Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2020

Open Access 01.12.2020 | Letter to the Editor

Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions

verfasst von: Li Zhang, Lin Tian, Xiaoyang Dai, Hua Yu, Jiajia Wang, Anhua Lei, Mengmeng Zhu, Jianpo Xu, Wei Zhao, Yuqing Zhu, Zhen Sun, Hao Zhang, Yongxian Hu, Yanlin Wang, Yuming Xu, George M. Church, He Huang, Qinjie Weng, Jin Zhang

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2020

Abstract

The Chimera antigen receptor (CAR)-T cell therapy has gained great success in the clinic. However, there are still major challenges for its wider applications in a variety of cancer types including lack of effectiveness due to the highly complex tumor microenvironment, and the forbiddingly high cost due to the personalized manufacturing procedures. In order to overcome these hurdles, numerous efforts have been spent focusing on optimizing Chimera antigen receptors, engineering and improving T cell capacity, exploiting features of subsets of T cell or NK cells, or making off-the-shelf universal cells. Here, we developed induced pluripotent stem cells (iPSCs)-derived, CAR-expressing macrophage cells (CAR-iMac). CAR expression confers antigen-dependent macrophage functions such as expression and secretion of cytokines, polarization toward the pro-inflammatory/anti-tumor state, enhanced phagocytosis of tumor cells, and in vivo anticancer cell activity. This technology platform for the first time provides an unlimited source of iPSC-derived engineered CAR-macrophage cells which could be utilized to eliminate cancer cells.
Hinweise
Li Zhang, Lin Tian, Xiaoyang Dai, Hua Yu and Jiajia Wang have contributed equally to this work

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13045-020-00983-2.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CAR
Chimera antigen receptor
DC
Dendritic cells
ERK
Extracellular-regulated protein kinase
HSC
Hematopoietic stem cell
ICT
Immune-checkpoint therapy
IFN-γ
Interferon gamma
IP
Intraperitoneal injection
iPSC
Induced pluripotent stem cells
LPS
Lipopolysaccharide
MPS
Mononuclear phagocyte system
PBMC
Peripheral blood mononuclear cells
qRT-PCR
Quantitative reverse transcription PCR

To the Editor,

Recently, CAR-iPSC-differentiated CAR-expressing T cells and NK cells have been reported to have potent cytotoxic activity against cancer cells, and they represent a new family of engineered stem cell-derived immune cells for CAR therapies [1, 2]. Myeloid cells such as macrophages have been utilized as a type of effector cells to combat cancer cells by means of their phagocytosis function [3, 4]. However, immortalized macrophage cell lines are not applicable to clinical settings, and bone marrow or PBMC-derived primary macrophages are not efficiently engineered, thus leaving iPSC-derived macrophage cells as a great source for myeloid cell-based immunotherapy. Upon challenge with antigen-expressing cancer cells, CAR-expressing iPSC-induced macrophage (CAR-iMac) cells showed antigen-dependent macrophage functions. Expression of a CAR targeting tumor-associated antigen conferred CAR-iMac cells in vitro and in vivo anti-tumor effects.
First, we started from deriving iPSCs from peripheral blood mononuclear cells (PBMC) of a healthy donor with non-integrating episomal vectors encoding reprogramming factors (Fig. 1a), and isolated single iPSC clones (Additional file 1: Fig. S1). The materials and methods are shown in detail in the Additional file 2. Then, we compared different CD19 CARs and chose the conventional CAR to introduce into the iPSCs by lentiviral transduction (Fig. 1a and Additional file 1: Fig. S2a–g). Then, we established a protocol of myeloid/macrophage differentiation to induce CAR-iPSCs toward myeloid cell lineages (Additional file 1: Fig. S2h). Differentiated cells showed typical macrophage marker gene expression (Fig. 1b, Additional file 1: Fig. S4b–i). The products can be collected from 20 to 30 day for multiple times to allow further expansion to have a final yield of above 50-fold of the starting iPSCs (Additional file 1: Fig. S3a), with high purity indicated by ~ 100% of CD11b and CD14 expression at later days (Fig. 1b). Key macrophage marker genes were induced, whereas pluripotent marker genes disappeared (Fig. 1c; Additional file 3: Table S1). RNA-sequencing using differentiated cells showed that iPSCs clustered with precursor cells, and late-day differentiated cells clustered with primary macrophage cells, or untransduced iPSC-differentiated macrophage cells from previous studies [5, 6](Fig. 1d, e). GO analyses showed strong enrichment of innate immunity-related functions in 28-day differentiated cells (Fig. 1f; Additional file 4: Table S2).
Next, we further dissected their subpopulations by performing single-cell RNA-sequencing analysis. These cells clustered away from undifferentiated CAR-iPSCs (Fig. 1g), and they appeared to be largely homogenous with only a small number of cells not clustered with the main population (Fig. 1h). Blasting the differentiated single cells in a database of human cell atlas containing single-cell RNA-sequencing data revealed that these iMac cells mainly clustered with macrophages (Fig. 1i and Additional file 1: Fig. S6a). Trajectory analysis revealed that CAR-iMac cells went through a path from HSC to macrophage and DC cells without major branches (Fig. 1j, Additional file 1: Fig. S5c).
Moreover, all 10 clusters of differentiated cells showed strong signatures of M2 state [710] (Fig. 1k, Additional file 1: Fig. S6b). We further compared the single-cell data in the 10 clusters with bulk RNA-seq data from the LPS/IFN-γ-polarized M1 cells or IL-4/IL-10-polarized M2 cells, by examining M1/M2-associated genes (Fig. 1l, Additional file 1: Fig. S5d), and found that most clusters were more similar to the M2 state particularly when using metabolism genes as markers [5, 6, 1115].
Next, we incubated the CAR (CD19)-iMac cells, CAR (meso)-iMac cells, or control iMac cells with CD19-expressing K562 leukemia cells or mesothelin-expressing OVCAR3/ASPC1 ovarian/pancreatic cancer cells. Compared with K562 alone, K562-CD19 cells were more likely to be phagocytosed by CAR (CD19)-iMacs (Fig. 2a, b), and compared with control cells, CAR (meso)-iMac showed increased phagocytosis activity against OVCAR3 and ASPC1 cells (Fig. 2g, h and Additional file 1: Fig. S7g). Intracellular signaling such as phosphorylation of ERK and NF-κB(P65) proteins were increased in CAR-iMacs co-cultured with CD19-expressing K562 cells compared to K562 cells, or to CAR-iMac cells cultured alone (Fig. 2c). We also examined cytokine gene expression in CAR(CD19)-iMac and CAR (meso)-iMac cells when they were incubated with tumor cells and found antigen-dependent increase in M1 pro-inflammatory cytokine expression(Fig. 2d, j and Additional file 1: Fig. S7h). Moreover, transcriptional analysis showed that CAR(CD19)-iMac cells and CAR(meso)-iMac cells showed strong enrichment of up-regulated genes in GO or KEGG terms of “positive regulation of cytokine secretion,” “antigen processing and presentation,” and “Toll-like receptor signaling pathway,” indicating these cells are more wired toward the pro-inflammatory state, when they encounter the antigen (Fig. 2e, f, i and Additional file 1: Fig. S7i).
When injected into NSG mice, these CAR-iMac cells expanded in vivo till around day 3 for about two fold, and persisted till more than 20 days and gradually disappeared after around 30 days (Fig. 2k). To test their anti-tumor cell activity, we first intraperitoneally injected ovarian cancer cells HO8910 expressing a luciferase gene into NSG mice. In order to further polarize CAR-iMac cells toward M1, we treated them with IFN-γ and washed IFN-γ away before injection (Additional file 1: Fig. S8). CAR(meso)-iMac-treated mice showed reduced tumor burden compared to the control group on day 4, 11, and 14 (Fig. 2l, m). These data demonstrate that the CAR confers anti-cancer cell activities in iMacs in vivo. The efficacy and persistency need to be further improved by designing more effective CARs or further modifying the CAR-iMac cells to stay constitutively active.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13045-020-00983-2.

Acknowledgements

We thank George Q. Daley and Thorsten M. Schlaeger for providing reprogramming plasmids. We thank the Core Facilities of Zhejiang University School of Medicine for technical support and Center for Drug Safety Evaluation at Zhejiang University for animal technical support. We thank Ming Chen (College of Life Sciences, Zhejiang University) for computational analyses support and Harvey Lodish (MIT) for helpful Discussion. We thank CellOrigin for supporting this project.
The animal study was performed in compliance with relevant regulatory standards. All animal studies are approved by ZJU IRB committee.
Written informed consent for publication was obtained from all participants.

Competing interests

A patent has been filed related to the data presented. JZ and GC are founders and scientific advisors for CellOrigin.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Themeli M, Kloss CC, Ciriello G, Fedorov VD, Perna F, Gonen M, et al. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat Biotechnol. 2013;31(10):928–33. CrossRef Themeli M, Kloss CC, Ciriello G, Fedorov VD, Perna F, Gonen M, et al. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat Biotechnol. 2013;31(10):928–33. CrossRef
2.
Zurück zum Zitat Li Y, Hermanson DL, Moriarity BS, Kaufman DS. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23(2):181-92e5. CrossRef Li Y, Hermanson DL, Moriarity BS, Kaufman DS. Human iPSC-derived natural killer cells engineered with chimeric antigen receptors enhance anti-tumor activity. Cell Stem Cell. 2018;23(2):181-92e5. CrossRef
3.
Zurück zum Zitat Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, et al. Chimeric antigen receptors that trigger phagocytosis. Elife. 2018;7:e36688. CrossRef Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, et al. Chimeric antigen receptors that trigger phagocytosis. Elife. 2018;7:e36688. CrossRef
4.
Zurück zum Zitat Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38(8):947–53. CrossRef Klichinsky M, Ruella M, Shestova O, Lu XM, Best A, Zeeman M, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38(8):947–53. CrossRef
5.
Zurück zum Zitat Zhang H, Xue C, Shah R, Bermingham K, Hinkle CC, Li W, et al. Functional analysis and transcriptomic profiling of iPSC-derived macrophages and their application in modeling Mendelian disease. Circ Res. 2015;117(1):17–28. CrossRef Zhang H, Xue C, Shah R, Bermingham K, Hinkle CC, Li W, et al. Functional analysis and transcriptomic profiling of iPSC-derived macrophages and their application in modeling Mendelian disease. Circ Res. 2015;117(1):17–28. CrossRef
6.
Zurück zum Zitat Douvaras P, Sun B, Wang M, Kruglikov I, Lallos G, Zimmer M, et al. Directed differentiation of human pluripotent stem cells to microglia. Stem Cell Rep. 2017;8(6):1516–24. CrossRef Douvaras P, Sun B, Wang M, Kruglikov I, Lallos G, Zimmer M, et al. Directed differentiation of human pluripotent stem cells to microglia. Stem Cell Rep. 2017;8(6):1516–24. CrossRef
7.
Zurück zum Zitat Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Investig. 2012;122(3):787–95. CrossRef Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Investig. 2012;122(3):787–95. CrossRef
8.
Zurück zum Zitat Viola A, Munari F, Sanchez-Rodriguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Front Immunol. 2019;10:1462. CrossRef Viola A, Munari F, Sanchez-Rodriguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Front Immunol. 2019;10:1462. CrossRef
9.
10.
Zurück zum Zitat Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25(7):771–84. CrossRef Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25(7):771–84. CrossRef
11.
Zurück zum Zitat Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958–69. CrossRef Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958–69. CrossRef
12.
Zurück zum Zitat Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40(2):274–88. CrossRef Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40(2):274–88. CrossRef
13.
Zurück zum Zitat Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13. CrossRef Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13. CrossRef
14.
Zurück zum Zitat Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61. CrossRef Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61. CrossRef
15.
Zurück zum Zitat Lachmann N, Ackermann M, Frenzel E, Liebhaber S, Brennig S, Happle C, et al. Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Rep. 2015;4(2):282–96. CrossRef Lachmann N, Ackermann M, Frenzel E, Liebhaber S, Brennig S, Happle C, et al. Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Rep. 2015;4(2):282–96. CrossRef
Metadaten
Titel
Pluripotent stem cell-derived CAR-macrophage cells with antigen-dependent anti-cancer cell functions
verfasst von
Li Zhang
Lin Tian
Xiaoyang Dai
Hua Yu
Jiajia Wang
Anhua Lei
Mengmeng Zhu
Jianpo Xu
Wei Zhao
Yuqing Zhu
Zhen Sun
Hao Zhang
Yongxian Hu
Yanlin Wang
Yuming Xu
George M. Church
He Huang
Qinjie Weng
Jin Zhang
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00983-2

Weitere Artikel der Ausgabe 1/2020

Journal of Hematology & Oncology 1/2020 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.