You are on page 1of 5

Biomedicine & Pharmacotherapy 139 (2021) 111605

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

CAR-macrophage: A new immunotherapy candidate against solid tumors


Yizhao Chen a, Zhiying Yu c, Xuewen Tan a, Haifeng Jiang a, Zhen Xu a, Yilong Fang a, Dafei Han a,
Wenming Hong b, Wei Wei a, *, Jiajie Tu a, c, **
a
Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative
Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
b
Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, China
c
Department of Gynecology, The First Affiliated Hospital of Shenzhen University, Health Science Center; Shenzhen Second People’s Hospital, Shenzhen, China

A R T I C L E I N F O A B S T R A C T

Keywords: Chimeric antigen receptor (CAR)-T cell therapy has been shown to be an effective treatment for hematological
CAR-M tumors, but the treatment of solid tumors still lacks effectiveness. In the tumor microenvironment, macrophages
Phagocytosis are the innate immune cells with the highest infiltration rate. Tumor-associated macrophages (TAMs) stimulate
Solid tumor
angiogenesis, increase tumor invasion, and mediate immunosuppression. Because macrophages can infiltrate
CAR-T
CAR-NK
solid tumor tissue and interact with almost all cellular components in the tumor microenvironment (including
Immunotherapy tumor cells, immune cells such as T-cells, NK cells, DCs, and other resident non-immune cells), researchers are
trying to use macrophages modified with CAR (CAR-M) against solid tumors. This review describes recent reports
of CAR-M-based tumor treatments and summarizes their shortcomings and future applications.

1. Introduction malignancies [5,6]. After the success of CAR-T therapy, some re­
searchers applied CAR strategy to NK cells and developed CAR NK cell
Tumor immunotherapy is developed on the basis of the study of therapy, which is similar to CAR-T therapy. They all kill tumor cells
tumor immune escape, by manipulating the immune system to reac­ thruough CAR targeting, the difference is that this therapy relies on the
tivate the anti-tumor immune response and overcome the pathway of broad cytotoxicity and rapid killing ability of NK cells [7]. However, due
tumor immune escape [1]. The purpose of tumor immunotherapy is to to the high complexity of the tumor microenvironment in solid tumors,
modify immune cells in order to restore their ability to attack cancer CAR-T cell-mediated immunotherapy has shown low success rates [8].
cells [2]. The main immunotherapies which are currently available are Currently, tumor immunotherapy has mainly focused on adaptive im­
chimeric antigen receptor (CAR)-T cell therapy, immune checkpoint mune systems, such as T cells and B cells [9,10]. However, innate im­
inhibitor and cancer vaccines [3]. CAR-T cell therapy is a cellular mune cells, such as macrophages, have not been widely studied in
immunotherapy proposed earlier, which is designed to avoid tumor cells cancer treatment [11]. Taking into account their capabilities of phago­
escaping TCR(T cell receptor) recognition. It refers to the method of cytosis, antigen presentation and penetration in the tumor microenvi­
transferring genetic material with specific antigen recognition domain ronment, macrophages should be considered in the treatment of solid
and T cell activation signal into T cells by gene cloning technology, so cancer [12,13].
that T cells can be directly activated by binding with specific antigen on The two main cell types of mononuclear-phagocyte systems (MPS)
the surface of tumor cells [4]. CAR-T cells have been successfully used to are monocytes and monocyte-derived macrophages, both of which have
treat malignant tumors in the circulatory system, such as B cell-source phagocytic functions [14]. MPS selectively devour external targets, such

Abbreviations: CAR, chimeric antigen receptor; TAMs, tumor-associated macrophages; NK, natural killer; DCs, dendritic cells; M, macrophages; TCR, T cell re­
ceptor; MPS, mononuclear-phagocyte systems; IL, interleukin; TGF, transforming growth factor; CAR-P, chimeric antigen receptor-phagocytes; iPSCs, induced
pluripotent stem cells; ECM, extracellular matrix; MMPs, matrix metalloproteinase; TIMPs, tissue inhibitors of metalloproteinases; HER2, human epidermal growth
factor receptor 2; INF, interferon; CRS, cytokine release syndrome; PBMCs, peripheral blood monouclear cells; GVHD, graft versus host disesse; NR, not reported.
* Correspondence to: 81# Meishan Road, Shushan District, Hefei City, Anhui Province, China.
** Corresponding author at: Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of
Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China.
E-mail addresses: wwei@ahmu.edu.cn (W. Wei), tujiajie@ahmu.edu.cn (J. Tu).

https://doi.org/10.1016/j.biopha.2021.111605
Received 22 December 2020; Received in revised form 30 March 2021; Accepted 12 April 2021
Available online 23 April 2021
0753-3322/© 2021 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Y. Chen et al. Biomedicine & Pharmacotherapy 139 (2021) 111605

as microorganisms, rather than devouring the body’s own cells or the effect. This suggests that macrophages need additional signals to guide
extracellular matrix [15]. However, because of the mutated genome and their activity to fight tumors [23]. It was later discovered that in addi­
powerful "domestication" ability of tumor cells, macrophages are unable tion to the positive signals for macrophage phagocytosis, there were
to identify and attack tumors. Even within the tumor tissues, macro­ opposite signals that prevented macrophages from devouring the target
phages do not play a role in phagocytosis or antigen presentation, but tumor cells [24]. Self-labeling is common in cancer cells and is used as a
instead transition into an immunosuppressive M2 type that prevents the general mechanism to avoid being swallowed by macrophages. Labels
immune system from removing tumor cells [16]. M2 macrophages can include the "don’t eat me" signal between CD47 and the macrophage
repair tissue damage and build new blood vessels, and are also known as membrane receptor SIRPα [25]. These signals have become a popular
alternative active macrophages. Secreted cytokines (such as interleukin research area since their discovery.
[IL]− 6 and transforming growth factor [TGF]-β) and the low-oxygen Given the current success of CAR-T cell therapy and the potential for
environment of tumor tissues lead macrophages to mistakenly recog­ development of CAR-natural killer cells, researchers have paid great
nize them as tissues which need to be repaired. Macrophages subse­ interest in developing CAR-macrophages (CAR-M) for tumor immuno­
quently start repairing tumor tissues, building new blood vessels which therapy. The emergence of CAR-M opens up a new possibility for
lead to the further development of cancer [17,18]. In addition to treating solid tumors: modifying human macrophages with specific
phagocytosis, the high plasticity and polarization of macrophages are CARs to improve phagocytic activity and antigen presentation of mac­
also worthy of attention. Diversity and plasticity are significant char­ rophages against tumors [26]. CAR-M therapy refers to the transfer of
acteristics of monocyte macrophage lineage cells [19]. Macrophages the edited specific CAR gene into macrophages in order to equip them
produce different phenotypes in different tissues to different microen­ with the ability to bind to the tumor cell surface via specific antigen
vironment stimuli and signals, and may undergo M1 or M2 activation. In identification and subsequently activate macrophage activity against
immune response, macrophage polarization can reflect Th1-Th2 polar­ tumor cells [27].
ization of T cells [20]. Moreover, macrophages polarized into M1 or M2
phenotypes can be reversed to some extent. In short, M1 phenotype is 2. Current attempts
highly expressed in inflammatory cytokines and has strong anti micro­
bial and tumor activity, while M2 is considered to promote tissue The current attempts to use CAR-M to treat cancer are quite diverse
remodeling and tumor growth [21]. It is of great significance to reverse (Fig. 1 and Table 1). Researchers have designed chimeric antigen
the M1-M2 phenotype in the treatment of tumor. receptor-phagocytes (CAR-P) which can guide macrophages to devour
Attempts to combat cancer using macrophages began several de­ specific targets. They demonstrated that CAR-P, which express the
cades ago [22]. However, the evaluation of efficacy in these early clin­ intracellular domain of Megf10 or FcRv, promote the phagocytic po­
ical trials showed that macrophage transplantation therapy had little tential of target antigens. A series of experiments have shown that CAR-

Fig. 1. Structure diagram of CAR-M (A) Schematics show the structure of CAR-P. An αCD19 (orange) scFv directs CAR specificity, Intracellular signaling domains
from Megf10(green), FcRγ(blue) or tandem connection of fcrv and p85 subunit of PI3K(orange) activate engulfment. All constructs include a transmembrane domain
from CD8 and a C-terminal GFP. (B) An αHER2(purple) scFv directs CAR specificity, Intracellular signaling domains from CD3ζ(wathet blue) activate engulfment. (C)
CAR-147 is composed of a single-chain antibody fragment targeting human HER2, the hinge region of mouse IghG1, and the transmembrane and intracellular regions
of the mouse CD147 molecule. The HER2-scFv sequence was designed by Sangon Biotech based on reports in the NCBI database (PDB: 3H3B_D). (D) Schematics show
the structure of CAR-iMac, An αCD19 (orange) scFv directs CAR specificity, Intracellular signaling domains from FcγRI(grey) or CD3ζ activate engulfment. Compared
with FcγRI, the expression and secretion of cytokines in CD3ζ is higher.
All strucutures are modified from [22].

2
Y. Chen et al. Biomedicine & Pharmacotherapy 139 (2021) 111605

Table 1
Summary of the common CAR constructs used in CAR-M cell.
Target antigen Target cells CAR construction Macrophage source Reference

CD19 K562 cell CD19 + 4–1BB+CD3ζ induced pluripotent stem cells (iPSC) Li Zhang et al. [29]
HER2 SKOV3 ovarian cancer cell line HER2 + CD3ζ Primary human monocytes Michael Klichinsky et al. [32]
HER2 4T1 cells overexpressing human HER2-EGFP HER2 +CD147 Raw264.7 macrophages Wenlong Zhang et al. [31]
CD19 CD19 cancerous Raji B cells CD19 + Megf10 J774A.1 murine macrophage Meghan A Morrissey et al. [28]
CD19 + FcRV
CD19 +FcRV+PI3K

Table 2
CAR-M-based clinical trials.
NO.NCT Star year Stage Tumors M source CAR structure Gene transfer

03608618 2018 I Advanced Ovarian and Peritoneal Mesothelioma PBMC(including T cell, NK cell and monocyte) NR mRNA transfection
04660929 2020 I HER2 Overexpressing Solid Tumors Primary human macrophages NR Adenovirus
transfection

The data used in the table are from https://clinicaltrials.gov/.

PMegf10 specifically triggered the phagocytic effect of the target ligand. After CAR-147 was co-cultured with HER2 + human breast cancer cells,
During the process of phagocytosis, local signaling cascades initiate the expression of multiple MMPs in CAR-147 macrophages was induced,
phagocytosis via tyrosine phosphorylation. The TCR-CD3ζ chain may proving that CAR-147 can specifically identify the antigen HER2 and
promote the phagocytic effect of CAR-P through the recruitment of syk effectively activate the expression of MMP in macrophages. CAR-147
kinase. Whole-cell phagocytosis is rare, while biting or nibbling of target macrophages did not inhibit tumor cell proliferation in vitro, but
cells is more frequent, suggesting that the interaction between CAR-P intravenous injection of CAR-147 macrophages significantly inhibited
macrophages and target cells is not sufficient to trigger direct whole- tumor growth in 4T1 breast cancer mouse models. It was also found that
cell phagocytosis. Previous studies have found that PI3K signals play the proportion of T cells in tumors treated with CAR-147 macrophages
an important role in the internalization of large targets and promote was much higher than that in tumors treated with controlled macro­
phagocytosis by macrophages. Researchers have linked the PI3K p85 phages, demonstrating that CAR-147 macrophages can destroy the tu­
subunit to CAR-P-FcRv to form a "tandem" CAR (CAR-Ptandem). CAR- mor’s extracellular matrix and promote T cell infiltration into tumors. In
Ptandem has better whole-cell phagocytosis, implying that assembling addition, it was found that CAR-147 macrophages can significantly in­
a motif of phagocytic effectors could increase the whole-cell phagocytic crease the levels of IL-12 and IFNγ in tumor tissue, thus playing an anti-
activity of CAR-P [28]. tumor role [31].
Zhang et al. [29] used induced pluripotent stem cells (iPSCs) to ex­ Researchers at the University of Pennsylvania have designed
press CAR structure and differentiate into macrophages, named adenovirus-induced CAR-M using an anti-HER2 CAR containing the
CAR-iMac. Experiments have shown that iPSCs, which express CAR, can CD3ζ intracellular domain. First, the specificity of antigen-specific
differentiate into macrophage-like cells. In the absence of antigen, phagocytosis of HER2 + tumor cells was validated in vitro. In two
CAR-iMac is closer to the M2 polarization state. However, in the pres­ ectopic mice models of solid tumor transplantation, a single injection of
ence of specific antigens, such as leukemia and lymphoma cells, anti-HER2 CAR-M reduced tumor load and prolonged the survival time
CAR-mediated signals promote phagocytosis in CAR-iMac and lead to of mice. In a humanized mouse model, it was also found that HER2 CAR-
transition of CAR-iMac to the inflammatory M1 subtype. Moreover, M transduced by adenovirus was able to transform M2 macrophages into
stimulation by solid tumor cells expressing antigens also promoted the M1 macrophages, induce an inflammatory tumor microenvironment,
phagocytosis and immune activity of CAR-iMac. Although CAR-iMac and enhance anti-tumor cytotoxicity of T-cells. In addition, it was also
exhibits anti-tumor activity, its efficiency can be further improved by found that HER2 CAR-M may produce epitope diffusion, which provides
designing a more effective CAR structure or gene modification to keep new ideas for avoiding tumor immune escape [32,33].
them closer to the M1 state. In general, iPSC-derived macrophages may
become an important source of cells for myeloid-based cancer 3. The clinical applications of CAR-macrophages for the
immunotherapy. treatment of solid tumors
An important reason why CAR-T is not effective for solid tumors is
that T cells have difficulty entering into the tumor tissues. This is Until November 2020, two clinical trials based on the CAR-M strat­
because the physical barriers formed by the extracellular matrix (ECM) egy have been approved by the FDA. The first is a drug candidate from
of solid tumor and the consequent high tissue pressure prevent T cells CARISMA Therapeutics, CT-0508, which treats tumor patients with
from entering the tumor tissue. ECMs are produced by highly organized relapsed/refractory HER2 over-expression with anti-HER2 CAR macro­
fiber molecules, glycoproteins, and other large molecules. Its synthesis phages (Phase I clinical trial). The study recruited 18 patients with HER2
and degradation are mainly regulated by matrix metalloproteinase overexpressed solid tumors for the first time to study the effects of
(MMPs) and tissue inhibitors of metalloproteinases (TIMPs). MMP de­ adenovirus transduction CAR-M in humans.The other is MaxCyte’s
grades almost all ECM and substrate membranes, and TIMP is an MCY-M11, which uses mRNA-targeted PBMCs(including CAR-M) to
important enzyme family that inhibits MMP activity. Macrophages are express mesothelin-CAR, treating patients with relapsed/refractory
an important source of MMP, and studies have shown that injecting bone ovarian cancer and peritoneal mesothelioma. Volunteers were recruited
marrow-derived macrophages into mice can significantly reduce for Phase I clinical trial (Table 2).
cirrhosis and improve liver function [30]. As for the clinical translation of CAR-M, there are several aspects that
Researchers designed a CAR, named CAR-147, consisting of a single- need attention: first, the safety and effectiveness of CAR-M is the basis of
strand antibody fragment targeting human HER2, a hinger of IghG1, and this therapy, although it has been verified by animal experiments, the
a trans-membrane and intracellular region of CD147 molecules in mice. safety and effectiveness of CAR-M in human body still need to be

3
Y. Chen et al. Biomedicine & Pharmacotherapy 139 (2021) 111605

verified; second, reliable source and expansion are the necessary con­ but it also has many shortcomings to be overcome. Therefore, attention
ditions for the clinical application of CAR-M, which can be prepared by should be given to maximize the effectiveness and safety of CAR-M in
PBMC or by iPSCs. In addition, unlike T cells, macrophage has a low risk clinical treatment in the future. At first, the original structure of CAR
of GVHD, which means that products can be made in advance for pa­ was designed for T cells, but there is no CAR structure specifically for
tients to use on demand.Third, there is also a problem that has to be macrophages. However, in CAR-NK related studies, it has been proved
considered. At present, CAR-M gene transfer using viral transfection, that the location of car binding epitope and its distance from the cell
which may induce insertion mutations and have an uncontrollable effect surface affect antigen binding and cell activation [43]. Therefore,
on treatment. CRISPR/Cas9 genome targeting system provides a new starting from the structure of CAR-M, we can consider adjusting the
possibility to solve this problem, which takes only a week to complete structure of car by associating different domains, to explore the most
CAR-T gene editing [34]. suitable car structure for macrophages to enhance the phagocytic effect
of CAR-M on cancer cells. Secondly, by using genetic engineering
4. Discussion and prospective technology, CAR-M can also express anti-tumor cytokines or
co-stimulators to enhance antigen-presenting ability. The development
Similar to CAR-T and CAR-NK cells, CAR-M cells consist of extra­ of CAR-M exerts other anti-tumor functions, such as secreting MMPs to
cellular signaling domains that identify specific tumor antigens, trans- degrade the extracellular matrix or inducing epitope spreading capa­
membrane regions, and intracellular domains. At present, the study of bility [44]. Thirdly, combining CAR-M therapy with other immuno­
the extracellular signal domain identifies several common tumor targets, therapy is also a potential attempt. By blocking the "don’t eat me" signal
such as CD19 and HER2. CAR-M research focuses on the activation and CD47-SIRPα may enhance CAR-M’s phagocytosis of targeted cancer
enhancement of the phagocytic effect via different intracellular do­ cells. In addition, for patients with heavy burden of solid tumors, it’s
mains. Beyond the structure of CAR-M, the method to transfer the CAR worth to try to combine CAR-M with CAR-T. CAR-M can repress the
gene also needs to be considered, as researchers at the University of release of cytokines throughout the treatment process, which may
Pennsylvania have identified the M1 polarization induced by the reduce the risk of CRS and neurotoxicity from CAR-T therapy [45].
chimeric adenovirus vector Ad5f35. Finally, the mechanisms and regulatory factors that affect phagocytosis
Unlike CAR-T cells, CAR-M cells present the following three advan­ and antigen-presenting of macrophages, such as the physical properties
tages. 1) While T cells cannot enter the tumor environment because of of the phagocytic target, should be further clarified [46].
the physical barriers formed by the matrix around the tumor cells,
macrophages can significantly immerse in the tumor environment [35]. Funding
TAM plays an essential role in tumor invasion, metastasis, immuno­
suppression, and angiogenesis [36]. CAR-M can reduce the ratio of TAM This study was supported by the National Natural Science Founda­
and affect the cellular phenotype of TAM, which has positive effects on tion of China (31900616, 81673444, 82003795), Natural Science
cancer treatment. 2) In addition to the phagocytosis of tumor cells, Foundation of Anhui Province for young scholars (1908085QH379),
CAR-M is also able to promote the ability of antigen presentation and China Postdoctoral Science Foundation (2020M672831), Sanming
enhance the cytotoxic effects of T cells. 3) Compared to CAR-T, CAR-M Project of Medicine in Shenzhen (SZSM201812041) and Clinical
has a limited time in circulation and less non-tumor toxicity [37]. Research Funding from Shenzhen Second People’s Hospital (4001023).
Cytokine release syndrome (CRS) is an inflammatory syndrome caused
by multiple cytokines that are released by CAR-T cells, including IL-1,
Author contributions
IL-2, IL-4, IL-6, IL-8, IL-10, and tumor necrosis factor (TNF) α [38].
However, some studies have suggested that CRS is also related to IL-6 YC and ZY drafted the manuscript. XT, HJ, ZX, YF, DH, WH, WW and
released by macrophages. Interestingly, while investigating CAR-M, JT revised the manuscript.
two different results were found: IL-6 levels in mice treated with
CAR-147 macrophages were lower than in the control group, while
CAR-iMac cells showed an increase in the production of IL-6 in an Conflict of interest statement
antigen-dependent manner [39]. This may be related to the structure
and import method of CAR-M, or the different tumor types, which re­ The authors have declared no conflicts of interest.
quires further study.
Although CAR-M has great potential to become a powerful cancer Data availability
immunotherapy, many problems need to be overcome in order to ach­
ieve the desired results. The first is the limitation number of cells: No new data generated.
macrophages do not proliferate either in vitro or after injection in vivo.
Patients can accept macrophages only in limited amounts, which may
References
affect the effectiveness of treatment [39]. The second is related to the
migration characteristics of macrophages in vivo. After injection, [1] L.B. Kennedy, A.K.S. Salama, A review of cancer immunotherapy toxicity, CA
exogenous macrophages pass through the lung, and then most of them Cancer J. Clin. 70 (2) (2020) 86–104.
remain in the liver, which is not conducive to the treatment of cancer [2] J. Lai, S. Mardiana, I.G. House, K. Sek, M.A. Henderson, L. Giuffrida, A.X.Y. Chen,
K.L. Todd, E.V. Petley, J.D. Chan, E.M. Carrington, A.M. Lew, B.J. Solomon, J.
[40]. The third is the complex tumor microenvironment. Although A. Trapani, K. Kedzierska, M. Evrard, S.J. Vervoort, J. Waithman, P.K. Darcy, P.
CAR-M has achieved good results in the mouse model, the actual tumor A. Beavis, Adoptive cellular therapy with T cells expressing the dendritic cell
microenvironment in humans is much more complex than the animal growth factor Flt3L drives epitope spreading and antitumor immunity, Nat.
Immunol. 21 (8) (2020) 914–926.
model [41]. Finally, due to the high heterogeneity of tumor cells, the [3] Y. Yu, J. Cui, Present and future of cancer immunotherapy: a tumor
expression of target antigens may not be sufficient. This problem has microenvironmental perspective, Oncol. Lett. 16 (4) (2018) 4105–4113.
been very prominent in CAR-T therapy. Clinical studies have found that [4] A.D. Fesnak, C.H. June, B.L. Levine, Engineered T cells: the promise and challenges
of cancer immunotherapy, Nat. Rev. Cancer 16 (9) (2016) 566–581.
most of the tumor cells removed by CAR-T cells have high expression
[5] A. Jin, J. Feng, G. Wei, W. Wu, L. Yang, H. Xu, Y. Zhang, J. Cui, A.H. Chang, Y. Hu,
levels of the target antigen [42]. Predictably, this will also be a major H. Huang, CD19/CD22 chimeric antigen receptor T-cell therapy for refractory
obstacle to the development of CAR-M therapy. acute B-cell lymphoblastic leukemia with FLT3-ITD mutations, Bone Marrow
As mentioned above, CAR-M therapy has shown its effective anti- Transpl. 55 (4) (2020) 717–721.
[6] R. Havard, D.M. Stephens, Anti-CD19 chimeric antigen receptor T cell therapies:
tumor ability in animal experiments. Compared with CAR-T and CAR- Harnessing the power of the immune system to fight diffuse large B cell lymphoma,
NK, CAR-M has its unique advantages as a new cell immunotherapy, Curr. Hematol. Malig. Rep. 13 (6) (2018) 534–542.

4
Y. Chen et al. Biomedicine & Pharmacotherapy 139 (2021) 111605

[7] K. Rezvani, R. Rouce, E. Liu, E. Shpall, Engineering natural killer cells for cancer [31] W. Zhang, L. Liu, H. Su, Q. Liu, J. Shen, H. Dai, W. Zheng, Y. Lu, W. Zhang, Y. Bei,
immunotherapy, molecular therapy, J. Am. Soc. Gene Ther. 25 (8) (2017) P. Shen, Chimeric antigen receptor macrophage therapy for breast tumours
1769–1781. mediated by targeting the tumour extracellular matrix, Br. J. Cancer 121 (10)
[8] M. Martinez, E.K. Moon, CAR T cells for solid tumors: new strategies for finding, (2019) 837–845.
infiltrating, and surviving in the tumor microenvironment, Front. Immunol. 10 [32] M. Klichinsky, M. Ruella, O. Shestova, X.M. Lu, A. Best, M. Zeeman, M. Schmierer,
(2019) 128. K. Gabrusiewicz, N.R. Anderson, N.E. Petty, K.D. Cummins, F. Shen, X. Shan,
[9] P. Kosti, J. Maher, J.N. Arnold, Perspectives on chimeric antigen receptor T-cell K. Veliz, K. Blouch, Y. Yashiro-Ohtani, S.S. Kenderian, M.Y. Kim, R.S. O’Connor, S.
immunotherapy for solid tumors, Front Immunol. 9 (2018) 1104. R. Wallace, M.S. Kozlowski, D.M. Marchione, M. Shestov, B.A. Garcia, C.H. June,
[10] R.Y. Alhabbab, Targeting cancer stem cells by genetically engineered chimeric S. Gill, Human chimeric antigen receptor macrophages for cancer immunotherapy,
antigen receptor T cells, Front. Genet. 11 (2020) 312. Nat. Biotechnol. 38 (8) (2020) 947–953.
[11] L. Cassetta, J.W. Pollard, Targeting macrophages: therapeutic approaches in [33] M. Mukhopadhyay, Macrophages enter CAR immunotherapy, Nat. Methods 17 (6)
cancer, Nat. Rev. Drug Discov. 17 (12) (2018) 887–904. (2020) 561, 561-561.
[12] E. Dolgin, Cancer-eating immune cells kitted out with CARs, Nat. Biotechnol. 38 [34] T.L. Roth, C. Puig-Saus, R. Yu, E. Shifrut, J. Carnevale, P.J. Li, J. Hiatt, J. Saco,
(5) (2020) 509–511. P. Krystofinski, H. Li, V. Tobin, D.N. Nguyen, M.R. Lee, A.L. Putnam, A.L. Ferris, J.
[13] J.L. Guerriero, Macrophages: the road less traveled, changing anticancer therapy, W. Chen, J.N. Schickel, L. Pellerin, D. Carmody, G. Alkorta-Aranburu, D. Del
Trends Mol. Med. 24 (5) (2018) 472–489. Gaudio, H. Matsumoto, M. Morell, Y. Mao, M. Cho, R.M. Quadros, C.
[14] A. Viola, F. Munari, R. Sanchez-Rodriguez, T. Scolaro, A. Castegna, The metabolic B. Gurumurthy, B. Smith, M. Haugwitz, S.H. Hughes, J.S. Weissman, K. Schumann,
signature of macrophage responses, Front Immunol. 10 (2019) 1462. J.H. Esensten, A.P. May, A. Ashworth, G.M. Kupfer, S.A.W. Greeley, R. Bacchetta,
[15] P.J. Murray, Macrophage polarization, Annu Rev. Physiol. 79 (2017) 541–566. E. Meffre, M.G. Roncarolo, N. Romberg, K.C. Herold, A. Ribas, M.D. Leonetti,
[16] G.L. Wolf, Macrophages hold the key to cancer’s inner sanctum, Med. Hypotheses A. Marson, Reprogramming human T cell function and specificity with non-viral
122 (2019) 111–114. genome targeting, Nature 559 (7714) (2018) 405–409.
[17] I. Rhee, Diverse macrophages polarization in tumor microenvironment, Arch. [35] D.H. Yoon, M.J. Osborn, J. Tolar, C.J. Kim, Incorporation of immune checkpoint
Pharm. Res. 39 (11) (2016) 1588–1596. blockade into chimeric antigen receptor T cells (CAR-Ts): combination or built-in
[18] B. Ruffell, N.I. Affara, L.M. Coussens, Differential macrophage programming in the CAR-T, Int. J. Mol. Sci. 19 (2) (2018).
tumor microenvironment, Trends Immunol. 33 (3) (2012) 119–126. [36] R.T. Netea-Maier, J.W.A. Smit, M.G. Netea, Metabolic changes in tumor cells and
[19] A. Sica, A. Mantovani, Macrophage plasticity and polarization: in vivo veritas, tumor-associated macrophages: a mutual relationship, Cancer Lett. 413 (2018)
J. Clin. Invest 122 (3) (2012) 787–795. 102–109.
[20] S.K. Biswas, A. Mantovani, Macrophage plasticity and interaction with lymphocyte [37] J.N. Brudno, J.N. Kochenderfer, Recent advances in CAR T-cell toxicity:
subsets: cancer as a paradigm, Nat. Immunol. 11 (10) (2010) 889–896. mechanisms, manifestations and management, Blood Rev. 34 (2019) 45–55.
[21] A. Shapouri-Moghaddam, S. Mohammadian, H. Vazini, M. Taghadosi, S. [38] M. Norelli, B. Camisa, G. Barbiera, L. Falcone, A. Purevdorj, M. Genua, F. Sanvito,
A. Esmaeili, F. Mardani, B. Seifi, A. Mohammadi, J.T. Afshari, A. Sahebkar, M. Ponzoni, C. Doglioni, P. Cristofori, C. Traversari, C. Bordignon, F. Ciceri,
Macrophage plasticity, polarization, and function in health and disease, J. Cell R. Ostuni, C. Bonini, M. Casucci, A. Bondanza, Monocyte-derived IL-1 and IL-6 are
Physiol. 233 (9) (2018) 6425–6440. differentially required for cytokine-release syndrome and neurotoxicity due to CAR
[22] C. Lin, J. Zhang, Chimeric antigen receptor engineered innate immune cells in T cells, Nat. Med. 24 (6) (2018) 739–748.
cancer immunotherapy, Sci. China Life Sci. 62 (5) (2019) 633–639. [39] J. Kim, J.-S. Bae, Tumor-associated macrophages and neutrophils in tumor
[23] C. Alvey, D.E. Discher, Engineering macrophages to eat cancer: from “marker of microenvironment, Mediat. Inflamm. 2016 (2016) 1–11.
self” CD47 and phagocytosis to differentiation, J. Leukoc. Biol. 102 (1) (2017) [40] D. van der Heide, R. Weiskirchen, R. Bansal, Therapeutic targeting of hepatic
31–40. macrophages for the treatment of liver diseases, Front. Immunol. 10 (2019) 2852.
[24] X. Li, R. Liu, X. Su, Y. Pan, X. Han, C. Shao, Y. Shi, Harnessing tumor-associated [41] S.M. Toor, V. Sasidharan Nair, J. Decock, E. Elkord, Immune checkpoints in the
macrophages as aids for cancer immunotherapy, Mol. Cancer 18 (1) (2019) 177. tumor microenvironment, Semin. Cancer Biol. 65 (2020) 1–12.
[25] H. Yang, R. Shao, H. Huang, X. Wang, Z. Rong, Y. Lin, Engineering macrophages to [42] G.V. Kochneva, G.F. Sivolobova, A.V. Tkacheva, A.A. Gorchakov, S.V. Kulemzin,
phagocytose cancer cells by blocking the CD47/SIRPa axis, Cancer Med. 8 (9) Combination of oncolytic virotherapy and CAR T/NK cell therapy for the treatment
(2019) 4245–4253. of cancer, Mol. Biol. 54 (1) (2020) 3–16.
[26] Z. Huang, Z. Zhang, Y. Jiang, D. Zhang, J. Chen, L. Dong, J. Zhang, Targeted [43] C. Zhang, P. Oberoi, S. Oelsner, A. Waldmann, A. Lindner, T. Tonn, W.S. Wels,
delivery of oligonucleotides into tumor-associated macrophages for cancer Chimeric antigen receptor-engineered NK-92 Cells: an off-the-shelf cellular
immunotherapy, J. Control Release 158 (2) (2012) 286–292. therapeutic for targeted elimination of cancer cells and induction of protective
[27] M.T. Villanueva, Macrophages get a CAR, Nat. Rev. Drug Discov. 19 (5) (2020) antitumor immunity, Front. Immunol. 8 (2017) 533.
308, 308-308. [44] Y. Komohara, Y. Fujiwara, K. Ohnishi, M. Takeya, Tumor-associated macrophages:
[28] M.A. Morrissey, A.P. Williamson, A.M. Steinbach, E.W. Roberts, N. Kern, M. potential therapeutic targets for anti-cancer therapy, Adv. Drug Deliv. Rev. 99 (Pt
B. Headley, R.D. Vale, Chimeric antigen receptors that trigger phagocytosis, Elife 7 B) (2016) 180–185.
(2018). [45] T. Giavridis, S.J.C. van der Stegen, J. Eyquem, M. Hamieh, A. Piersigilli,
[29] L. Zhang, L. Tian, X. Dai, H. Yu, J. Wang, A. Lei, M. Zhu, J. Xu, W. Zhao, Y. Zhu, M. Sadelain, CAR T cell-induced cytokine release syndrome is mediated by
Z. Sun, H. Zhang, Y. Hu, Y. Wang, Y. Xu, G.M. Church, H. Huang, Q. Weng, macrophages and abated by IL-1 blockade, Nat. Med 24 (6) (2018) 731–738.
J. Zhang, Pluripotent stem cell-derived CAR-macrophage cells with antigen- [46] D. Mevorach, Apoptotic cells reprogram resident macrophages to support chimeric
dependent anti-cancer cell functions, J. Hematol. Oncol. 13 (1) (2020) 153. antigen receptor (CAR) T cell therapy against peritoneal solid tumor, Biol. Blood
[30] L. Cassetta, T. Kitamura, Macrophage targeting: opening new possibilities for Marrow Transplant. 26 (3) (2020) S314.
cancer immunotherapy, Immunology 155 (3) (2018) 285–293.

You might also like