected and non-transfected macrophages were co-incubated with stained OPM2 targets (E:T 1:1) in the presence or absence of MOR202 (10 µg/ml) for 24 h. The number and the viability of myeloma cells were measured by flow cytometry in the presence of 123count eBeads to determine absolute numbers of viable MM cells Table 3). The qPCR analyses revealed that monocytes displayed a significantly increased CYP27B1mRNA expression during IMiD therapy in opposition to monocytes from patients without treatment (Fig. 5a) (CYP27B1 in untreated: 0.05 ± 0.05 A.U. vs. CYP27B1 in IMiD-treated: 0.83 ± 0.3 A.U., p = 0.002). In addition, in monocytes from proteasome inhibitor-treated patients CYP27B1 mRNA expression remained, on average, unchanged. To directly examine whether lenalidomide regulates the CYP27B1 expression in MAMs, we longitudinally examined BM specimens of individual MM patients before and during lenalidomide therapy (n = 5) simultaneously for CYP27B1 and CD68 (for patient characteristics, please refer to Sup- plemental Table 4). Whereas a considerable number of MAMs expressed CYP27B1 during lenalidomide treatment, we found only low levels of CYP27B1 in MAMs in the untreated MM patient cohort (Fig. 5b). Noticeably, overall CYP27B1 tissue expression was found enhanced indicating a rather general (lenalidomide-triggered) phenomenon that is not only restricted to macrophages. Taken together, these results indicate that monocytes and MAMs have an enhanced expression of CYP27B1 under lenalidomide therapy, implicating that these macrophages may possess increased tumoricidal effector mechanisms, which obviously requires further ...
Context 3
... mentioned previously lenalidomide increases the expression of CYP27B1 in macrophages. In fact, this step is required for lenalidomide to spark the beneficial effects of vitamin D in terms of improving the antibody-dependent activity of macrophages against MM cells. To assess whe- ther lenalidomide therapy impacted the vitamin D pathway in vivo, we initially analyzed isolated monocytes from untreated MM patients and from IMiD-treated patients (for patient characteristics, please refer to Supplemental Fig. 4 MOR202-dependent elimination of MM cells is enhanced by lenalidomide and 25D3. a Generated macrophages were coincubated with VPD-labeled myeloma cell lines (as indicated) in different effector target ratios (1:1, 5:1, and 10:1) in the presence or absence of MOR202 (10 µg/ml) for 24 h. The number and the viability of mye- loma cells were measured by flow cytometry in the presence of 123count eBeads to determine absolute numbers of surviving MM cells. The percentage of MOR202-mediated killing was then calcu- lated using the following formula: MOR202-mediated killing = 100- ((absolute number of surviving MM cells in the presence of MOR202/ absolute number of surviving MM cells in the presence of IgG1) × 100%). b VPD-labeled MM target cells (as indicated) were added to lenalidomide-treated macrophage effectors (1 µM, 72 h) at E/T ratios of 1:1 with or without 25D3 (≈10 nM) in the presence or absence of MOR202 (10 µg/ml). After 24 h, surviving MM cells were enumerated by flow cytometric analysis of CD11b−/VPD+ cells in the presence of cell count beads. Percentage of MOR202-mediated killing was then calculated as described above. c CPD-labeled MM target cells (as indicated, red) were added to lenalidomide-treated macrophage (CFSE stained, green) effectors at E/T ratios of 1:1 with or without 25D3 in the presence or absence of MOR202. After 2 h phagocytosis was analyzed by confocal microscope at (630×). Scale bar: 20 µm. d Macrophages were treated with lenalidomide (1 µM) for 48 h with or without 25D3 (≈10 nM). After 24 h of incubation, macrophages were transfected with control siRNA or siRNA against CYP27B1. Trans- fected and non-transfected macrophages were co-incubated with stained OPM2 targets (E:T 1:1) in the presence or absence of MOR202 (10 µg/ml) for 24 h. The number and the viability of myeloma cells were measured by flow cytometry in the presence of 123count eBeads to determine absolute numbers of viable MM cells Table 3). The qPCR analyses revealed that monocytes displayed a significantly increased CYP27B1mRNA expression during IMiD therapy in opposition to monocytes from patients without treatment (Fig. 5a) (CYP27B1 in untreated: 0.05 ± 0.05 A.U. vs. CYP27B1 in IMiD-treated: 0.83 ± 0.3 A.U., p = 0.002). In addition, in monocytes from proteasome inhibitor-treated patients CYP27B1 mRNA expression remained, on average, unchanged. To directly examine whether lenalidomide regulates the CYP27B1 expression in MAMs, we longitudinally examined BM specimens of individual MM patients before and during lenalidomide therapy (n = 5) simultaneously for CYP27B1 and CD68 (for patient characteristics, please refer to Sup- plemental Table 4). Whereas a considerable number of MAMs expressed CYP27B1 during lenalidomide treatment, we found only low levels of CYP27B1 in MAMs in the untreated MM patient cohort (Fig. 5b). Noticeably, overall CYP27B1 tissue expression was found enhanced indicating a rather general (lenalidomide-triggered) phenomenon that is not only restricted to macrophages. Taken together, these results indicate that monocytes and MAMs have an enhanced expression of CYP27B1 under lenalidomide therapy, implicating that these macrophages may possess increased tumoricidal effector mechanisms, which obviously requires further ...
Context 4
... represent an abundant component of the stromal cell compartment in MM and have in principle the ability to eliminate MM cells opsonized by MOR202. To evaluate whether primary MAMs exert MOR202-mediated cyto- toxicity, we first measured cytotoxic activity of purified were isolated (n = 4), differentiated for 3 days with M-CSF and co- incubated with CPD-labeled myeloma cells (OPM2) in the presence or absence of 25D3 and MOR202 (10 µg/ml). After 2 h phagocytosis was analyzed by confocal microscope (630×). Scale bar: 20 µm MAMs, against autologous MM cells (n = 5). MAMs (CD163+/CD15−) and MM cells (CD138+) were isolated by flow cytometry-based sorting (Supplemental Fig. 12) and co-cultured in three different effector-to-target ratios (1:1, 5:1, and 10:1) with or without MOR202 (10 µg/ml) for 24 h. Comparable to the results for in vitro generated macro- phages (Supplemental Fig. 13) and MM cell lines (Fig. 4a), freshly isolated autologous MAMs from patients showed a potent capacity to target primary MM cells that were treated with MOR202. Significant killing was detected at an effector/target ratio of 5:1 (42 ± 12%, p = 0.048) and reached 45 ± 14% at an effector/target ratio of 10:1 ( Fig. 6a and Supplemental Fig. 15). To further demonstrate the relevance of the finding that the combination of lenalido- mide and vitamin D supplementation enhances MOR202- mediated cytotoxicity, isolated MAMs were treated with lenalidomide and then co-cultured with autologous MM cells in the presence of vitamin D-sufficient medium. Consistent with the results shown with macrophages from healthy donors (Fig. 4b), the combination of lenalidomide and 25D3 resulted in a significant enhancement of MOR202-mediated cytotoxicity by MAMs, in contrast to conditions with lenalidomide or 25D3 alone (untreated: 27 ± 8%, lenalidomide: 42 ± 3%, 25D3: 24 ± 16%, lena + 25D3: 73 ± 11%, p = 0,03) (Fig. 6b). Finally, we studied MOR202-mediated cytotoxicity of monocytes from untreated MM patients and during IMiD therapy. Interest- ingly, the MOR202-dependent cytotoxic capacity of monocytes isolated from patients not previously treated with IMiD toward MM cells (E:T 5:1) was similar in control and 25D3 supplemented medium (ctrl: 30 ± 6%, +25D3: 34 ± 6%, p = 0.41, Fig. 6c). Monocytes isolated from IMiD- treated patients, on the other hand, displayed a highly sig- nificant increase in MOR202-mediated killing in the pre- sence of 25D3 specifically (ctrl: 40 ± 3%, +25D3: 63 ± 3%, p = 0.0005, Fig. 6c). In line with the previously observed in vitro effect of lenalidomide and 25D3 on ADCP (Fig. 4c), patient-derived macrophages during IMiD therapy display an increased uptake of MM cells in the presence of 25D3 (Fig. ...
Context 5
... (RPMI8226: p = 0.0005; OPM2: p = 0.0064; MM1S: p = 0.001) ( Fig. 3b and Supplemental Fig. 4a), but no differences in the expression of CD138, SLAMF7, and CD47. These results were confirmed on the mRNA level by qPCR (Supplemental Fig. 4b). Of note, we did not observe any direct cytotoxic effects of 1,25D3 in our experimental settings (Supplemental Fig. 5a, b). Next, we examined whether vitamin D increases the expression of CD38 on primary MM cells of treatment naïve patients. There- fore, we treated BM samples of untreated MM (n = 6) patients for 48 h with 1,25D3 and analyzed the CD38 expression on MM cells (CD45 low , CD19 low , and CD138 high ) by flow cytometry. Similar to the MM cell ...
Context 6
... number and the viability of myeloma cells were measured by flow cytometry in the presence of 123count eBeads to determine absolute numbers of viable MM cells Table 3). The qPCR analyses revealed that monocytes displayed a significantly increased CYP27B1mRNA expression during IMiD therapy in opposition to monocytes from patients without treatment (Fig. 5a) (CYP27B1 in untreated: 0.05 ± 0.05 A.U. vs. CYP27B1 in IMiD-treated: 0.83 ± 0.3 A.U., p = 0.002). In addition, in monocytes from proteasome inhibitor-treated patients CYP27B1 mRNA expression remained, on average, unchanged. To directly examine whether lenalidomide regulates the CYP27B1 expression in MAMs, we longitudinally examined BM ...
Context 7
... of individual MM patients before and during lenalidomide therapy (n = 5) simultaneously for CYP27B1 and CD68 (for patient characteristics, please refer to Sup- plemental Table 4). Whereas a considerable number of MAMs expressed CYP27B1 during lenalidomide treatment, we found only low levels of CYP27B1 in MAMs in the untreated MM patient cohort (Fig. 5b). Noticeably, overall CYP27B1 tissue expression was found enhanced indicating a rather general (lenalidomide-triggered) phenomenon that is not only restricted to macrophages. Taken together, these results indicate that monocytes and MAMs have an enhanced expression of CYP27B1 under lenalidomide therapy, implicating that these ...
Context 8
... to the results for in vitro generated macro- phages (Supplemental Fig. 13) and MM cell lines (Fig. 4a), freshly isolated autologous MAMs from patients showed a potent capacity to target primary MM cells that were treated with MOR202. Significant killing was detected at an effector/target ratio of 5:1 (42 ± 12%, p = 0.048) and reached 45 ± 14% at an effector/target ratio of 10:1 ( Fig. 6a and Supplemental Fig. 15). To further demonstrate the relevance of the finding that the combination of lenalido- mide and vitamin D supplementation enhances MOR202- mediated cytotoxicity, isolated MAMs were treated with lenalidomide and then co-cultured with autologous MM cells in the presence of vitamin D-sufficient medium. ...

Citations

... VD has been shown to induce differentiation and decrease the proliferation of malignant cells [5]. Its anti-tumor activity was demonstrated for solid tumors, for instance, by enhancing the response to chemotherapy [13], as well as hematological malignancies, including MM [14,15]. The exact role of VD in MM has been reviewed by our team elsewhere [16]. ...
... They demonstrated in vitro that VD is a key molecule for restoring and maintaining the effector functions of myeloma-associated macrophages and that VD supplementation in combination with IMiDs can enhance the therapeutic efficacy of anti-CD38 antibodies. However, clinical effectiveness needs to be verified in clinical studies [15]. On top of vitamin D itself, its analogs, such as EB1089, also exhibit anti-MM activity. ...
Article
Full-text available
Multiple myeloma (MM) is a plasma cell malignancy that, despite recent advances in therapy, continues to pose a major challenge to hematologists. Currently, different classes of drugs are applied to treat MM, among others, proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies. Most of them participate in an interplay with the immune system, hijacking its effector functions and redirecting them to anti-MM activity. Therefore, adjuvant therapies boosting the immune system may be potentially beneficial in MM therapy. Vitamin D (VD) and vitamin K (VK) have multiple so called "non-classical" actions. They exhibit various anti-inflammatory and anti-cancer properties. In this paper, we investigated the influence of VD and VK on epigenetic alterations associated with the proliferative potential of MM cells and the development of BTZ resistance. Our results showed that the development of BTZ resistance is associated with a global decrease in DNA methylation. On the contrary, both control MM cells and BTZ-resistant MM cells exposed to VD alone and to the combination of VD and VK exhibit a global increase in methylation. In conclusion, VD and VK in vitro have the potential to induce epigenetic changes that reduce the proliferative potential of plasma cells and may at least partially prevent the development of resistance to BTZ. However, further ex vivo and in vivo studies are needed to confirm the results and introduce new supplementation recommendations as part of adjuvant therapy.
... Stromal cells in the BM niche might protect MM PCs by inducing the production of antiapoptotic molecules [71]. Furthermore, ADCP dysfunction through CD47 amplification in MM cells is related to reduced phagocytosis and tumor escape [72][73][74][75]. In addition, low frequencies of effector T lymphocytes, with reduced expression of the costimulatory molecule CD28 and pro-inflammatory M1 macrophages, are observed in patients with R/R MM or during disease progression [63,76,77]. ...
Article
Full-text available
CD38 and B-cell maturation antigens (BCMAs) are prevalently expressed on neoplastic plasma cells in multiple myeloma (MM), making them ideal therapeutic targets. Anti-CD38 monoclonal antibodies, such as approved daratumumab and isatuximab, are currently the milestone in MM treatment because they induce plasma cell apoptosis and kill through several mechanisms, including antibody-dependent cellular cytotoxicity or phagocytosis. BCMA is considered an excellent target in MM, and three different therapeutic strategies are either already available in clinical practice or under investigation: antibody–drug conjugates, such as belantamab-mafodotin; bispecific T cell engagers; and chimeric antigen receptor-modified T cell therapies. Despite the impressive clinical efficacy of these new strategies in the treatment of newly diagnosed or multi-refractory MM patients, several mechanisms of resistance have already been described, including antigen downregulation, the impairment of antibody-dependent cell cytotoxicity and phagocytosis, T- and natural killer cell senescence, and exhaustion. In this review, we summarize the current knowledge on the mechanisms of action and resistance of anti-CD38 and anti-BCMA agents and their clinical efficacy and safety.
... Accordingly, it can be hypothesized that VD and VK can mutually potentiate each other's effect, and their parallel administration can be beneficial in MM. Anti-MM properties of VD were demonstrated predominantly in in vitro studies [37,38]. Additionally, there were conducted human studies, both clinical trials and correlational, revealing the role of VD in MM patients [39][40][41]. ...
... The activation of the VD pathway by lenalidomide as well as the concurrent supplementation of VD enhances the therapeutic efficacy of MOR202. The obtained results suggest that maintaining proper 25(OH)D3 concentrations in MM patients may be of clinical significance [37]. Another study investigated the antiproliferative effects of 1.24(OH)2D2 in combination with bisphosphonate pamidronate on an MM H929 cell line. ...
Article
Full-text available
Multiple myeloma (MM) remains an incurable hematological malignancy. Bortezomib (BTZ) is a proteasome inhibitor widely used in MM therapy whose potent activity is often hampered by the development of resistance. The immune system is vital in the pathophysiology of BTZ resistance. Vitamins D (VD) and K (VK) modulate the immune system; therefore, they are potentially beneficial in MM. The aim of the study was to evaluate the effect of BTZ therapy and VD and VK supplementation on the proliferation potential and gene expression profiles of MM cells in terms of the development of BTZ resistance. The U266 MM cell line was incubated three times with BTZ, VD and VK at different timepoints. Then, proliferation assays, RNA sequencing and bioinformatics analysis were performed. We showed BTZ resistance to be mediated by processes related to ATP metabolism and oxidative phosphorylation. The upregulation of genes from the SNORDs family suggests the involvement of epigenetic mechanisms. Supplementation with VD and VK reduced the proliferation of MM cells in both the non-BTZ-resistant and BTZ-resistant phenotypes. VD and VK, by restoring proper metabolism, may have overcome resistance to BTZ in vitro. This observation forms the basis for further clinical trials evaluating VD and VK as potential adjuvant therapies for MM patients.
... Thus, harnessing and enhancing macrophage-mediated ADCP through repolarization of M1/M2 macrophages is poised to become a novel and effective strategy for immunotherapy. Lenalidomide was shown to improved MOR202 (an anti-CD38 mAb)-mediated tumoricidal activity of MAMs against primary MM cells by restoring the defective vitamin D pathway in these MAMs with reduced CYP27B1 level (225). In addition, lenalidomide and pomalidomide mediated a substantial CD38 upregulation on MM cell lines, which also contributes to a synergistic enhancement of cytotoxic activity by combining MOR202 with IMiDs (213). ...
Article
Full-text available
Immunomodulatory drugs (IMiDs) such as thalidomide, lenalidomide and pomalidomide are antitumor compounds that have direct tumoricidal activity and indirect effects mediated by multiple types of immune cells in the tumor microenvironment (TME). IMiDs have shown remarkable therapeutic efficacy in a set of B-cell neoplasms including multiple myeloma, B-cell lymphomas and chronic lymphocytic leukemia. More recently, the advent of immunotherapy has revolutionized the treatment of these B-cell neoplasms. However, the success of immunotherapy is restrained by immunosuppressive signals and dysfunctional immune cells in the TME. Due to the pleiotropic immunobiological properties, IMiDs have shown to generate synergetic effects in preclinical models when combined with monoclonal antibodies, immune checkpoint inhibitors or CAR-T cell therapy, some of which were successfully translated to the clinic and lead to improved responses for both first-line and relapsed/refractory settings. Mechanistically, despite cereblon (CRBN), an E3 ubiquitin ligase, is considered as considered as the major molecular target responsible for the antineoplastic activities of IMiDs, the exact mechanisms of action for IMiDs-based TME re-education remain largely unknown. This review presents an overview of IMiDs in regulation of immune cell function and their utilization in potentiating efficacy of immunotherapies across multiple types of B-cell neoplasms.
... After labeling MOR202 with a fluorescent dye, enhanced binding of the antibody to the surface of MM cells after incubation with 25(OH)D3 was found. The study concluded that the active form of VD not only acts on the effector cells but also increases the vulnerability towards anti-CD38 antibodies by enhancing CD38 expression on MM cells [35]. ...
Article
Full-text available
Multiple myeloma (MM) is a plasma cell malignancy with multifactorial etiology. One of the underlying mechanisms is immune system dysregulation. Immunotherapy is being widely introduced into various MM treatment protocols. Nevertheless, little is known about boosting the immune system with supportive treatment. Although classical actions of vitamin D (VD) are very well established, their non-classical actions related to the modulation of the immune system in MM are still a subject of ongoing research. In this literature review, we intend to summarize research conducted on VD and MM, both in vitro and in vivo, with particular emphasis on immune system modulation, the induction of the differentiation of malignant MM cells, synergic activity with anti-MM drugs, and MM-associated peripheral neuropathy.
... 37 Pomalidomide or lenalidomide can also increase the activity of anti-CD38 antibodies including Dara by upregulation of CD38. 38 Dara's half-life estimated following the first 16 mg/kg dose was 9 d. Based on population PK analysis, the mean half-life associated with nonspecific linear elimination was approximately 18 d. ...
... Progression-free survival 2 and OS were not mature. Median time to first response in responders was similar in both groups: 32 d (IQR [30][31][32][33][34][35][36][37][38][39][40] in the IsaKd and 33 d in the control group. Duration of response and time to next treatment was longer in the Isatuximab group than in the control group. ...
Article
Full-text available
CD38 is a transmembrane glycoprotein with ectoenzymatic activity and is highly and uniformly expressed on multiple myeloma (MM) cells. CD38 is expressed also at relatively low levels on normal lymphoid and myeloid cells, and in some tissues of non-hematopoietic origin. The specificity of this target has increased interest in new drugs and triggered the development of the CD38 monoclonal antibodies Daratumumab (fully human) and Isatuximab (chimeric). CD38 antibodies have pleiotropic mechanisms of action including Fc-dependent immune effector mechanisms, direct apoptotic activity, and immunomodulatory effects by the elimination of CD38+ immune-suppressor cells. Monoclonal antibody-based therapy has revolutionized MM therapy in the latest years increasing depth of response. This product review will focus on anti-CD38 monoclonal antibodies Daratumumab and Isatuximab efficacy, safety, pharmacokinetic and pharmacodynamic data from clinical trials.
... Increased number of CD163+ cells and serum soluble CD163 were observed in CTCL, atopic dermatitis, and psoriasis comparing with normal skin; increased lesion macrophages also been associated with disease progression and an overall worse prognosis in CTCL [11]. Moreover, although the exact mechanisms remain unknown, chemotherapy agents targeted on TAMs via reprograming M2 macrophages or promoting antitumor activities have proven effective in various hematopoietic malignancies including multiple myeloma and CNS lymphomas [12][13][14][15]. ...
Article
Full-text available
Polarization of tumor associated macrophages (TAMs) has been shown to have prognostic significance in different cancer types. This study evaluates the macrophage subtypes that predominates in GMF. Cases of GCTCL from 2007–2020 were identified (n = 6), clinical data was extracted from the electronic medical record, and all pathology slides were reviewed to confirm the diagnosis. Immunohistochemistry (IHC) studies were performed to characterize M1 and M2 macrophage polarization. CD68 (PGM1), pSTAT1, and CD163 were used as pan macrophage, M1, and M2 markers, respectively. The macrophages with positive staining at hot spot per high power field were counted and recorded for data analysis. The average age of patients was 60.5 years [range, 21–78], five patients (83%) were women and 1 (17%) was a man. Five patients were Caucasian (83%), and 1 was Black/African American (17%). Two patients had late stage GMF with M2 (CD163) predominance and the other three had early stage GMF with M1 (pSTAT1) predominance. Our study suggests that macrophage polarization present in GMF tends to be M1 in early stages and M2 in advanced stages. Additional studies are needed to further elucidate the microenvironment of macrophages present in GMF. Such findings may lead to prognostic and therapeutic advances in GMF.
... Although CD38 high NK cell population is reduced by CD38 mAb, it brings few destructive effects to the efficacy and safety during combined therapy, because IMiDs, such as Lenalidomide, markedly enhance NK cell-mediated ADCC by increasing the number and activity of activated CD16 + NK cells [71,98,101]. In addition, IMiDs also enhance CD38 mAb-mediated ADCP by promoting the tumoricidal activity of macrophages [102]. The IMiD-mediated recovery of CD16 + NK cells makes the combined therapy possibly a good choice to conquer the negative affect of reduction of CD38 high NK cells in CD38 mAbs (Figure 6a) treatment [103]. ...
Article
Full-text available
CD38 is highly expressed on multiple myeloma (MM) cells and plays a role in regulating tumor generation and development. CD38 monoclonal antibodies (mAbs) have been used as an effective therapy for MM treatment by various mechanisms, including complement-dependent cytotoxic effects, antibody-dependent cell-mediated cytotoxicity, antibody-dependent cellular phagocytosis, programmed cell death, enzymatic modulation, and immunomodulation. Although CD38 mAbs inhibit the proliferation and survival of MM cells, there are substantial side effects on antitumoral NK cells. The NK-mediated immune response needs to be further evaluated to minimize the adverse effects of NK cell loss. The killing effect of CD38 mAbs on CD38high NK cells should be minimized and the potential combination of CD38low/- NK cells and CD38 mAbs should be maximized to better benefit from their therapeutic efficacy against MM. CD38 mAb effects against MM can be maximized by combination therapies with immunomodulatory imide drugs (IMiDs), proteasome inhibitors (PIs), anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) antibodies, or cellular therapies for the treatment of MM, especially in patients with relapsed or refractory MM (R/R MM) and drug-resistant MM.
... Daratumumab-induced NK cell-mediated ADCC (41,42). Vitamin D can enhance cytotoxic activity in vitro; therefore, vitamin D supplementation in an IMID combined test could further improve the therapeutic effect of anti-CD38 antibodies (43). That study also found that the use of DNA methyltransferases (DNMTs) could achieve similar effects. ...
Article
Full-text available
Cluster of differentiation 38 (CD38) is a cell surface glycoprotein and multifunctional extracellular enzyme. As a NADase, CD38 produces adenosine through the adenosine energy pathway to cause immunosuppression. As a cell surface receptor, CD38 is necessary for immune cell activation and proliferation. The aggregation and polarization of macrophages are affected by the knockout of CD38. Intracellular NAD⁺ levels are reduced by nuclear receptor liver X receptor-alpha (LXR) agonists in a CD38-dependent manner, thereby reducing the infection of macrophages. Previous studies suggested that CD38 plays an important role in the regulation of macrophage function. Therefore, as a new marker of macrophages, the effect of CD38 on macrophage proliferation, polarization and function; its possible mechanism; the relationship between the expression level of CD38 on macrophage surfaces and disease diagnosis, treatment, etc; and the role of targeting CD38 in macrophage-related diseases are reviewed in this paper to provide a theoretical basis for a comprehensive understanding of the relationship between CD38 and macrophages.
... Lenalidomide is an immunomodulatory agent, with a pleomorphic activity on the TME, typically used in combination with an anti-CD20 monoclonal antibody for the treatment of both previously untreated and relapsed patients with FL. [54][55][56] While the majority of translational studies in FL have focused on characterizing its effects on T-cells, the effects of lenalidomide on TAMs have been better described in CLL and multiple myeloma (MM), with favorable effects on ADCC and ADCP through regulation of the vitamin D pathway, and on macrophage phenotype, through activation of Rap1 GTPase. [57][58][59][60][61] In pre-clinical lymphoma models lenalidomide has been shown to enhance macrophage-mediated ADCC of rituximab-opsonized lymphoma cells, with a 60% killing of Raji and Farage lymphoma cell lines as compared to 30% with rituximab alone. 62 Studies aimed at determining the impact of lenalidomide on TAMs in FL and mechanisms of resistance in this disease subtype are highly needed. ...
Article
Full-text available
The survival and proliferation of follicular lymphoma (FL) cells is strongly dependent on macrophages, their presence being necessary for the propagation of FL cells in vitro. To this regard, as shown also for the majority of solid tumors, a high tissue content of tumor-associated macrophages (TAMs), particularly if showing a pro-tumoral phenotype (also called M2) has strongly associated with a poor outcome among FL patients treated with chemotherapy. The introduction of rituximab, an anti-CD20 antibody which can be used by TAMs to performed antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis, has challenged this paradigm. In the rituximab-era, in fact, clinical studies have yielded conflicting results in FL, showing variable outcomes based on the type of employed regimen. This has highlighted for the first time that the impact of TAM on the prognosis of FL patients may depend on the administered treatment, emphasizing the need to better understand how currently available therapies affect macrophage function in FL. We summarize here the impact of approved and novel therapies for FL on the biology of TAMs, including radiation therapy, chemotherapy, anti-CD20 monoclonal antibodies, lenalidomide, and targeted agents, and describe their effects on macrophage phagocytosis, polarization and function. While novel agents targeting the CD47/SIRPα axis are being developed and showing promising activity in FL, a deeper understanding of macrophage biology and their complex pathways will help to develop novel and safer therapeutic strategies for patients with this type of lymphoma.