Advancing Immunotherapy Beyond CAR T-cells: Exploring the Potential of CAR-Macrophages in Solid Tumors
https://www.the-scientist.com/car-macrophages-tackle-challenges-in-solid-cancer-treatment-68604

Advancing Immunotherapy Beyond CAR T-cells: Exploring the Potential of CAR-Macrophages in Solid Tumors

Cancer, with its increasing incidence and mortality rates, remains one of the most significant health challenges globally. Despite advancements in conventional therapies such as surgery, radiation, and chemotherapy, many patients with metastatic or recurrent disease still face poor outcomes. In recent years, the field of cancer treatment has witnessed a paradigm shift with the emergence of targeted therapies and immunotherapies. These advancements, rooted in our expanding knowledge of cancer biology and immunology, have paved the way for precision medicine—a more tailored and less toxic approach to cancer management.

Among the various immunotherapeutic strategies, CAR-T cell therapy has garnered considerable attention for its remarkable success in hematological malignancies. By genetically modifying T cells to express chimeric antigen receptors (CARs) capable of recognizing tumor-specific antigens, CAR-T cells have demonstrated unprecedented efficacy in diseases like acute lymphoblastic leukemia (ALL) and certain types of lymphomas. However, the translation of CAR-T therapy to solid tumors has been met with challenges, including poor tumor infiltration and an immunosuppressive microenvironment.

In response to these challenges, researchers have turned their attention to alternative immune effector cells, such as macrophages, for CAR-based therapy. Macrophages, with their inherent ability to phagocytose pathogens and debris, as well as their presence within the tumor microenvironment, offer a promising platform for solid tumor immunotherapy. In this article, we explore the current status, challenges, recent advances, and potential applications of CAR-M cell therapy in the management of solid tumors.

Advantages of CAR-M Cells

CAR-M cells, derived from macrophages, possess several distinct advantages over CAR-T cells in the context of solid tumor therapy. Unlike T cells, macrophages naturally infiltrate tumors and constitute a significant portion of the tumor-infiltrating immune cells. This inherent ability to migrate to the tumor site addresses one of the major limitations of CAR-T therapy. Additionally, macrophages exhibit robust phagocytic activity, allowing them to engulf and destroy tumor cells directly. Furthermore, macrophages play a crucial role in antigen presentation, thereby facilitating the initiation of adaptive immune responses against cancer cells.

Another significant advantage of CAR-M cells lies in their versatility in terms of cell source. While CAR-T cells are primarily derived from patients' own T cells (autologous therapy), CAR-M cells can be generated from various sources, including peripheral blood mononuclear cells (PBMCs), induced pluripotent stem cells (iPSCs), and cell lines such as THP-1. This flexibility not only simplifies the manufacturing process but also enables the development of allogeneic CAR-M cell therapies, potentially broadening their applicability and accessibility.

Furthermore, CAR-M cells offer a reduced risk of graft-versus-host disease (GvHD) compared to CAR-T cells. Since macrophages are tissue-resident cells with limited systemic circulation, the likelihood of off-target effects and immune-mediated complications is diminished, making CAR-M therapy potentially safer for patients.

Clinical Applications and Trials

The clinical translation of CAR-M therapy is still in its early stages, with several ongoing trials aimed at evaluating its safety and efficacy in patients with solid tumors. These trials utilize various CAR-M constructs targeting different tumor-associated antigens, including HER2, Glypican 3 (GPC3), and mesothelin (MSLN), among others.

One notable example is the Phase I clinical trial (NCT04660929) conducted by CARISMA Therapeutics, which investigated the use of HER2-targeted CAR-M cells in solid tumors. In this study, CAR-M cells were engineered using a chimeric adenoviral vector to target HER2-expressing tumor cells. Preliminary results from this trial demonstrated promising anti-tumor activity and a favorable safety profile, paving the way for further exploration of CAR-M therapy in solid tumors.

Additionally, ongoing preclinical studies are exploring novel CAR-M constructs and combination therapies to enhance their efficacy and overcome potential limitations. These include strategies to improve CAR-M trafficking and persistence within the tumor microenvironment, mitigate off-target toxicity, and optimize CAR-M bioengineering and manufacturing processes.

Biological Properties of Macrophages

To understand the therapeutic potential of CAR-M cells, it is essential to consider the biological properties of macrophages, particularly their role within the tumor microenvironment. Macrophages exhibit remarkable phenotypic plasticity, with distinct polarization states ranging from the pro-inflammatory M1 phenotype to the immunosuppressive M2 phenotype.

In the context of solid tumor therapy, M1 macrophages play a crucial role in anti-tumor immunity. These classically activated macrophages are characterized by their ability to produce pro-inflammatory cytokines, such as interleukin-12 (IL-12), and mediate tumor cell killing through phagocytosis and the release of reactive oxygen and nitrogen species. Furthermore, M1 macrophages can act as antigen-presenting cells, thereby initiating adaptive immune responses against cancer cells.

On the other hand, tumor-associated macrophages (TAMs) often exhibit an M2-like phenotype, which is associated with tumor-promoting activities such as immunosuppression, angiogenesis, and tissue remodeling. Targeting these immunosuppressive TAMs and reprogramming them into an anti-tumor M1 phenotype represents a promising therapeutic strategy for solid tumors.

Limitations and Optimization Strategies

Despite the promise of CAR-M therapy, several challenges must be addressed to maximize its clinical utility. These include limitations related to CAR-M bioengineering, storage, expansion, persistence within the tumor microenvironment, and potential toxicity risks.

Strategies to Overcome Limitations in CAR-M Bioengineering

To enhance CAR-M cell production and transduction efficiency, researchers have explored various viral and non-viral gene delivery methods. Modified lentiviral vectors containing accessory proteins such as Vpx have shown promising results in delivering transgenes to myeloid cells, overcoming barriers such as SAMHD1-mediated restriction. Additionally, chimeric adenovirus vectors have been utilized to efficiently transduce human macrophages and maintain their pro-inflammatory M1 phenotype.

Non-viral methods, including mRNA transfection and bacterial plasmid DNA, offer alternative approaches for CAR-M cell engineering. Moreover, the use of polymer nanocarriers has been investigated to enhance gene delivery and expression in macrophages, further optimizing their anti-tumor potential.

Strategies to Enhance Antitumor Activity of CAR-M

Several strategies have been proposed to enhance the antitumor activity of CAR-M cells, including promoting their polarization toward the pro-inflammatory M1 phenotype. Studies have demonstrated that CAR-M cells targeting HER2 or mesothelin can induce M1 polarization and cytokine secretion upon interaction with tumor cells, leading to enhanced tumor cell killing and immune activation.

Strategies to Enhance Trafficking and Persistence of CAR-M within the TME

?Improving CAR-M trafficking and persistence within the immunosuppressive tumor microenvironment (TME) is critical for maximizing their therapeutic efficacy. One approach involves engineering CAR-M cells to express chemokine receptors or ligands that facilitate their migration toward tumor sites. For example, CAR-M cells expressing CCR7 ligands were shown to promote T cell infiltration into tumors and suppress tumor growth.

Another strategy focuses on modulating the expression of matrix metalloproteinases (MMPs) by CAR-M cells to facilitate tumor penetration and remodeling of the extracellular matrix (ECM). By enhancing MMP expression, CAR-M cells can overcome physical barriers within the TME and improve their ability to eradicate tumor cells.

Overcoming CAR-M Toxicity

The potential for cytokine release syndrome (CRS) and off-target toxicity represents significant hurdles in CAR-M therapy. Strategies to mitigate these risks include optimizing CAR-M cell dosing and administration regimens, as well as implementing strategies to enhance their specificity and selectivity toward tumor cells.

Additionally, ongoing research is focused on developing strategies to minimize the systemic distribution of CAR-M cells and enhance their tumor-targeting capabilities. For example, the use of tumor-targeting peptides or antibodies conjugated to CAR-M cells could potentially improve their tumor specificity while reducing off-target effects.

Conclusion

CAR-M cell therapy represents a promising frontier in cancer immunotherapy, leveraging the unique properties of macrophages to overcome key challenges associated with solid tumor treatment. While still in the early stages of development, CAR-M therapy holds immense potential for revolutionizing cancer treatment and improving patient outcomes. Continued research and clinical trials are essential for further elucidating the therapeutic potential of CAR-M cells and optimizing their clinical application in the management of solid tumors.

?

Alex Jones (PhD/CEO)

???????? ?????????????? for Cell Therapy & Research

7 个月

You may be interested in a cytometry and cell sorting for Cell and Gene Therapy event in May, if so take a look (https://www.dhirubhai.net/events/cytometryandsortingincelltherap7186409732901863426/ )

回复

要查看或添加评论,请登录

社区洞察

其他会员也浏览了