Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Nov 8;13(1):6739.
doi: 10.1038/s41467-022-34548-3.

CDKN1A is a target for phagocytosis-mediated cellular immunotherapy in acute leukemia

Affiliations

CDKN1A is a target for phagocytosis-mediated cellular immunotherapy in acute leukemia

Awatef Allouch et al. Nat Commun. .

Abstract

Targeting the reprogramming and phagocytic capacities of tumor-associated macrophages (TAMs) has emerged as a therapeutic opportunity for cancer treatment. Here, we demonstrate that tumor cell phagocytosis drives the pro-inflammatory activation of TAMs and identify a key role for the cyclin-dependent kinase inhibitor CDKN1A (p21). Through the transcriptional repression of Signal-Regularity Protein α (SIRPα), p21 promotes leukemia cell phagocytosis and, subsequently, the pro-inflammatory reprogramming of phagocytic macrophages that extends to surrounding macrophages through Interferon γ. In mouse models of human T-cell acute lymphoblastic leukemia (T-ALL), infusion of human monocytes (Mos) engineered to overexpress p21 (p21TD-Mos) leads to Mo differentiation into phagocytosis-proficient TAMs that, after leukemia cell engulfment, undergo pro-inflammatory activation and trigger the reprogramming of bystander TAMs, reducing the leukemic burden and substantially prolonging survival in mice. These results reveal p21 as a trigger of phagocytosis-guided pro-inflammatory TAM reprogramming and highlight the potential for p21TD-Mo-based cellular therapy as a cancer immunotherapy.

PubMed Disclaimer

Conflict of interest statement

A.A. and J.-L.P. are listed as co-inventors on a patent application related to p21TD-Mo-based cellular therapy (p21 expressing monocytes for cancer cell therapy, WO2021013764). J.-L.P. is a founding member of Findimmune SAS, an immuno-oncology biotech company. J.-L.P. discloses research funding not related to this work from NH TherAguix SAS. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Tumor cell phagocytosis triggers the proinflammatory activation of macrophages.
a Schematic representation of the coculture of CMFDA-labeled MDMs with CMTMR-labeled malignant hematologic cells. b Confocal micrograph of MDMs and Jurkat cells after 8 h of coculture. c, d Percentage of phagocytosis of leukemia cells or PBLs (c) or CD34+ AML cells (d) in control (Co.)- or ZVAD (100 µM)-treated cocultures. e FACS dot plot of Phago+ MDMs or Phago- MDMs sorted after 2 h of coculture with MOLT4 cells. f, g Confocal micrographs and percentages of CMTMR+CMFDA+ MDMs at 2 h (**p = 0.0079) (f, g) and 96 h (h, i) after Phago+ MDMs and Phago- MDMs sorting. jn Phago+ MDMs were analyzed in comparison to Phago- MDMs to characterize modulated genes by a microarray (**p = 0.0022, *p = 0.0152) (j), CD163 membrane expression by FACS (**p = 0.0039) (k), and IRF5 expression by western blot (WB) analysis (l) and the supernatant (SN) was evaluated for indicated proinflammatory cytokines by WB analysis (l) or for IFNγ by a cytokine microarray (*p = 0.0286) (m) or ELISA (***p = 0.0008) (n) at 96 h (j, k, m) and 7 d (l, n) after FACS sorting. o, p Transwell coculture model of Phago+ MDMs and Phago- MDMs at 2 h after FACS sorting (o), and iNOS expression identified by WB analysis of Phago- MDMs cocultured in the bottom chambers for 15 d (p). In b, l, p and e, f, h, the data are representative of n = 3 and n = 5 donors. In c, n, d, and g, i, the data are presented as the mean±SEM from n = 3, n = 6, and n = 5 donors. In d, the CD34+ cells were from n = 4 AML patients. In j, box plots show centre line as median, box limits as upper and lower quartiles, and whiskers as a minimum to maximum values, from n = 3 donors. In k and m, the data are donor matched from n = 9 and n = 4 donors. Exact p-values are indicated and determined with two- (g) or one-tailed (m) unpaired Mann-Whitney test, the Kolmogorov–Smirnov test (j), a two-tailed paired Wilcoxon test (k), and a two-tailed unpaired t test (n). Source data are provided as a Source Data file.
Fig. 2
Fig. 2. Macrophage-expressed p21 governs the phagocytosis of leukemia cells through the repression of SIRPα transcription.
a p21 expression in control (siCo.) or p21-silenced (sip21) MDMs after 24 h silencing. b Confocal micrograph of siCo. or sip21 CMFDA+ MDMs cocultured with CMTMR+ Jurkat cells. ce Percentages of Jurkat (c), MOLT4 (d) or patient CD34+ AML (e) phagocytosis by siCo. or sip21 MDMs (**p = 0.0078, *p = 0.0312, ****p = 6.1e-5). f Phagocytosis (Fluorescence Units (FU)) of pHrodo Green E. coli bioparticle by siCo. or sip21 MDMs. g Percentages of CrCD47MOLT4 or CrCo.MOLT4 phagocytosis by MDMs transduced with control (Co.TD) or p21-expressing (p21TD) lentiviral vectors (LVs) after 72 h of transduction (**p = 0.0061, **p = 0.0053, **p = 0.0048). hm SIRPα and p21 proteins (h, k) or mRNAs (i, j, l, m) in siCo. or sip21 MDMs (**p = 0.0024, **p = 0.0020) (hj) or in Co.TD-MDMs or p21TD-MDMs (**p = 0.0063, *p = 0.0291) (km). n ChIP-qPCR assays performed with control MDMs, Co.TD-MDMs (**p = 0.0016) or p21TD-MDMs (**p = 0.0017, *p = 0.0194) immunoprecipitated with control IgG or anti-p21 antibodies and analyzed by qPCR at SIRPα promoter. o, p Luciferase activities in siCo. or sip21 MDMs (o) or in Co.TD-MDMs or p21TD-MDMs (p) transduced with a lentiviral vector expressing a luciferase reporter gene under the control of the SIRPα promoter (*p = 0.0431, *p = 0.0458). qs MDMs derived from monocytes transduced with Co.TD and/or p21TD LVs and/or SIRPα-expressing LVs (SIRPαTD) assessed for the indicated proteins (q) and for phagocytosis of mCherry+ MOLT4 cells (**p = 0.0079, *p = 0.0472, ***p = 0.0002, ***p = 0.0004) (r, s). In a, b, h, k, q and r, the data are representative of n = 3 donors. In c, d, e and o, p, the data are donor matched from n = 8, n = 6, n = 9, n = 4 and n = 3 donors. In e, the CD34+ cells were from n = 6 AML patients. In i, j and g, l, m, n, s, the data are presented as the mean±SEM from n = 5 and n = 3 donors. Exact p-values are indicated and determined with two-tailed paired Wilcoxon (c, d, e), two- (i, j, l, n) or one-tailed (m) ratio-paired t, and one-tailed paired t (o, p) tests and two-way ANOVA with Sidak’s (g) or one-way ANOVA Tukey’s (s) multiple comparison tests. Source data are provided as a Source Data file.
Fig. 3
Fig. 3. Prophylactic adoptive transfer of p21TD-Mos decreases the leukemic burden and prolongs mouse survival in a model of human T-ALL.
a Schematic representation showing prophylactic adoptive transfer of CFSE-labeled control (Co.TD), p21 (p21TD) and/or SIRPα (SIRPαTD) genetically engineered human monocytes (Mos) into NSG mice previously treated with total body irradiation (TBI), which were then engrafted with mCherry+ MOLT4 cells 7 days later. bk Engrafted mice that received the indicated monocytes were assessed at 21 d to measure body (**p = 0.0079) (b, c) and spleen (*p = 0.00317) weights (d, e); the leukemic burdens in the blood, bone marrow (BM) and spleen by FACS (**p = 0.0079) (f, g, h) and in liver tissues by confocal microscopy (**p = 0.0079) (i, j); and survival (***p = 0.0003) (k). l Survival of engrafted mice that received indicated monocytes and were treated 21 d after monocyte transfer with control (Co.) or clodronate (Clodro)-containing liposomes (****p = 4.8e-9, ****p = 0.1.1e-5). m Survival of engrafted mice that received the indicated monocytes (****p = 3.2e-5, ****p = 7e-10, ****p = 2.1e-7, ****p = 1e-6). n Survival of engrafted mice that received Co.TD-Mos, p21TD-Mos, SIRPαTD-Mos or p21+SIRPαTD-Mos and were treated on d 15 after Mo transfer for 14 d with daily injections of isotype control (IgG) or anti-CD47 blocking antibodies (100 μg/mouse) (****p = 9.4e-12, p = 0.0942 (n.s.), ****p = 2.9e-7, ****p = 2.7e-6, ****p < 1e-15, ****p = 4.5e-14, ****p = 9e-10). In b, d and i, the data are representative of n = 5 mice/group. In c, e, f, g, h and j, the data are presented as the mean±SEM from n = 5 mice/group. In k, m, n and l, the survival data are from n = 5 and n = 6 mice/group. Exact p-values are indicated and determined with two-tailed unpaired Mann-Whitney (c, e, f, g, h, j) and log-rank Mantel–Cox (k, l, m, n), tests. Source data are provided as a Source Data file.
Fig. 4
Fig. 4. Prophylactic adoptive transfer of p21TD-Mos triggers the proinflammatory activation of TAMs and prolongs the survival of human T-ALL-engrafted mice in an IFNγ-dependent manner.
a, b Confocal micrographs of phagocytic (mCherry+CFSE+) macrophages in the spleen (a) and CD163 membrane expression identified by FACS on sorted single-positive CFSE+ (Phago-) MDMs or phagocytic mCherry+CFSE+ (Phago+) MDMs from the spleen and BM (*p = 0.033) (b) of mCherry+ MOLT4 cell-engrafted NSG mice that received Co.TD-Mos or p21TD-Mos, as shown in Fig. 3a, on d 21 after Mo transfer. ce Percentages of iNOS-expressing (iNOS+) cells, determined by immunofluorescence staining, among Phago+ MDMs (c), MDMs (d) or Phago- MDMs (****p = 4.6e-5, ****p = 5e-5, ***p = 0.0004) (e) sorted from tumor-free (d) or mCherry+ MOLT4 cell-engrafted (c, e) NSG mice that received Co.TD-Mos or p21TD-Mos; cells collected on d 21 and 35 after Mo transfer. f Percentages of iNOS+ cells among human TAMs (hCD14+hCD11b+hCD163+CFSE+), sorted from the BM and spleen of mCherry+ MOLT4 cell-engrafted NSG mice adoptively transferred with CFSE+ human Mo; cells collected on d 35 after Mo transfer. These cells were cocultured, as shown in Supplementary Fig. 16, in the bottom chambers of Transwell devices with Phago- MDMs or Phago+ MDMs sorted from the spleen and BM of mCherry+ MOLT4 cell-engrafted NSG mice adoptively transferred with Co.TD-Mos or p21TD-Mos (d 35 after Mo transfer) or with control MDMs differentiated in vitro from Co.TD-Mos or p21TD-Mos (****p = 1.4e-8, ****p = 4.2e-5, ****p = 3.8e-10). g, h Schematic representation (g) and survival Fig. 4h (****p = 1.2e-7, ***p = 0.0002) (h) of mCherry+ MOLT4 cell-engrafted NSG mice that received Co.TD-Mos or p21TD-Mos and were treated 15 d after Mo transfer with isotype control (IgG) or anti-IFNγ blocking antibodies. In (a), the data are representative of n = 5 mice/group. In b, the data are mouse matched from n = 3 mice/group. In cf, the data are presented as the mean±SEM from n = 3 mice/group. In h, the survival data are from n = 7 mice/group. Exact p-values are indicated and determined with one-way ANOVA with Friedman’s (b) and Sidak’s (f) or two-way ANOVA with Tukey’s (e) multiple comparison tests or determined with log-rank Mantel-Cox (h), test. Source data are provided as a Source Data file.
Fig. 5
Fig. 5. Curative adoptive transfer of p21TD-Mos prolongs mouse survival in diagnosed or relapsed T-ALL-derived PDX models.
a Schematic representation showing curative adoptive transfer of Co.TD-Mos or p21TD-Mos into NSG mice engrafted with T-ALL PDXs. bd Survival of mice engrafted with T-ALL PDX#1 (****p = 1.1e-5) (b), PDX#2 (****p = 1.9e-6) (c) or PDX#3 (**p = 0.003) (d) and treated curatively, as shown in a, with Co.TD-Mos or p21TD-Mos. PDX#1 and PDX#2 were from the same patient but isolated at the diagnosis and relapse stages, respectively. In bd, the survival data are from n = 5 mice/group. Exact p-values are indicated and determined with the log-rank Mantel-Cox (bd) test. Source data are provided as a Source Data file.

References

    1. Chao MP, Majeti R, Weissman IL. Programmed cell removal: a new obstacle in the road to developing cancer. Nat. Rev. Cancer. 2011;12:58–67. - PubMed
    1. Feng M, et al. Programmed cell removal by calreticulin in tissue homeostasis and cancer. Nat. Commun. 2018;9:3194. - PMC - PubMed
    1. Jaiswal S, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271–285. - PMC - PubMed
    1. Cohen LJ, et al. Commensal bacteria make GPCR ligands that mimic human signalling molecules. Nature. 2017;549:48–53. - PMC - PubMed
    1. Barkal AA, et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature. 2019;572:392–396. - PMC - PubMed

Publication types

Substances