Introduction to CAR-M
CAR-macrophages (CAR-Ms) represent an innovative cell-based immunotherapy modality in which autologous or allogeneic macrophages are genetically engineered to express a chimeric antigen receptor. These receptors are designed to target specific antigens expressed on
tumor cells or other diseased tissues. The overarching objective of CAR-M therapy is to harness the inherent phagocytic and antigen-presenting functions of macrophages to boost anticancer immunity and simultaneously modify the tumor microenvironment (TME) in favor of immune activation. This technology is emerging as a complement—and in some cases an alternative—to established CAR-T cell therapies, particularly in the treatment of
solid tumors, where conventional T cells often fail to infiltrate and persist efficiently.
Definition and Mechanism
At its most basic level, CAR-Ms are defined as macrophages that have been genetically reprogrammed to express a chimeric antigen receptor. The CAR construct usually consists of an extracellular single-chain variable fragment (scFv) specific to a
tumor-associated antigen (TAA), a hinge region, a transmembrane domain, and one or more intracellular signaling domains. When the engineered macrophage engages with its target antigen through the scFv, intracellular signaling cascades are triggered that enhance the phagocytic ability of the macrophage, promote cytokine and chemokine release, and can even lead to repolarization of tumor-associated macrophages (TAMs) from an immunosuppressive (M2-like) state to a pro-inflammatory (M1-like) phenotype. This dual function of direct tumor cell clearance through phagocytosis and modulation of the TME distinguishes CAR-Ms from other cell therapies.
Development History
The development of CAR-M technology has evolved over the past decade from early preclinical studies focused on optimizing gene transfer techniques (e.g., adenoviral vectors such as Ad5f35) to generate stably modified macrophages, to more sophisticated strategies that include the use of induced pluripotent stem cells (iPSCs) for manufacturing off-the-shelf CAR-M products. Companies like
Resolution Therapeutics,
CellOrigin Biotechnology,
Evotec SE, and
Oncoinsight are at various stages of development with CAR-M candidates targeting a range of indications. Initially, the research concentrated on proof-of-concept preclinical models in cancer, primarily demonstrating antigen-specific phagocytosis and TME remodeling. The first-in-human clinical trials, such as CT-0508 targeting HER2-overexpressing tumors, have further propelled the field, encouraging ongoing development and more refined molecular designs.
Current Indications for CAR-M
CAR-M therapies are primarily investigated as anticancer treatments. In addition, emerging research also hints at potential non-cancer applications where modulation of the immune environment may be beneficial. The general-specific-general structure of these investigations underscores that while the bulk of the current activities focus on cancer indications, the versatility of macrophages opens avenues for non-oncological inflammatory and possibly regenerative conditions.
Cancer Indications
The overwhelming majority of CAR-M studies have been directed toward various types of cancers, with a particular emphasis on solid tumors. These indications include, but are not limited to:
1. HER2-Overexpressing Tumors:
Several clinical trials have been designed with anti-HER2 CAR-Ms. For instance, the CT-0508 trial in which genetically modified macrophages are administered to patients with HER2-overexpressing tumors is a landmark study. HER2 is a well-known oncogenic driver in breast cancer and other solid tumors, and the targeting of HER2 has shown promise due to its critical role in tumor growth and progression. CAR-Ms directed against HER2 not only phagocytose tumor cells but also help convert the residing M2-like TAMs into pro-inflammatory M1 macrophages, thereby stimulating downstream T-cell responses.
2. Gastrointestinal and Digestive System Disorders:
An interesting aspect of CAR-M development is the investigation of agents like RTX-001 from Resolution Therapeutics Ltd., which is in Phase 1/2 trials and indicated for a subset of digestive system disorders that are oncologic in nature. In these cases, the target antigen may be overexpressed in cancers arising in the digestive tract, thereby exploiting the unique ability of macrophages to penetrate the dense stroma of gastrointestinal tumors.
3. Urogenital and Endocrine-Related Neoplasms:
CAR-M candidates, such as SY-001 developed by CellOrigin Biotechnology, are being evaluated for their efficacy in tumors that span several therapeutic areas, including neoplasms of the urogenital system, as well as endocrine and metabolic diseases. The inclusion of multiple therapeutic areas in the development strategy reflects the versatility of CAR-M constructs whose targets might be shared among cancers in diverse organ systems.
4. Ovarian, Pancreatic, and Other Solid Tumors:
Preclinical evidence has shown that engineered macrophages can infiltrate and remodel the TME not only in breast cancer and HER2-driven cancers but also in other solid tumors such as ovarian and pancreatic cancers. The design of CAR-Ms with the capability to trigger both phagocytosis and T-cell recruitment provides a two-pronged attack against these tumors. These studies are supported by a growing body of literature emphasizing the importance of overcoming the stromal barrier through the use of myeloid cells, which naturally possess migratory capabilities in solid tumors.
5. Hematological Malignancies:
Although CAR-M research has been predominantly concentrated on solid tumors, a few preclinical studies and early discovery projects are exploring their potential in hematologic malignancies. For instance, products such as iPSC-derived CAR-M (from SNC Stem Cell) are at the discovery stage and are being investigated for their role in potentially targeting neoplasms in hematological settings. The advantages of using macrophages—such as their antigen presentation capabilities—may complement the direct cytolytic effects traditionally associated with CAR-T cells in these indications.
6. Glioblastoma and Central Nervous System Tumors:
Certain preclinical studies have explored the use of CAR-Ms in the treatment of glioblastoma multiforme (GBM). Given the fact that macrophages and microglia naturally reside within the central nervous system, engineering these cells to express CARs offers a unique therapeutic approach for intracranial tumors. One study demonstrated that chimeric antigen receptor–engineered microglia/macrophages have the capacity not only to engulf glioma stem cells but also to stimulate T-cell–mediated responses, thereby establishing a robust immune environment within the post-surgical cavity of GBM tumors.
7. Mesothelin-Expressing Tumors:
Mesothelin has emerged as a promising target antigen for several solid tumors, including mesothelioma, ovarian cancer, and pancreatic cancer. Preclinical data have shown that anti-mesothelin CAR-Ms can mediate efficient tumor clearance via antigen-specific phagocytosis and secretion of pro-inflammatory cytokines. The modular design of CAR-Ms allows them to be armed with different signaling domains to optimize their antitumor activity depending on the target, making mesothelin one of the key antigens in various ongoing studies.
Non-Cancer Indications
While cancer remains the primary focus, there is also emerging interest in leveraging the unique properties of CAR-engineered macrophages for non-oncological conditions. Although still in early exploratory stages, these applications capitalize on the inherent ability of macrophages to modulate inflammatory responses and tissue homeostasis.
1. Inflammatory and Autoimmune Diseases:
Macrophages play a central role in both the initiation and resolution of inflammation. Engineered CAR-Ms could theoretically be designed to target specific inflammation-associated antigens or even autoantigens that drive chronic inflammatory and autoimmune diseases. For example, the modulation of cytokine release through CAR signaling may help temper exaggerated inflammatory responses in diseases such as rheumatoid arthritis or inflammatory bowel disease. Research in this area is still preclinical but holds promise as a novel way to temper immune responses without globally suppressing the immune system.
2. Tissue Remodeling and Fibrosis:
Given that macrophages are key regulators of tissue repair and remodeling, there are potential applications for CAR-M therapies in fibrotic conditions. These conditions, which are characterized by excessive extracellular matrix deposition that can lead to organ dysfunction (e.g., pulmonary fibrosis or liver cirrhosis), may benefit from CAR-M cells specifically engineered to degrade fibrotic matrices or to secrete enzymes such as matrix metalloproteinases (MMPs) that can break down scar tissue. Early preclinical strategies are testing the concept of using engineered macrophages as “living converters” of the tissue microenvironment to promote regeneration rather than fibrosis.
3. Organ Transplantation:
In the realm of transplantation, the immunomodulatory potential of macrophages can be harnessed to induce tolerance and reduce the risk of graft rejection. Although most current research in cell therapy for transplantation has focused on CAR-Treg cells, the concept of tailoring CAR-Ms to modulate alloimmune responses is emerging. Such approaches might involve programming macrophages to express inhibitory molecules or secrete cytokines that foster tolerance, thereby protecting transplanted tissues. This area remains highly exploratory and requires further in-depth preclinical studies before clinical translation.
Clinical Trials and Research
The investigation of CAR-M therapies spans both clinical trials and preclinical research settings, which together create a comprehensive development pathway that addresses both the immediate therapeutic potential and the iterative improvement of these technologies.
Ongoing Clinical Trials
Several early-phase clinical trials involving CAR-Ms have already been initiated as the field transitions from preclinical promise to clinical application. A notable example includes the phase I trial evaluating CT-0508, an autologous HER2-targeted CAR-M product developed by Carisma Therapeutics, which represents the first clinical exploration of CAR-M therapy in human subjects. In this trial, patients with HER2-overexpressing solid tumors—who have either exhausted approved HER2-targeted therapies or have shown recurrences—are receiving CAR-M infusions. The trial employs both monotherapy as well as combination sub-studies with checkpoint inhibitors like pembrolizumab, aimed at reinforcing antitumor T-cell responses via TME modulation.
Other trials include candidates from CellOrigin Biotechnology and Evotec SE that target broad neoplasms, including digestive system cancers, urogenital tumors, and other malignancies. Products such as SY-001, iPSC-derived CAR-Ms (e.g., iPS-CAR-Mac01, Mac02, etc.), and SIRPα knockout CAR-Ms are in various phases of development ranging from preclinical to Phase I/II studies. Although the published literature from synapse indicates that many of these studies are preclinical or in the discovery phase, there is a clear clinical trajectory aimed at validating both safety and efficacy in humans.
These clinical trials are pivotal for several reasons. They not only assess the safety profile and potential off-target effects of CAR-M infusions in the complex human physiological setting but also begin to demonstrate whether the unique attributes of macrophages—such as their capacity to penetrate solid tumor stroma and to reprogram the TME—translate into meaningful therapeutic outcomes. The early data signaling feasibility and tolerability are encouraging and have prompted further expansion into additional cancer types and possibly into non-cancer indications in the future.
Preclinical Studies
Preclinical research on CAR-M therapies has been both extensive and multifaceted. Numerous in vitro experiments and animal studies have demonstrated that CAR-Ms can mediate antigen-specific phagocytosis and enhance antitumor immunity by secreting pro-inflammatory cytokines and remodeling the TME.
1. Animal Models:
Studies in murine models have been instrumental in validating the antitumor effects of CAR-Ms. For instance, preclinical evaluations conducted with engineered macrophages targeting HER2 have shown significant tumor load reduction and prolonged survival in xenograft models of ovarian cancer and breast cancer. Additionally, research involving CAR-Ms in glioblastoma models has revealed that these cells are capable of homing into the brain and eradicating tumor cells, thereby delaying tumor recurrence after surgical resection.
2. iPSC-derived CAR-Macrophages:
The advent of iPSC technology has opened new doors for the production of CAR-Ms at scale. Several studies have reported on iPSC-derived CAR-M products (e.g., iPS-CAR-Mac03, Mac01, Mac02, Mac04) that exhibit robust antigen-specific activity and can be standardized for clinical application. These preclinical studies emphasize not only the antitumor efficacy but also the potential for better manufacturing logistics and reduced batch-to-batch variability compared to autologous products.
3. Mechanistic Studies:
Detailed mechanistic investigations have focused on understanding how CAR-Ms signal to induce phagocytosis and cytokine release. These studies have compared various intracellular domains—including those derived from CD3ζ, FcRγ, and other costimulatory molecules—to optimize the antitumor activity of CAR-Ms. One key finding is that certain intracellular domain combinations not only boost phagocytic capacity but also lead to a sustained M1 phenotype even in the immunosuppressive TME, which is critical for efficient tumor clearance.
4. Combination Studies:
Preclinical models have also examined the synergistic potential of CAR-M therapies when combined with other modalities such as checkpoint inhibitors (anti-PD1), conventional chemotherapy, or other cell-based therapies like CAR-T cells. These studies suggest that the immunomodulatory effect of CAR-Ms on the TME can enhance the overall antitumor response, even in models where single-agent therapies have limited efficacy.
Challenges and Future Perspectives
Despite the promising progress to date, there remain significant technical, biological, and clinical challenges that must be overcome before CAR-M therapies can be widely implemented across different indications. Addressing these challenges will not only improve the efficacy and safety profiles of CAR-Ms but also expand the range of indications—for cancer and potentially non-cancer conditions—that can be treated with this innovative approach.
Technical and Biological Challenges
1. Gene Transfer Efficiency and Stability:
One of the critical challenges in manufacturing CAR-Ms is achieving efficient and stable gene transfer into macrophages. Unlike T cells, macrophages possess robust innate immune mechanisms that can limit the uptake and stable expression of viral vectors. Although adenoviral vectors (e.g., Ad5f35) have been successfully employed to generate CAR-Ms, the risk of insertional mutagenesis and phenotypic alteration remains an important technical barrier. Continued advances in vector design—including nonviral gene delivery systems and CRISPR/Cas9-based approaches—are being explored to mitigate these issues.
2. Maintenance of the M1 Phenotype:
The tumor microenvironment is notorious for its immunosuppressive milieu, which can push macrophages toward an M2 phenotype. For CAR-Ms to retain their effective antitumor functions, it is essential to maintain a durable M1 pro-inflammatory phenotype even in the presence of immunosuppressive signals. Preclinical studies have shown that specific viral vectors and intracellular signaling domain combinations help sustain the M1 state, but further refinements are needed to ensure long-term efficacy in clinical settings.
3. Trafficking, Infiltration, and Persistence:
Effective antitumor activity requires engineered macrophages to not only home to tumor sites but also to infiltrate and persist within the TME. While macrophages are inherently adept at trafficking into tumors, heterogeneity in tumors and physical barriers such as dense extracellular matrices may still limit their accumulation and sustained activity. Strategies to enhance chemokine receptor expression or modulate adhesion molecules are actively being researched as potential solutions.
4. Off-Target Effects and Safety Concerns:
As with any targeted therapy, specificity for tumor-associated antigens is paramount. There is potential for “on-target, off-tumor” toxicity if the antigen targeted by the CAR is also expressed on healthy tissues. Preclinical studies are focusing on identifying antigens that are highly expressed on tumor cells while sparing normal tissues, but this remains an ongoing challenge. Early clinical trials, such as those using HER2-targeted CAR-Ms, are carefully monitoring cytokine release syndrome and other adverse events to better understand the safety profile.
5. Manufacturing and Scalability:
The production of autologous CAR-Ms is inherently challenging due to the limited number of macrophages present in peripheral blood and the lack of in vitro expansion methods compared to T cells. iPSC-derived CAR-Ms represent one promising avenue to overcome these limitations, offering the potential for off-the-shelf products that can be manufactured at scale. However, standardizing these processes and ensuring safety and efficacy remains an active area of research.
Future Research Directions
1. Optimization of CAR Constructs:
Future research will continue to focus on optimizing the CAR design for macrophages. This includes evaluating various intracellular signaling domains and costimulatory modules that can enhance not only phagocytic activity but also cytokine secretion and antigen presentation. Iterative modifications in the CAR structure—guided by detailed mechanistic and preclinical studies—are expected to yield constructs that are more potent and have improved persistence within the TME.
2. Combination Therapies and Multipronged Approaches:
The integration of CAR-M therapies with other immunotherapeutic strategies, such as checkpoint inhibitors (e.g., anti-PD1), may offer synergistic benefits. Future trials are likely to explore combination regimens that target both the myeloid and lymphoid compartments of the tumor, thereby creating a more comprehensive antitumor immune response. Moreover, the combination with traditional cytotoxic therapies may further enhance the overall therapeutic efficacy.
3. Expanding Non-Cancer Applications:
Although current clinical investigations primarily focus on cancer, especially solid tumors, emerging research suggests that engineered macrophages could be adapted for non-cancer indications. Areas such as autoimmune disease, chronic inflammatory conditions, tissue fibrosis, and even modulation of immune responses in transplantation offer exciting avenues for future exploration. These applications will require careful tailoring of the CAR constructs to modulate immune responses without causing excessive inflammation or tissue damage.
4. Personalized and Adaptive Therapy:
As our understanding of tumor biology and the TME deepens, there is an opportunity to develop personalized CAR-M therapies. Detailed tumor profiling could inform the selection of target antigens and the design of CAR constructs tailored to individual patients’ tumors. This personalized approach could enhance efficacy while minimizing risks. Furthermore, adaptive strategies that allow CAR-Ms to dynamically respond to changes in the TME over time could improve long-term outcomes.
5. Advanced Preclinical Models and Clinical Translation:
Finally, there is a significant need for improved preclinical models that more faithfully replicate the complexities of the human TME and tumor heterogeneity. Enhanced animal models, along with advanced in vitro systems such as organoids, will be critical in bridging the gap from bench to bedside. Such models will provide more robust safety and efficacy data, thereby accelerating the clinical translation of next-generation CAR-M therapies.
Conclusion
In summary, CAR-macrophage therapies represent a promising and rapidly evolving modality in immuno-oncology, with the primary indications being various types of cancer—especially solid tumors such as HER2-overexpressing tumors, gastrointestinal cancers, ovarian and pancreatic cancers, glioblastoma, and even some hematological malignancies. More recently, there is also growing preclinical interest in exploring non-cancer indications including inflammatory and fibrotic conditions as well as potential applications in transplantation tolerance. The current landscape is characterized by active early-phase clinical trials (e.g., CT-0508 for HER2-positive tumors) and extensive preclinical studies that underscore the dual mechanisms of direct tumor cell phagocytosis and TME modulation. Nevertheless, a number of technical and biological challenges remain, including efficient gene transfer, maintenance of a durable M1 phenotype in immunosuppressive environments, targeted trafficking and persistence, off-target safety concerns, and manufacturing scalability.
Looking forward, future research efforts are expected to refine CAR designs, explore combinatorial treatment regimens, and expand the range of target indications through personalized approaches. The continuous evolution of cell engineering techniques—including the adoption of nonviral gene transfer methods and iPSC-derived products—will be key in surmounting current hurdles and achieving broader clinical success. Ultimately, the promising preclinical and early clinical results support the notion that CAR-M therapies have the potential to redefine therapeutic strategies for hard-to-treat cancers and possibly other chronic diseases, paving the way for a new generation of cell-based immunotherapies.
In conclusion, CAR-M therapies are being investigated primarily for cancer indications with a strong focus on solid tumors, given their unique ability to infiltrate the dense tumor microenvironment and trigger both direct and indirect antitumor responses. As research matures, the potential expansion into non-cancer applications also presents exciting possibilities. The current breadth of clinical trials and preclinical studies, as detailed in the synapse-sourced literature, reflects a dynamic field that embraces both the complexity of modern oncology and the versatility of engineered immune cells. Continued collaborative efforts in translational research are essential to fully realize the clinical potential of CAR-Ms, ensuring improved therapeutic outcomes and diversified applications in the near future.