CAR-macrophages: tailoring cancer immunotherapy DOI Creative Commons

Dewan Chettri,

Bibhu Prasad Satapathy,

Rohit Yadav

et al.

Frontiers in Immunology, Journal Year: 2025, Volume and Issue: 15

Published: Jan. 14, 2025

The ability of effector immune cells to target and eliminate tumor by focusing on tumor-associated antigens is crucial for the success immunotherapy. Chimeric Antigen Receptor (CAR)-modified have revolutionized cancer immunotherapy, primarily with CAR-T showing remarkable in hematological cancers. Numerous cell-based therapies, such as cell, TIL, CAR-NK T-cell receptor (TCR)-based are currently undergoing clinical pre-clinical evaluation across various types. However, these therapies possess limitations, including their inability penetrate stoma, change TME, exaggerated inflammatory responses. To overcome this, developing advanced flexible strategies precisely while preserving homeostasis patients imperative. One promising approach involves using engineered macrophages (TAMs), which plastic nature constitute approximately 50% microenvironment (TME) indispensable both progression regression. CAR-macrophages aim reprogram toward M1 TAM phenotype enable them immune-suppressive TME facilitate immune-mediated destruction tumors. Body textThe typically comprises cells, more than total cell mass play a role or Tumor-associated extremely can adapt function response environmental stimuli. TAMs within exist two distinct phenotypes: (regulatory) M2 (trophic). produces proinflammatory cytokines like IL-12 TNF-α, activates T attack demonstrate antitumor activity. presence tumors often linked better outcomes due regulate growth. advances, intrinsic extrinsic factors drive polarization tumor-infiltrating into [1], supports development. These M2-polarized secrete pro-tumoral angiogenic factors, IL-10, TGF-β, VEGF, contributing an immunosuppressive environment that favors growth [2]. Various efforts under progress engineering express receptors recognize vide range (TAA) pipeline, allowing selectively destroy cells. macrophages, known Macrophages (CAR-Macrophages) capable enough priming reactive T-cells keeping natural tumor-targeting [3]. Thus, it clear CAR-Macrophage (CAR-M) therapy would be potential asset immunotherapy many solid Underlying MechanismCAR-Macrophage recognizes specific through its chimeric antigen (CAR), leading enhanced phagocytosis concomitant presentation subsequent activation. CAR-Ms modulate secreting cytokines, metalloproteinases (MMPs), Reactive oxygen species (ROS) serine proteases altogether infiltration anti-tumor responses Each mechanisms (Figure 1) contributes overall effectiveness CAR-Macrophages targeting eliminating making therapeutic CAR-engineered were shown actively migrate sites, could locally deliver and/or cytotoxic substances antigen-specific environments when used drug delivery system, significantly alter [4]. leverage tumor-homing tendencies cargo induce activity niche. With help regenerative technologies, induced pluripotent stem (iPSCs) make CD19 / mesothelin+ M1programmed bear strong anti-tumoral [5]. Engineered derived from diverse sources, primary human monocytes/macrophages, (iPSCs), hematopoietic progenitor (HSPCs). Abdin et al. (2023) iPSCs, HSPCs generate anti-CD19 CAR-Ms. They generated αCD19 CAR constructs lentiviral vectors verified via sequencing. CD34+ isolated cord blood, transduced, differentiated macrophages. iPSCs cultured, mesoderm-primed, defined cytokines. Cancer lines patient-derived samples transduced co-cultured assess phagocytosis. Then they flow cytometry, confocal microscopy, western blotting verify action displayed CD19+ pro-inflammatory responses, adaptive recruitment, scRNA sequencing revealed activation pathways cytokine upregulation [6]. In another study Zhang (2023), utilized CRISPR-Cas9 gene editing integrate anti-GD2 AAVS1 locus (hPSCs). developed serum- feeder-free differentiation protocol (CAR-Ms) arterial endothelial-to-hematopoietic transition (EHT). this way demonstrated potent against GD2-expressing neuroblastoma melanoma vitro vivo. It useful generating off-the-shelf CAR-Ms, advancing applications [7]. improve efficiency functionality hPSC shen (2024) optimized monolayer-based system achieving stable expression tumoricidal vitro. address diminished vivo activity, employed interferon-γ monophosphoryl lipid-A innate activation, repolarizing hPSC-CAR-Ms Additionally, activating collaborative innate-adaptive amplifying effects [8]. Using high-throughput screening, Mukalel identified oxidized lipid nanoparticles (oLNPs) monocyte tropism effective mRNA delivery. C14-O2 oLNP successfully CD19-CAR monocytes vivo, significant B depletion, highlighting [9]. Similarly, numerous novel approaches being developed, efficacy existing methods continues undergo constant refinement enhancement. ongoing advancements precision, scalability, current ensuring meet evolving needs applications, particularly fields cellular. Reprogramming macrophage-directed strategy delivering [10] innovative improving rejection [11]. past, we amply returned/conditioned syngeneic donor mice dictated promoted high-grade highly invasive neuroendocrine pancreas. Most intriguingly, adoptive transfer reprogrammed (which regarded surrogate CAR-M) potentially normalized vasculature aided PanNETs [12-14]. On basis believe HER2 CD47-specific M, sensing clearing dead containing neighboring (immune) also skewed Th1 lymphocytes (CD8+ cells), secretion major effectors viz IFN-γ & IL-2, neutralizing exhaustion markers PD-1, TIGIT, LAG-3 [15]. This explained entitle niche [16]. mechanism might responsible HER2-directed CAR-macrophage (CT-0508) so far phase-1 trial (NCT04660929). interestingly was safe tolerable variety indicated CAR-M based raised hope large future. Challenges ProspectiveAlthough CAR-M-based several advantages however few bottlenecks still associated them. issues do not proliferate post-administration, amount tolerate limited, reduce interventions. Moreover, exogenous tend accumulate liver after passing lungs, neutralize potentials. complexity over murine model factor possesses challenge effective. Such heterogeneous restrict wide antigens, impact recognition/binding TSA/TAA –M TME. whether support local tumor-supportive should carefully addressed, especially at research stage. problem has been observed likely pose obstacle general. Both stability durability genetic modifications hurdle off-target stabilized CRISPR-based approach. prevent excessive inflammation, paramount maximizing therapies. achieved floxing iNOS Arginase CD47 promoter affording palliative iNOsFloxed/ArginaseFloxedCD47+CAR-M adverse impact. would, only enhance but ensure successful trials employ CAR-M. fidelity CAR–M program pave new patients.

Language: Английский

Cancer immunotherapies: advances and bottlenecks DOI Creative Commons
Rui Rui, Liqun Zhou, Shiming He

et al.

Frontiers in Immunology, Journal Year: 2023, Volume and Issue: 14

Published: Aug. 24, 2023

Immunotherapy has ushered in a new era cancer treatment, and immunotherapy continues to be rejuvenated. The clinical goal of is prime host immune system provide passive or active immunity against malignant tumors. Tumor infiltrating leukocytes (TILs) play an immunomodulatory role tumor microenvironment (TME) which closely related escape cells, thus influence progress. Several immunotherapies, include checkpoint inhibitors (ICIs), vaccine, adoptive cell transfer (ACT), have shown great efficacy promise. In this review, we will summarize the recent research advances immunotherapy, including molecular mechanisms effects as well limitations immunotherapy.

Language: Английский

Citations

176

Challenges and new technologies in adoptive cell therapy DOI Creative Commons
Pengchao Zhang, Guizhong Zhang, Xiaochun Wan

et al.

Journal of Hematology & Oncology, Journal Year: 2023, Volume and Issue: 16(1)

Published: Aug. 18, 2023

Abstract Adoptive cell therapies (ACTs) have existed for decades. From the initial infusion of tumor-infiltrating lymphocytes to subsequent specific enhanced T receptor (TCR)-T and chimeric antigen (CAR)-T therapies, many novel strategies cancer treatment been developed. Owing its promising outcomes, CAR-T therapy has revolutionized field ACTs, particularly hematologic malignancies. Despite these advances, still limitations in both autologous allogeneic settings, including practicality toxicity issues. To overcome challenges, researchers focused on application CAR engineering technology other types immune engineering. Consequently, several new based developed, CAR-NK, CAR-macrophage, CAR-γδT, CAR-NKT. In this review, we describe development, advantages, possible challenges aforementioned ACTs discuss current aimed at maximizing therapeutic potential ACTs. We also provide an overview various gene transduction employed immunotherapy given their importance Furthermore, possibility that capable creating a positive feedback circuit, as healthy systems do, could address flaw single type ACT, thus serve key players future immunotherapy.

Language: Английский

Citations

82

The CAR macrophage cells, a novel generation of chimeric antigen-based approach against solid tumors DOI Creative Commons
Kaveh Hadiloo,

Siavash Taremi,

Mahmood Heidari

et al.

Biomarker Research, Journal Year: 2023, Volume and Issue: 11(1)

Published: Nov. 28, 2023

Abstract Today, adoptive cell therapy has many successes in cancer therapy, and this subject is brilliant using chimeric antigen receptor T cells. The CAR with its FDA-approved drugs, could treat several types of hematological malignancies thus be very attractive for treating solid cancer. Unfortunately, the cannot functional cancers due to unique features. This treatment method harmful adverse effects that limit their applications, so novel treatments must use new cells like NK cells, NKT macrophage Among these innate features, are more tumor seem a better candidate prior methods. have vital roles microenvironment and, direct effect, can eliminate efficiently. In addition, being part immune system, attended sites. With high infiltration, modulations effective. review investigates last achievements CAR-macrophage future immunotherapy method.

Language: Английский

Citations

45

Human anti-PSCA CAR macrophages possess potent antitumor activity against pancreatic cancer DOI
Zahir Shah, Lei Tian, Zhixin Li

et al.

Cell stem cell, Journal Year: 2024, Volume and Issue: 31(6), P. 803 - 817.e6

Published: April 24, 2024

Language: Английский

Citations

28

Advances in Engineered Macrophages: A New Frontier in Cancer Immunotherapy DOI Creative Commons
Shuaixi Yang, Yuhang Wang,

Jiachi Jia

et al.

Cell Death and Disease, Journal Year: 2024, Volume and Issue: 15(4)

Published: April 1, 2024

Abstract Macrophages, as pivotal cells within the tumour microenvironment, significantly influence impact of and reactions to treatments for solid tumours. The rapid evolution bioengineering technology has revealed vast potential engineered macrophages in immunotherapy, disease diagnosis, tissue engineering. Given this landscape, goal harnessing innovating a novel strategy immunotherapy cannot be overstated. diverse strategies realm cancer encompassing macrophage drug delivery systems, chimeric antigen receptor therapy, synergistic treatment approaches involving bacterial outer membrane vesicles macrophages, are meticulously examined review. These methodologies designed enhance therapeutic efficacy against tumours, particularly those that drug-resistant metastatic. Collectively, these immunotherapies poised supplement refine current paradigms, thus heralding new frontier fight malignant

Language: Английский

Citations

22

A new era of cancer immunotherapy: combining revolutionary technologies for enhanced CAR-M therapy DOI Creative Commons
Na Li,

Shinan Geng,

Zhenzhen Dong

et al.

Molecular Cancer, Journal Year: 2024, Volume and Issue: 23(1)

Published: June 1, 2024

Abstract Significant advancements have been made in the application of chimeric antigen receptor (CAR)-T treatment for blood cancers during previous ten years. However, its effectiveness treating solid tumors is still lacking, necessitating exploration alternative immunotherapies that can overcome significant challenges faced by current CAR-T cells. CAR-based immunotherapy against shows promise with emergence macrophages, which possess robust phagocytic abilities, antigen-presenting functions, and ability to modify tumor microenvironment stimulate adaptive responses. This paper presents a thorough examination latest progress CAR-M therapy, covering both basic scientific studies clinical trials. study examines primary obstacles hindering realization complete potential as well strategies be employed these hurdles. With revolutionary technologies like situ genetic modification, synthetic biology techniques, biomaterial-supported gene transfer, provide wider array resources manipulating tumor-associated we suggest combining advanced methods will result creation new era therapy demonstrates improved efficacy, safety, availability. Graphical

Language: Английский

Citations

20

M1 polarization enhances the antitumor activity of chimeric antigen receptor macrophages in solid tumors DOI Creative Commons
Yi Huo, Han Zhang,

Longqi Sa

et al.

Journal of Translational Medicine, Journal Year: 2023, Volume and Issue: 21(1)

Published: March 28, 2023

Abstract Background Chimeric antigen receptor macrophage (CAR-M) therapy is a novel cancer immunotherapy approach that integrates CAR structure and functions. CAR-M has shown unique impressive antitumor effects in for solid tumors. However, the polarization state of macrophages can affect effect CAR-M. We hypothesized activity CAR-Ms may be further improved after inducing M1-type polarization. Methods In this report, we constructed HER2-targeting CAR-M, which was composed humanized anti-HER2 scFv, CD28 hinge region FcγRI transmembrane domain intracellular domain. Phagocytosis, tumor-killing capacities, cytokine release were detected with or without M1-polarization pretreatment. Several syngeneic tumor models used to monitor vivo M1-polarized CAR-Ms. Results After LPS combined interferon-γ vitro, found phagocytic capacities against target cells significantly enhanced. The expression costimulatory molecules proinflammatory cytokines also increased By establishing several vivo, demonstrated infusing polarized could effectively suppress progression prolong survival tumor-bearing mice enhanced cytotoxicity. Conclusions our eliminate HER2-positive both vitro M1 ability resulting stronger therapeutic immunotherapy.

Language: Английский

Citations

39

New cell sources for CAR-based immunotherapy DOI Creative Commons
Marzieh Mazinani, Fatemeh Rahbarizadeh

Biomarker Research, Journal Year: 2023, Volume and Issue: 11(1)

Published: May 6, 2023

Chimeric antigen receptor (CAR) T cell therapy, in which a patient's own lymphocytes are engineered to recognize and kill cancer cells, has achieved striking success some hematological malignancies preclinical clinical trials, resulting six FDA-approved CAR-T products currently available the market. Despite impressive outcomes, concerns about treatment failure associated with low efficacy or high cytotoxicity of cells remain. While main focus been on improving exploring alternative cellular sources for CAR generation garnered growing interest. In current review, we comprehensively evaluated other rather than conventional generation.

Language: Английский

Citations

25

CAR Macrophages: a promising novel immunotherapy for solid tumors and beyond DOI Creative Commons
Jialin Lu,

Yuqing Ma,

Q. Quentin Li

et al.

Biomarker Research, Journal Year: 2024, Volume and Issue: 12(1)

Published: Aug. 23, 2024

Abstract With the advent of adoptive cellular therapy, chimeric antigen receptor (CAR)-T cell therapy has gained widespread application in cancer treatment and demonstrated significant efficacy against certain hematologic malignancies. However, due to limitations CAR-T treating solid tumors, other immune cells are being modified with CAR address this issue. Macrophages have emerged as a promising option, owing their extensive functions, which include presentation, powerful tumor phagocytosis, particularly active trafficking microenvironment. Leveraging unique advantages, CAR-macrophages (CAR-M) expected enhance effectiveness treatments novel form immunotherapy, potentially overcoming major challenges associated CAR-T/NK therapy. This review outlines primary mechanism underlying CAR-M recent progressions while also discussing further applications.

Language: Английский

Citations

14

The next frontier in immunotherapy: potential and challenges of CAR-macrophages DOI Creative Commons
Jing Li,

Ping Chen,

Wenxue Ma

et al.

Experimental Hematology and Oncology, Journal Year: 2024, Volume and Issue: 13(1)

Published: Aug. 5, 2024

Abstract Chimeric antigen receptor macrophage (CAR-MΦ) represents a significant advancement in immunotherapy, especially for treating solid tumors where traditional CAR-T therapies face limitations. CAR-MΦ offers promising approach to target and eradicate tumor cells by utilizing macrophages’ phagocytic antigen-presenting abilities. However, challenges such as the complex microenvironment (TME), variability expression, immune suppression limit their efficacy. This review addresses these issues, exploring mechanisms of action, optimal construct designs, interactions within TME. It also delves into ex vivo manufacturing CAR-MΦ, discussing autologous allogeneic sources importance stringent quality control. The potential synergies integrating with existing cancer like checkpoint inhibitors conventional chemotherapeutics are examined highlight possible enhanced treatment outcomes. Furthermore, regulatory pathways scrutinized alongside established protocols cells, identifying unique considerations essential clinical trials market approval. Proposed safety monitoring frameworks aim manage adverse events, cytokine release syndrome, crucial patient safety. Consolidating current research insights, this seeks refine therapeutic applications, overcome barriers, suggest future directions transition from experimental platforms standard care options.

Language: Английский

Citations

12