Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced...

28
1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell Functions for Liquid and Solid Tumors Li Zhang 1, * , Lin Tian 1, * , Xiaoyang Dai 2, * , Hua Yu 1, * , Jiajia Wang 2, * , Anhua Lei 1 , Wei Zhao 1 , Yuqing Zhu 1 , Zhen Sun 1 , Hao Zhang 3 , George M. Church 4 , He Huang 3 , Qinjie Weng 2, † , Jin Zhang 1, † 1 Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, & The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. Institute of Hematology, Zhejiang University, Hangzhou 310058, China 2 Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research, Zhejiang University, Hangzhou 310058, China. 3 The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. Institute of Hematology, Zhejiang University, Hangzhou 310058, China. 4 Department of Genetics and Wyss Institute for Biologically Inspired Engineering, Harvard Medical School, Boston, Massachusetts 02115, United States. * These authors contributed equally to this work Correspondence: [email protected], [email protected] One sentence summary: We developed CAR-expressing iPSC-induced macrophage cells that have antigen-dependent phagocytosis and pro-inflammatory functions and anti-cancer cell activity for both liquid and solid tumor cells. 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 However, there are still major challenges for its wider applications in a variety of cancer (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprint this version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270 doi: bioRxiv preprint

Transcript of Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced...

Page 1: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent

Anti-Cancer Cell Functions for Liquid and Solid Tumors

Li Zhang1, *, Lin Tian1, *, Xiaoyang Dai2, *, Hua Yu1, *, Jiajia Wang2, *, Anhua Lei1, Wei Zhao1,

Yuqing Zhu1, Zhen Sun1, Hao Zhang3, George M. Church4, He Huang3, Qinjie Weng2, †, Jin

Zhang1, †

1Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, &

The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang,

China. Institute of Hematology, Zhejiang University, Hangzhou 310058, China

2Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer

Drug Research, College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and

Research, Zhejiang University, Hangzhou 310058, China.

3The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang,

China. Institute of Hematology, Zhejiang University, Hangzhou 310058, China.

4Department of Genetics and Wyss Institute for Biologically Inspired Engineering, Harvard

Medical School, Boston, Massachusetts 02115, United States.

*These authors contributed equally to this work

†Correspondence: [email protected], [email protected]

One sentence summary: We developed CAR-expressing iPSC-induced macrophage cells that

have antigen-dependent phagocytosis and pro-inflammatory functions and anti-cancer cell

activity for both liquid and solid tumor cells.

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

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 2: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

2  

types including lack of effectiveness due to the highly complex tumor microenvironment, and

the forbiddingly high cost due to 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 T cells. Here, we developed induced pluripotent

stem cells (iPSCs)-derived, CAR-expressing macrophage cells (CAR-iMac). These cells

showed antigen-dependent macrophage functions such as expression and secretion of cytokines,

polarization toward the pro-inflammatory/anti-tumor state, and phagocytosis of tumor cells, as

well as some in vivo anti-cancer cell activity for both liquid and solid tumors. 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 or modulate the tumor

microenvironment in liquid and solid tumor immunotherapy.  

Introduction

Human iPSCs derived from patients’ own cells can be differentiated to multiple cell types and

used for disease modeling, drug screening cell models and cell-based therapies1. Recently,

engineered T cells have shown great success in treating cancers in the clinic2. However, there

are a few outstanding challenges for engineering primary immune cells, including variable

transduction efficiency, highly heterogeneous genetic outcomes upon editing the genome of the

targeted cells, and limited cell resources for certain cases such as from hematological cancer

patients for which immune cell itself is transformed. iPSC-derived immune cells, due to their

flexibility of expansion and genome editing at the iPSC stage3, 4, in theory have advantages in

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 3: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

3  

dealing with those challenges above, and have been proved to be effective in treating B cell and

ovarian cancer cells in pre-clinical settings such as iPSC differentiated T cells5 and NK cells6.  

Other than lymphoid lineage cytotoxic cells, a larger number of myeloid cells exist in the tumor

microenvironment7. Recently, myeloid cells such as macrophages have been utilized as a type

of effector cells to combat cancer cells by means of their phagocytosis function8. However, the

immortalized cell lines used in the previous study are not applicable to clinical settings, and

bone marrow-derived macrophages are not easily engineered, thus leaving iPSC-derived

macrophage cells as a great source for myeloid cell-based cancer immunotherapy. In addition

to phagocytosis function, it is believed that macrophages in a polarized pro-inflammatory state

can alter the tumor microenvironment through secreting cytokines to stimulate T cells9.

Moreover, it was reported that tumor macrophages transition to a pro-inflammatory polarized

state upon immune-checkpoint therapy (ICT) treatment in animal models10, and an

inflammatory signature of tumor samples predicts clinical benefits in ICT-treated patients11.

Applying the above features of macrophages to combat tumor cells is currently under intensive

investigation largely by targeting endogenous macrophages in vivo12, but not much effort has

been spent on adoptive transfer therapies using monocyte/macrophages13, partly due to the

difficulties in obtaining or expanding enough cells, and the low efficiency in engineering them

in order to harness these cells for combating cancer. Here, we used human iPSCs to introduce

a CD19 or mesothelin-targeting CAR, and differentiated them into macrophages, a product we

named as CAR-iMac. We thoroughly characterized these cells with bulk/single cell RNA-

sequencing and functional analyses, and showed they are heterogeneous and functional

macrophage cells. Upon challenge with antigen-expressing cancer cells, CAR-iMac cells show

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 4: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

4  

antigen-dependent increase of phagocytosis, strong induction of antigen-dependent cytokine

expression and secretion, and polarization toward a more inflammatory M1-like state.

Transplanting CD19 or Mesothelin CAR-iMac cells to liquid and solid tumor models showed

some in vivo anti-tumor effect. This CAR-iMac system provides a proof-of-principle platform

to use pluripotent stem cells to obtain a large amount of engineered macrophage cells with

antigen-dependent functions for adoptive transfer-based immune cell therapies. 

Results

Engineering a CAR-expressing iPSC line for producing macrophages 

We aim to develop engineered macrophages that have the following features: (1) the

differentiated macrophages are capable of phagocytosing tumor cells and producing cytokines

that may alter the tumor microenvironment; (2) these functions should be antigen-dependent;

and (3) the engineered cells should have a high percentage of transgene expression and can be

produced in large scale (Fig. 1a). First, we started from deriving iPSCs from Peripheral blood

mononuclear cells (PBMC) of a healthy donor with non-integrable episomal vectors encoding

OCT4, SOX2, KLF4, L-MYC and LIN28A (Supplementary Fig. 1a,b). Isolated single iPSC

clones showed pluripotent markers OCT3/4 and LIN28A/B expression (Supplementary Fig.

1c), and formed teratomas when injected to immune-deficient mice (Supplementary Fig. 1d).

We originally designed a CD19 CAR supposedly more suitable for macrophages by replacing

the 4-1BB and CD3ζ intracellular domains (T-CAR) with the macrophage CD86 and FcγRI

intracellular domains (M-CAR) (Supplementary Fig. 2a), and transduced M-CAR and T-CAR

into the THP-1 human monocyte cell line with lentivirus and achieved comparable levels of

expression (Supplementary Fig. 2b,c). To our surprise, even though the M-CAR-transduced

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 5: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

5  

cells showed increased level of cytokine expression and secretion when encountering the

antigen CD19-expressing K562 cells compared with the untransduced control, the increase was

more significant for the T-CAR-transduced cells (Supplementary Fig. 2d,e), suggesting T-CAR

is capable of stimulating monocyte/macrophage intracellular signaling and activating

macrophage specific genes. Based on these results, the CD19-targeting T-CAR transgene

(named as CAR thereafter) was introduced into the iPSCs by lentiviral transduction (Fig. 1a

and Supplementary Fig. 2f). qPCR showed mRNA of the CAR transgene was highly expressed

in these iPSCs (Supplementary Fig. 2g). We thus engineered a CAR-expressing iPSC line

amenable for producing differentiated products with antigen-dependent functions. 

CAR-expressing iPSCs can differentiate to macrophage cells 

Next, we established a protocol of myeloid/macrophage differentiation to induce iPSCs toward

myeloid cell lineages (Fig. 1b). After a period of mesoderm induction, hematopoietic

specification, myeloid expansion, and macrophage maturation, differentiated cells showed

typical macrophage marker gene expression such as CD14, CD11b, CD163 and CD86 (Fig. 1c),

as well as CAR expression on cell surface detected with an antibody against CD19 CAR

(Supplementary Fig. 2h). Consistently, key macrophage genes were induced such as AIF1,

CSF1R and SPI1, whereas pluripotent marker genes disappeared such as LIN28A and POU5F1

(Fig. 1d). RNA-sequencing using undifferentiated cells, early-day precursor cells and 18-day,

28-day 38-day differentiated cells showed that iPSCs clustered with precursor cells, and late-

day differentiated cells clustered with primary macrophage cells, or iPSC-differentiated

macrophage cells from previous studies14, 15 (Fig. 2a,b). However, it is challenging to classify

iMac to M0, M1 or M2 types of macrophages, likely because of the heterogeneous feature of

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 6: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

6  

the primary and differentiated cells from the published bulk RNA-sequencing data. Similarly,

selected M1 and M2 marker genes both showed high expression determined by qRT-PCR

(Supplementary Fig. 3a, b). GO and KEGG pathway analyses showed strong enrichment of

innate immunity-related functions in 18 or 28-day differentiated cells such as: “positive

regulation of cytokine production”, “regulation of innate immune responses” and “regulation

of phagocytosis” (Fig. 2c, d). Quantitative PCR also validated that the differentiated cells

expressed high levels of Toll-like receptor, Fc receptor and cytokine receptor genes, HLA genes,

NF-κB pathway genes, and costimulatory signaling genes, etc (Supplementary Fig. 3c-h), all

of which are required for certain specific monocyte/macrophage/dendritic cell (DC) functions.

Importantly, these genes are expressed in comparable levels with primary PBMC-derived

macrophage cells (Supplementary Fig. 3i). Together, these data show CAR-expressing iPSCs

can differentiate to myeloid lineage and macrophage-like cells. 

Single cell RNA-seq analysis of CAR-iMac reveals a differentiation trajectory mainly

toward M2 macrophage and dendritic cells  

To dissect heterogeneity of these CAR-iMac cells, we performed single cell RNA-sequencing

analysis of differentiated cells. These cells clustered away from undifferentiated iPSCs (Fig.

3a), and they appeared to be largely homogenous with only a few sub-populations not clustered

with the main population (Fig. 3b). Blasting the differentiated single cells in a database of

human cell atlas containing single cell RNA-sequencing data of various cell types revealed that

these iMac cells mainly clustered with macrophages (C0 and C5) or DCs (dendritic cells, C1,

C2, C3 and C8) (Fig. 3b, c), with typical genes expressed in each clusters such as CD14 in

macrophages and CD1C and HLA-DRB1 in DCs (Supplementary Fig. 4a). Small populations

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 7: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

7  

of cells still retained the HSC (hematopoietic stem cell) signature (C9, C7, C4) or differentiated

toward other lineages (C6). Trajectory analysis revealed that CAR-iMac cells went through a

path from HSC to macrophage and DC cells without major branches (Fig. 3d).  

In order to determine whether each of these single cells or clusters are more polarized toward

the M1 or M2 state, we examined marker gene expression and found high and relatively

homogenous expression of M2 genes such as CD206 (Supplementary Fig. 4b). Moreover, all

10 clusters of differentiated cells showed strong signatures of “oxidative phosphorylation” and

“respiratory electron transport” which are usually associated with the M2 state, and weak

glycolysis or pro-inflammatory signatures associated with the M1 state16-19 (Fig. 3e). We further

compared cells in the 10 clusters with the LPS/IFN-γ-polarized M1 cells or IL-4/IL-10-

polarized M2 cells14, 15, by examining differentially expressed cytokine and chemokine genes

or M1/M2 associated metabolism genes (Fig. 3f, g ), and found that most clusters are more

similar to the M2 state, especially when using metabolism genes as markers. Together, these

data showed that in the absence of antigen, the first generation of CAR-iMac cells stayed in a

state closer to the classically defined M2 along the plastic macrophage spectrum20-23, likely

because of the presence of M-CSF during the differentiation process24. 

CAR-iMac Cells Show Antigen-dependent Functions When Co-cultured with Leukemia

and Lymphoma Cells in Vitro 

In order to test whether iPSC-derived CAR-expressing macrophages assumed the capability to

phagocytose tumor cells in an antigen-dependent manner, we incubated the CAR-iMac cells

labeled by a green dye with CD19-expressing K562 cells or K562 cells (K562 cell itself does

not express CD19) labeled by a red dye. Compared with K562 alone, K562-CD19 cells were

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 8: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

8  

more likely to be phagocytosed by CAR-iMac indicated by an increase from 51.9% to 76.4%

double positive cells which are the macrophages in the process of phagocytosing tumor cells

(Fig. 4a, b). Intracellular signaling such as phosphorylation of ERK and NF-κB P65 proteins

were increased in CAR-iMac co-cultured with CD19-expressing K562 cells compared to K562

cells, or to CAR-iMac cells cultured alone (Fig. 4c), indicating antigen engagement promotes

activation of macrophage intracellular signaling pathways.  

One important role of macrophages is to modulate the tumor microenvironment by producing

cytokines that either stimulate or suppress T cell activity9. In order to harness these features,

we first examined cytokine gene expression in CAR-iMac cells, and found antigen-dependent

increase of pro-inflammatory cytokine expression, such as IL1B, IL21, IL12A and IL6, when

CAR-iMac cells were incubated with CD19-expressing K562 cells (Fig. 4d), as well as antigen-

dependent increase of cytokine secretion in the culture medium such as IL-6 and IL-12

determined by ELISA from the same condition (Fig. 4e). Moreover, transcriptional analysis

showed that CAR-iMac cells co-cultured with K562-CD19, in comparison to those with K562,

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. 4f, g). We also examined another leukemia cell line Nalm6

expressing CD19, and consistently, the CAR-expressing iMac cells showed increased

production of pro-inflammatory cytokines and increased level of phosphorylation of ERK,

compared to CAR-iMac cells alone (Supplementary Fig. 5a, b), again demonstrating the role

of CAR-antigen engagement in conferring the stimulated macrophage activity.  

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 9: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

9  

We next investigated the effects of CAR-iMac or iMac cells on a CD19-expressing B cell

lymphoma cell line Raji. There is mild enhancement of phagocytosis conferred by the CAR

(Supplementary Fig. 5c, d). Moreover, CAR-iMac also had significantly increased pro-

inflammatory cytokine gene expression such as IL18, IL6 and IL1B, and other macrophage

function-specific genes compared to iMac without a CAR (Supplementary Fig. 5e). When we

examined intracellular signaling pathways, we found phosphorylation of AKT, SRC, STAT5

and ERK were all up-regulated when CAR-iMac cells encountered Raji cells (Supplementary

Fig. 5f). Taken together, the above data demonstrate that CAR-mediated signaling promotes

iMac phagocytosis and tuning toward the pro-inflammatory M1-like state upon encountering

the antigen on multiple types of leukemia and lymphoma cells (Fig. 4h).

CAR-iMac Cells Show Antigen-dependent Functions When Co-cultured with Solid

Tumor Cells in Vitro

To test iMac cells functions against solid tumors, we replaced the CD19 CAR scFv with a

mesothelin scFv in the lentiviral construct, and transduced the new construct to iPSCs followed

by differentiating them to CAR (meso)-iMac cells (Fig. 5a). We incubated the CAR (meso)-

iMac cells or control iMac cells with mesothelin-expressing OVCAR3 ovarian cancer cells.

Compared with control cells, CAR (meso)-iMac showed increased phagocytosis activity

against OVCAR3 (Fig. 5b, c). Moreover, transcriptome analysis showed that CAR (meso)-

iMac cells also had significantly up-regulated inflammatory and cytokine activity signatures

compared to the control when encountering OVCAR3 cells (Fig. 5d, e), which were validated

by increased pro-inflammatory M1 cytokine gene expression such as IL6, IL1A, IL1B, and TNF

(Fig. 5f). We also used a mesothelin-expressing pancreatic cancer line ASPC1, and found the

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 10: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

10  

same trends of up-regulated phagocytosis, M1 cytokine gene expression, and global

inflammatory signatures (Fig. 5g-j). Together, these data demonstrate CAR confers enhanced

phagocytosis and pro-inflammatory immune activities when CAR-iMacs are stimulated by

antigen-bearing solid tumor cells.

CAR-iMac Cells Show in Vivo Phagocytosis and Anti-Cancer Cell Function

In order to detect phagocytosis functions of CAR-iMac cells in vivo, we injected GFP-

expressing Raji cancer cells and CAR-iMac cells to NSG mice intravenously, and after three

days, the sorted human CD11b+ (from CAR-iMac cells) and GFP+ (from cancer cells) double

positive cells were examined with Giemsa staining (Fig. 6a). The macrophages showed multiple

nuclei, suggesting in vivo phagocytosis took place in the injected mice (Fig. 6a). To evaluate

the anti-cancer cell function of these CAR-iMac cells in vivo, we intraperitoneally injected

leukemia cells expressing a luciferase gene into NSG mice, and after tumors formed, mice

bearing comparable tumor loads were separated into two groups for treatment and controls. In

order to achieve better anti-cancer cell outcome, we further polarized the CAR-iMac cells

toward M1 by treating them with IFN-γ for 24 hours (Supplementary Fig. 6a), and then one

group of mice was injected intraperitoneally with the polarized CAR-iMac cells for treatment

(Fig. 6b). The treated mice showed smaller tumor size on day 10, with two mice having

complete remission (Fig. 6c). For solid tumors, we inoculated a high mesothelin-expressing

ovarian cancer cell line HO8910 intraperitoneally to NSG mice, followed by intraperitoneally

injecting IFN-γ-treated CAR (meso)-iMac cells (Fig. 6b). Two out of the five treated mice

showed tumor remission (Fig. 6d), and four out of five mice in the treated group lived for three

weeks but only two mice in the control group (Fig. 6d). The treatment did not work to the same

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 11: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

11  

extent in all mice, likely because not all the plastic iMac cells preserved in the same polarized

state in the complex and sometime immunosuppressive in vivo microenvironment in all mice.

Nevertheless, these data demonstrate CAR-iMac cells showed some anti-cancer cell functions

in vivo, but their efficiency needs to be further improved by designing more effective CARs or

further modifying the CAR-iMac cells to stay constitutively in M1 state. 

Discussion 

Recently, 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 therapies5, 6 A new member of this family, CAR-iMac

has three advantages. First, CAR-iMac can be utilized to specifically “eat” tumor cells or to

alter the tumor microenvironment, especially when its function is antigen-dependent, providing

a tool to directly kill tumor cells or to modulate a specific niche at the interface of tumor and

immune cells. Second, increasingly complicated genome engineering approaches can be

applied to iPSCs, followed by single clone expansion and thorough characterization to obtain a

clonal and genetically homogenous population with minimal off-target risk. Third, the seed

iPSC clone can be expanded in large scale, and when combined with the recently reported

techniques in making immune-tolerant universal iPSCs25-27, CAR-iMac can be developed into

an off-the-shelf product with unlimited sources. The last two points are particularly useful when

the effector cells from primary cell sources are difficult to engineer and to expand which is the

case for PBMC-derived macrophage cells. Thus, CAR-iMac is an excellent platform for

engineering-friendly and expandable macrophage cells, and a valuable addition to other iPSC

differentiated immune cells for further cancer immunotherapies. 

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 12: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

12  

There are several general challenges of using iPSC differentiated cells for clinical applications,

such as undifferentiated cell can form teratomas, yield of differentiation and heterogeneity of

differentiated cells. We have achieved high differentiation efficiency with a yield of ~10-100

myeloid cells per starting iPSC. Even though certain cells remained some gene expression

signature of stem/precursor cells, we have not seen teratomas formation when we transplanted

the bulk differentiated cells into mice. In terms of heterogeneity, differentiated cells collected

from the floating shedded cells are primarily positive for macrophage markers such as CD14

and CD11b (Fig. 1c). More careful examination with single cell RNA-sequencing data revealed

a clear trajectory of differentiation toward macrophages and dendritic cells (Fig. 3c), which by

themselves share overlapping transcriptome signatures and functions, and can be named

altogether as “mononuclear phagocyte system (MPS)” cells28. Notably, in CD19 CAR-T cell

therapies or immune checkpoint therapies, combinations of CD8+ and CD4+ T cells have

superior anti-cancer cell activity29-31. It is thus conceivable that MPS cells may achieve

synergistic effects from different subsets of cells by simultaneously phagocytosing tumor cells

or presenting antigens to T cells. Therefore, it will be important to compare the purified versus

mixed myeloid products for their efficacy, persistence and toxicity in future studies. It will also

be of high interest to use humanized mouse models to study the role of differentiated DC-like

cells in tumor antigen presentation and T cell stimulation.  

Another challenge to solve in order to take our findings to the clinic is to balance the anti-cancer

cell functions from pro-inflammatory cytokines and the toxicity induced by the same cytokines

such as IL-1 and IL-632, 33. In our system, we achieved antigen-dependent induction of these

cytokines, so that they can only be released in large amount within a restricted niche of antigen

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 13: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

13  

exposure. Indeed, our first generation of iMac cells were in a state closer to the immune-

suppressive M2 state and were tuned toward M1 after exposing to antigens, which suggests that

the cells may travel safely in the blood vessel, and may not become inflammatory until it

reaches to the tumor microenvironment (Fig. 4h). Another potential solution is to engineer

molecular switches to enable CAR or cytokine expression under control of endogenous

promoters34or exogenously administered small molecules35 so that inflammatory cytokine

expression is controlled by the cell state or controlled in a temporal way. Moreover, further

engineering the CAR intracellular domain to confer phagocytosis function without eliciting

inflammation -stimulating cytokines, such as using intracellular domain of FcγR36, or knocking

out endogenous inflammation-stimulating cytokine genes, are all possible solutions to explore.

Besides the safety issues, we explored the anti-tumor effect in animal models of both liquid and

solid tumors. The data showed that our first generation of CAR-iMac had some effect. While

our paper is being reviewed, Klichinsky et. al reported PBMC-derived macrophage transduced

with a CAR through adenovirus showed strong in vivo anti-tumor effect, and one key reason is

the adenoviral vector Ad5f35 used imparted a sustained pro-inflammatory M1 phenotype37.

Thus, there are many directions to improve the efficacy of our first generation of CAR-iMac

along this line. Our current iMac differentiation protocol contains M-CSF which is not only

required to support myeloid cell survival, but also unavoidably biases the differentiation toward

M2 state. Even though we added IFN-γ to polarize it to M1 state and CAR signaling tuned it

toward M1, but the epigenetic memory inherited may influence the ultimate state, particularly

for the plastic macrophage in the complicated in vivo microenvironment. Thus, a constitutive

M1 signaling is required to guarantee the cells to be constantly pro-inflammatory and anti-

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 14: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

14  

tumor. With the iMac system, this can be achieved by inserting certain M1 cytokine genes in

the genome of iPSC seed cells, or by further optimizing the intracellular domain of CAR to

confer M1 signaling, similar as the second signal provided by the “co-stimulatory” domain

from CD28 and 4-IBB in the second generation of CAR-T cells38, 39.  

Together, our data provided a proof-of-principle platform of iPSC-based, engineering-friendly

macrophage for adoptive transfer immune cell therapy, and suggest that further development

and optimization of CAR-iMac is warranted. Its kaleidoscopic applications in various contexts

of liquid and solid tumors, and in combination of genome engineering or in combination with

other types of immunotherapies are full of great promises for further immune cell therapies.

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 15: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

25  

References:

1. Rowe, R.G. & Daley, G.Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat Rev Genet 20, 377-388 (2019).

2. June, C.H. & Sadelain, M. Chimeric Antigen Receptor Therapy. The New England journal of medicine 379, 64-73 (2018).

3. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science (New York, N.Y.) 339, 823-826 (2013).

4. Wang, G. et al. Efficient, footprint-free human iPSC genome editing by consolidation of Cas9/CRISPR and piggyBac technologies. Nature protocols 12, 88-103 (2017).

5. Themeli, M. et al. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat Biotechnol 31, 928-933 (2013).

6. Li, Y., Hermanson, D.L., Moriarity, B.S. & Kaufman, D.S. Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity. Cell Stem Cell 23, 181-192 e185 (2018).

7. Condeelis, J. & Pollard, J.W. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124, 263-266 (2006).

8. Morrissey, M.A. et al. Chimeric antigen receptors that trigger phagocytosis. Elife 7 (2018).

9. Noy, R. & Pollard, J.W. Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49-61 (2014).

10. Gubin, M.M. et al. High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy. Cell 175, 1443 (2018).

11. Ayers, M. et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127, 2930-2940 (2017).

12. Pathria, P., Louis, T.L. & Varner, J.A. Targeting Tumor-Associated Macrophages in Cancer. Trends Immunol 40, 310-327 (2019).

13. Andreesen, R. et al. Adoptive transfer of tumor cytotoxic macrophages generated in vitro from circulating blood monocytes: a new approach to cancer immunotherapy. Cancer Res 50, 7450-7456 (1990).

14. Zhang, H. et al. Functional analysis and transcriptomic profiling of iPSC-derived macrophages and their application in modeling Mendelian disease. Circ Res 117, 17-28 (2015).

15. Douvaras, P. et al. Directed Differentiation of Human Pluripotent Stem Cells to Microglia. Stem Cell Reports 8, 1516-1524 (2017).

16. Sica, A. & Mantovani, A. Macrophage plasticity and polarization: in vivo veritas. The Journal of clinical investigation 122, 787-795 (2012).

17. Viola, A., Munari, F., Sanchez-Rodriguez, R., Scolaro, T. & Castegna, A. The Metabolic Signature of Macrophage Responses. Front Immunol 10, 1462 (2019).

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 16: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

26  

18. Galvan-Pena, S. & O'Neill, L.A. Metabolic reprograming in macrophage polarization. Front Immunol 5, 420 (2014).

19. Kelly, B. & O'Neill, L.A. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res 25, 771-784 (2015).

20. Mosser, D.M. & Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8, 958-969 (2008).

21. Xue, J. et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 40, 274-288 (2014).

22. Martinez, F.O. & Gordon, S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6, 13 (2014).

23. Martinez, F.O., Sica, A., Mantovani, A. & Locati, M. Macrophage activation and polarization. Front Biosci 13, 453-461 (2008).

24. Lachmann, N. et al. Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Reports 4, 282-296 (2015).

25. Deuse, T. et al. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat Biotechnol 37, 252-258 (2019).

26. Xu, H. et al. Targeted Disruption of HLA Genes via CRISPR-Cas9 Generates iPSCs with Enhanced Immune Compatibility. Cell Stem Cell 24, 566-578 e567 (2019).

27. Han, X. et al. Generation of hypoimmunogenic human pluripotent stem cells. Proc Natl Acad Sci U S A 116, 10441-10446 (2019).

28. Guilliams, M. et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 14, 571-578 (2014).

29. Sommermeyer, D. et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 30, 492-500 (2016).

30. Turtle, C.J. et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. The Journal of clinical investigation 126, 2123-2138 (2016).

31. Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 574, 696-701 (2019).

32. Giavridis, T. et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 24, 731-738 (2018).

33. Norelli, M. et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 24, 739-748 (2018).

34. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113-117 (2017).

35. Sakemura, R. et al. A Tet-On Inducible System for Controlling CD19-Chimeric Antigen Receptor Expression upon Drug Administration. Cancer Immunol Res 4, 658-668 (2016).

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 17: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

27  

36. Ruffell, B. & Coussens, L.M. Macrophages and therapeutic resistance in cancer. Cancer Cell 27, 462-472 (2015).

37. Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nature Biotechnology (2020).

38. Milone, M.C. et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Molecular therapy : the journal of the American Society of Gene Therapy 17, 1453-1464 (2009).

39. Maher, J., Brentjens, R.J., Gunset, G., Riviere, I. & Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol 20, 70-75 (2002).

40. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 32, 381-386 (2014).

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 18: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

28  

Figure legends

Fig. 1. Differentiation of CAR–iPSCs into macrophages. a, Schematics showing the

procedures of PBMC reprogramming, introduction of chimera antigen receptor and

differentiation to macrophage cells. b, Overview of the differentiation protocol to derive

macrophages from iPSCs. Representative microscopic pictures showing cells at different

stages during differentiation. The last picture is a magnified filed for matured macrophages

attached at the bottom of the plate at day 28. c, Flow cytometry analysis of iPSC-derived cells

at different stages of differentiation with stage specific markers. d, qRT-PCR showing

pluripotent marker gene and key macrophage marker gene expression at different stages of

differentiation. n=3, error bar: standard error of the mean.

Fig. 2. Transcriptional profiling of iPSC-derived CAR-iMac cells. a, Hierarchical clustering

of transcriptomes of iPSCs, differentiated cells, and primary and stem cell-differentiated

macrophages in different states. b, Principle component analysis (PCA) of the same samples as

in a. c, Top GO terms enriched in genes up-regulated on day 18 differentiated cells compared

with iPSCs. Right panel is an example of GSEA analysis of one GO term “Positive regulation

of cytokine production”. NES: normalized enrichment score. P=0: p-value is a very small

number. d, Top GO terms enriched in genes up-regulated on day 28 compared with iPSCs, and

GSEA analysis of “Positive regulation of cytokine production”.

Fig. 3. Single cell RNA-sequencing analysis shows CAR-iMac cells are closer to the M2 state

on the macrophage polarization spectrum. a, UMAP plot showing separation between human

iPSCs and CAR-iMac cells. b, UMAP plot showing sub-population clustering of CAR-iMac

cells. Ten clustered C0-C9 were identified and labeled as 0-9 with different colors. c, Heatmap

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 19: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

29  

showing blasting the C0-C9 clusters of cells illustrated in b against a human single cell atlas

database containing single cell RNA-seq data of hundreds of cell types including macrophages

(http://scibet.cancer-pku.cn). d, Trajectory analysis of differentiated cells along a pseudotime

axis. Left panel includes all clusters on the same trajectory map, and right panel is the

breakdown of each cluster on the same trajectory. The numbers on the trajectory mean branch

points or bifurcating events revealed by the trajectory analysis. MAC/DC means the cluster

matches both Macrophage and Dendritic cells. Cluster 6 is not assigned to a specific cell type.

e, Heatmap showing averaged expression of M1 or M2 signature pathway genes in different

clusters of cells illustrated in b. f, g, Heatmaps to compare (benchmark) the 10 clusters (C0-C9)

of CAR-iMac cells against previously published M1 or M2 polarized macrophages using either

top differentially expressed cytokine and chemoattractant genes f or using metabolism genes g

as signature genes. IPS_M1: Human iPS cells differentiated macrophages polarized by IFN-γ

and LPS; IPS_M2: Human iPS cells differentiated macrophages polarized by IL-4; HM_M1:

Human PBMC-derived macrophages polarized by IFN-γ and LPS; HM_M2: Human PBMC-

derived macrophages polarized by IL-4.

Fig. 4. CAR-iMac cells assume antigen- and CAR-dependent macrophage phagocytosis

function and pro-inflammatory signatures when co-cultured with liquid cancer cells. a,

Confocal microscopy pictures showing phagocytosis of K562 or K562-CD19 cells (red) by

CAR-iMac cells (green). iMac cells labeled by CMFDA were co-incubated with cancer cells

labeled by CTDR for 24 hours before imaging. b, Flow cytometry showing phagocytosis of

K562 or K562-CD19 cells by CAR-iMac cells. Labeled iMac cells were co-incubated with

labeled cancer cells and collected for FACS analysis. Double positive cells represent the iMac

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 20: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

30  

cells that engulfed cancer cells. c, Western blotting showing phosphorylation of ERK and NF-

κB P65 in CAR-iMac cells in the indicated conditions. d, qRT-PCR showing cytokine gene

mRNA expression when CAR-iMac cells were incubated with K562 or K562-CD19 cancer

cells for 24h, cancer cells were washed off before collecting CAR-iMac. n=3, error bar:

standard error of the mean. e, ELISA assay showing cytokine secretion in the medium when

CAR-iMac cells were incubated with K562 or K562-CD19 cancer cells. n=3, error bar: standard

error of the mean. **: p < 0.01, one-way ANOVA. f, Top GO terms enriched in genes up-

regulated in CAR-iMac cells co-cultured with K562-CD19 cells compared with CAR-iMac

cells co-cultured with K562 cells without the CD19 antigen. Right panel is GSEA analysis of

“positive regulation of cytokine production”. g, Top KEGG pathways enriched in genes up-

regulated in CAR-iMac cells co-cultured with K562-CD19 cells compared with CAR-iMac

cells co-cultured with CD19 cells. Right panel is GSEA analysis of “antigen processing and

presentation”. h, Schematic diagram showing CAR-iMac cells are more skewed toward M1-

like pro-inflammatory state upon encountering the antigen.

Fig. 5. iMac cells assume antigen- and CAR-dependent macrophage phagocytosis

functions and pro-inflammatory signatures when co-cultured with solid tumor cells. a, A

schematic picture to show the lentivirus construct of the CAR against mesothelin CAR (meso),

and the procedures to obtain CAR (meso)-iMac cells. b, Confocal microscopic images showing

phagocytosis of OVCAR3 cells (red) by iMac or CAR (meso)-iMac cells (green). c, Flow

cytometry showing phagocytosis of OVCAR3 ovarian cancer cells by iMac or CAR (meso)-

iMac cells. Labeled iMac cells were co-incubated with labeled cancer cells and collected for

FACS analysis. Squared parts represent the cancer cells engulfed by iMac cells. n=3. d, GO

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 21: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

31  

term analysis with RNA-seq data showing the up-regulated genes in CAR (meso)-iMac cells

compared with iMac cell when both cells were co-cultured with OVCAR3 cancer cells. e,

GSEA analysis of representative cytokine activity gene set showing it is up-regulated in CAR

(meso)-iMac cells compared with iMac cell when both cells were co-cultured with OVCAR3

cells. NES: normalized enrichment score. f, qRT-PCR showing cytokine gene mRNA

expression when iMac or CAR (meso)-iMac cells were incubated with OVCAR3 cells for 24h.

Cancer cells were washed off before collecting iMac or CAR-iMac cells. n=3. Error bar:

standard error of the mean. g, Flow cytometry showing phagocytosis of ASPC1 pancreatic

cancer cells by iMac or CAR (meso)-iMac cells. n=3. h, GO term analysis showing the up-

regulated genes in CAR (meso)-iMac cells compared with iMac cell when both cells were co-

cultured with ASPC1 cells. i, GSEA analysis of representative acute inflammatory response

gene set showing it is up-regulated in CAR (meso)-iMac cells compared with iMac cell when

both cells were co-cultured with ASPC1 cells. j, qRT-PCR showing cytokine gene mRNA

expression when iMac or CAR (meso)-iMac cells were incubated with ASPC1 cells for 24h.

Cancer cells were washed off before collecting CAR-iMac. n=3, error bar: standard error of the

mean.

Fig. 6. CAR-iMac cells showed in vivo phagocytosis function and anti-tumor effect. a,

Giemsa staining of FACS-sorted human CD11b and GFP double positive cells showing that

CAR-iMac cells phagocytosed Raji cells in vivo. 4x105 GFP-labeled cancer cells were

injected intravenously, and 6 hours later, 1x106 CAR-iMac were injected intravenously. At

day 3, cells were sorted for staining. Black arrows point to the multiple nucleus in the isolated

macrophages. b, A schematic picture of in vivo anti-tumor studies using luciferase

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 22: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

32  

(luc)-labeled CD19-expressing Nalm6 leukemia cells or mesothelin-expressing

HO8910 ovarian cancer cells in NSG mice treated with CAR-iMac cells. c,

Bioluminescent imaging pictures showing tumor development on the indicated days in mice

inoculated with Nalm6 leukemia cells and treated with iMac cells or PBS as described above.

d, Bioluminescent imaging pictures showing tumor development on the indicated days in

mice inoculated with HO8910 ovarian cancer cells and treated with iMac cells or PBS.

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 23: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

Fig. 1

a

b

diPSEB Day2EB Day7iMac Day18iMac Day28

C

CD45

CD34

CD11bCD11b

CD14 CD14

CD163

CD86

Blood cell marker

HSC marker

iPS cells

differentiated cells

Day 14 Day 45Day 14

101 102

0

20

40

60

80

100

103 104 105 106

Macrophage marker

101 102

0

20

40

60

80

100

103 104 105 106

101 102

0

20

40

60

80

100

103 104 105 106

101 102

0

20

40

60

80

100

103 104 105 106 101 102

0

20

40

60

80

100

103 104 105 106101 102

0

20

40

60

80

100

103 104 105 106

101 102

0

20

40

60

80

100

103 104 105 106 101 102

0

20

40

60

80

100

103 104 105 106

Mesoderm induction

Hematopoietic specification

Myeloid expansion

Macrophage maturation

Macrophagemaintainenance

20μm

D0 D2 D7 D20 D28

20μm20μm

TREM2

AIF1

CSF1RSPI1

0.1

1

10

100

1000

10000

100000

1000000

Rel

ativ

e ex

pre

ssio

n

LIN28L-MYC

CAR

Reprogramming Engineering Differentiation

Human PBMC iPSC CAR-iPSC CAR-iMac

KLF4 SOX2 OCT3/4

c

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 24: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

Day 18 differentiated cells vs iPS: up-regulated genes

Day 28 differentiated cells vs iPS: up-regulated genes

c

d

day 2

iPS

day 7

day 18

day 28

day 38

non−polarized macrophages

polarized with IFN−gamma and LPS

polarized with IL−4 and IL−13

polarized with IL−10

IPSDM M1

IPSDM M2

IPSDM Mac

p=0.0

positive regulation of cytokine production

NES=1.88

up-regulated at day 28 down-regulated at day 28

NES=1.78

p=0.0

positive regulation of cytokine production

up-regulated on day 18 down-regulated on day 18

pattern recognition receptor signaling pathway

positive regulation of cytokine production

response to molecule of bacterial origin

positive regulation of defense response

cytokine secretion

cellular response to lipopolysaccharide

regulation of immune effector process

regulation of innate immune response

toll-like receptor signaling pathway

regulation of inflammatory response

regulation of adaptive immune response

regulation of phagocytosis

-log10 p value

0 10 20 30

Macrophages polarized with IFN−gamma and LPS

Macrophages polarized with IL−4 and IL−13

day 2

iPS rep_1

iPS rep_2

iPS rep_3

day 7 rep_1

day 7 rep_2

day 7 rep_3

IPSDM Mac rep_3

IPSDM Mac rep_1

IPSDM Mac rep_2

IPSDM M2 rep_3

IPSDM M2 rep_2

IPSDM M2 rep_1

Non−polarized macrophages

day 18 rep_2

day 18 rep_1

day 28 rep_2

day 28 rep_1

day 38

IPSDM M1 rep_1

IPSDM M1 rep_2

IPSDM M1 rep_3

Macrophages polarized with IL−10

day 28 rep_3

CAR-iMac

iPS and precursor cells

a b

GO terms

GO terms

Fig. 2

pattern recognition receptor signaling pathway

0 10 20 30 40 50

-log10 p value

response to lipopolysaccharide

positive regulation of cytokine production

response to interferon-gamma

positive regulation of defense response

regulation of innate immune response

regulation of inflammatory response

I-kappaB kinase/NF-kappaB signaling

regulation of immune effector process

regulation of tumor necrosis factor production

antigen processing and presentation

regulation of toll-like receptor signaling pathway

iPS and precursor cellsCAR-iMac

PC1

PC

3

En

rich

men

t sc

ore

En

rich

men

t sc

ore

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 25: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

C0:MAC/DC C1:DC/MAC C2:DC/MAC C3:DC/MAC C4:HSC

C5:MAC/DC C6 C7:HSC C8:DC C9:HSC

-5 0 5 10

-5

0

5

10

UMAP1

UM

AP

2Fig. 3

M1 signatures

M2 signatures

a

b

c

gf

1.25

1.00

0.75

0.50

0.25

Skeletal muscle myoblasts

Foreskin fibroblast cells

Hepatocytes

Embryonic stem cells

Muscle

Monocyte

Microglia

Mast

Macrophage

ILC

HSCsHEK and 3T3 mix

H9 cells

Epithelial

Endothelial

DC

0.75

0.50

0.25

0.00

C1 C2 C3 C4 C5 C6 C7 C8 C9C0

HM

_M

2

IPS

_M1_rep3

HK2NDUFB3SDHANDUFV1COX5BCOX7CNDUFA5COX11COX18SDHBNDUFB2NDUFB9NDUFB6COX6NDUFS7LDHBNDUFA7NDUFS6UQCRQUQCRC1UQCRBCOX4I1LDHAUQCRHCOX20NDUFAF2COX8ANDUFB8NDUFS8COX6A1NDUFS5NDUFA11UQCR10COX7A2COX6B1NDUFV2NDUFB7

HM

_M

1

IPS

_M1_rep5

IPS

_M1_rep1

IPS

_M

1_

rep

2

IPS

_M

1_

rep

4

C9

C6

C4

C7

C8

C2

C1

C3

C0

C5

IPS

_M

2_

rep

2

IPS

_M

2_

rep

3

IPS

_M

2_

rep

1

IPS

_M

2_

rep

5

IPS

_M

2_

rep

4

2

1

0

-1

-10

UM

AP

2

-10 0 10

0

5

10

-5

UMAP1

Oxidative phosphorylation

Respiratory electron transport

Glycolysis / Gluconeogenesis

Hallmark TNF-α signaling via NF-κB

Hallmark inflammatory response

0 1 3 4 5 6 7 8 92

Hallmark interferon gamma response

Clusters

6

4

2

0

-2

-4

-6

IL18CCL15IL2RAIL15TNFSF10CD80IL6CD7CD38

IL4I1TNFSF13B

IL27IL23ACXCL11CD48CD82CCL8CXCL10CXCL2IL15RAIL1RNCD40TNFAIP2CXCL9CD83CD300EIL32TLR8IL1AIL1BIL10CD1ECD36CCL26CD209TNFRSF11ATREM2CD200R1CD93CD180CSF1RCCL13IP

S_

M1

_re

p4

HM

_M

1

IPS

_M

1_

rep

3

IPS

_M

1_

rep

5

IPS

_M

1_

rep

1

IPS

_M

1_

rep

2

HM

_M

2

IPS

_M

2_

rep

4

IPS

_M

2_

rep

2

IPS

_M

2_

rep

3

IPS

_M

2_

rep

1

IPS

_M

2_

rep

5

C4

C7

C6

C9

C1

C3

C0

C5

C2

C8

CCL19

e

C0:MAC/DC C1:DC/MAC

C2:DC/MAC C3:DC/MAC

C4:HSC C5:MAC/DC

C6 C7:HSC

C8:DC C9:HSC

d

C0C1C2C3C4C5C6C7C8C9

HM

_M

2

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 26: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

NES=1.41

p=0.0

positive regulation of cytokine secretion

NES=1.66

p=0.0

antigen processing and presentation

ba

dc

Fig. 4

f

g

e

CAR-iMac Cancer cell Merge

K562

K562-CD19

20μm

p-ERK

p-P65

GAPDH

CAR-iMac

positive regulation of cytokine production

pattern recognition receptor signaling pathway

positive regulation of cell motility

cell chemotaxis

positive regulation of defense responsepositive regulation of cytokine secretion

activation of innate immune response

toll-like receptor signaling pathway

chemokine-mediated signaling pathway

regulation of inflammatory response

I-κB kinase/NF-κB signaling

positive regulation of GTPase activity

-log10 p value

0 5 10 15

CAR-iMac with K562-CD19 vs CAR-iMac with K562: up-regulated genes

GO terms

KEGG terms

CAR-iMac with K562-CD19 vs CAR-iMac with K562: up-regulated genes

pg

/ml

IL-6

IL-1

2

02468

101000

1500

2000

2500**

**

IL23

CCL2

CCL13

IL12

ATNF

IL15 IL

6IL

1B

0

2

4

6

8102030405060

Rel

ativ

e ex

pre

ssio

n

CAR-iMac+K562CAR-iMac+K562-CD19

0 2 4 6

NOD-like receptor signaling pathway

Chemokine signaling pathway

NF-kappa B signaling pathway

C-type lectin receptor signaling pathway

Fc gamma R-mediated phagocytosis

Phagosome

Cytokine-cytokine receptor interaction

Antigen processing and presentation

-log10 p value

Can

cer

cell

K562 K562-CD19

CAR-iMac

51.9% 76.4%

M1 M2 M1 M2

h

En

rich

men

t sc

ore

En

rich

men

t sc

ore

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 27: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

iMac OVCAR3 Merge

iMac

iMac(meso)

20μm

20.9± 0.3 27.3± 1.4

iMac

OV

CA

R3

iMac CAR (meso)-iMac

7.9± 0.7 12.8± 0.6

iMac

AS

PC

1

iMac CAR (meso)-iMac

TNFIL

1BIL

1A IL6

0

2

4

6

Rel

ativ

e ex

pre

ssio

n

iMac+OVCAR3iMac(meso)+OVACR3

IL12

BIL

1BIL

1A IL6

TNF

0

5

10

15

Rel

ativ

e ex

pre

ssio

n

iMac+ASPC1iMac(meso) +ASPC1

a

c f

d e

0 10 20 30 40 50-log10 p value

immune response

inflammatory responseresponse to cytokine

regulation of defense response

immune effector process

response to lipopolysaccharide

cellular response to chemokinecytokine secretion

response to interferon-gamma

cytokine-mediated signaling pathway

0 10 20 30 40 50-log10 p value

immune response

immune effector process

defense response

inflammatory response

cytokine-mediated signaling pathway

cytokine secretion

cellular response to chemokine

chemokine-mediated signaling pathwayregulation of MAPK cascade

response to interferon-gamma

acute inflammatory response

NES=1.58

NES=1.54

cytokine activity

g j

h i

CAR-iMac with OVCAR3 vs iMac with OVCAR3: up-regulated genes

CAR-iMac with ASPC1 vs iMac with ASPC1: up-regulated genes

Fig 5b

En

rich

men

t sc

ore

En

rich

men

t sc

ore

iMac + OVCAR3CAR (meso)-iMac + OVCAR3

iMac + ASPC1CAR (meso)-iMac + ASPC1

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint

Page 28: Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells ... · 3/28/2020  · 1 Induced Pluripotent Stem Cell-derived CAR-Macrophage Cells with Antigen-dependent Anti-Cancer Cell

a

Fig. 6

cPBS CAR-iMac

Day -1

Day 3

Day 7

Day 10

Color ScaleMin=1.72e6Max=3.77e7Radiance(p/sec/cm2/sr)

Raji CAR-iMac Phagocytosing CAR- iMac

10 μm

b

Day -4

Inject Luc+ Nalm6 cells (4×105) I.P.

-1 day

+ IFN-γ(100 ng/ml)

Day 0

Inject CAR-iMac (1.2×107) I.P.

Day 3 Day 7 Day 10

Day -4Inject Luc+ HO8910 cells (7×105) I.P.

-1 day

+ IFN-γ(100 ng/ml)

Day 0

Inject CAR-iMac (3.5×106) I.P.

Day 3 Day 6 Day 10 Day 13 Day 17

Day 0 0h

I.V. GFP+ Tumor I.V. iMac

Day 0 6h

Sort human CD14+ / GFP+ iMac cells for Giemsa staining

Day 3

d

Day -1

Day 13

Day 3

Day 10

Day 17

Day 6

PBS CAR(meso)-iMac

(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. The copyright holder for this preprintthis version posted March 29, 2020. ; https://doi.org/10.1101/2020.03.28.011270doi: bioRxiv preprint