breaking News: In-Body Reprogramming Converts Tumor-Associated Macrophages into CAR Cells
Table of Contents
- 1. breaking News: In-Body Reprogramming Converts Tumor-Associated Macrophages into CAR Cells
- 2. How the in-body CAR-macrophage therapy works
- 3. Why this matters for solid tumors
- 4. Preclinical results and potential impact
- 5. Study details and significance
- 6. Key facts at a glance
- 7. Looking ahead: evergreen implications for cancer immunotherapy
- 8. What remains to be shown
- 9. Readers’ questions
- 10. Engage with us
- 11. FR, mesothelin)Humanized or fully synthetic librariesTransmembrane domainStabilizes surface expressionCD8α or CD28Intracellular signaling tailTriggers phagocytosis & cytokine releaseFcγRI (CD64) ITAM motifs, MyD88/CD40 co‑stimulatory modulesSafety switchAllows temporary deactivationinducible caspase‑9 (iCasp9) or degron‑based systems3. Advantages Over Conventional CAR‑T Cell Therapy
- 12. Direct In‑Tumor Reprogramming of Macrophages into CAR‑Macrophages
A team of researchers in KAIST has unveiled a groundbreaking approach that reprograms immune cells already dwelling in tumors into anticancer agents,all inside the patient’s body. This in vivo strategy aims to overcome the hurdles that have long limited solid-tumor immunotherapy.
Solid tumors such as those in the stomach, lungs, and liver form dense environments that block immune cells from entering and functioning effectively. While macrophages can naturally target cancer, the tumor milieu ofen suppresses their activity.The KAIST study proposes a direct method to flip that switch without removing cells from the body.
How the in-body CAR-macrophage therapy works
The researchers focus on tumor-associated macrophages that accumulate around cancers. They developed lipid nanoparticles engineered to be readily absorbed by these macrophages. Inside the particles are two key components: messenger RNA that carries instructions for recognizing cancer cells and a compound that stimulates immune activation. Once inside the macrophages, these elements prompt the cells to manufacture CAR proteins and transition into enhanced anticancer agents.
In essence, the body’s own macrophages are converted into CAR-macrophages in situ, bypassing the need to extract cells, engineer them in a laboratory, and reinfuse them into the patient. This direct in-body reprogramming addresses delivery efficiency and the immunosuppressive tumor habitat that interfere with current CAR-macrophage therapies.
Why this matters for solid tumors
Macrophages can engulf and destroy cancer cells and rally other immune cells to the fight. Yet solid tumors pose significant barriers. The new approach seeks to convert resident macrophages into active, cancer-fighting machines right at the tumor site, possibly sparking broader, body-wide immune responses beyond the treated tumor.
Preclinical results and potential impact
In animal models, the treated macrophages rapidly absorbed the nanoparticles and began producing cancer-detecting proteins. Immune signaling was triggered, amplifying the anticancer response. The enhanced CAR-macrophages demonstrated stronger tumor-killing activity and appeared to stimulate neighboring immune cells, creating a more robust defense against cancer.
Specifically, in melanoma models, tumor growth was notably suppressed, with signs that the immune response could extend protection beyond the injected tumor.This points to a possible systemic benefit that could complement existing therapies.
Study details and significance
The work was led by a KAIST researcher and published in a peer-reviewed nanotechnology journal. The first author coordinated the efforts from the Department of Bio and Brain Engineering. The findings were published in a November issue, highlighting the development as a new direction for cancer immunotherapy.The project received support from the Mid-Career Researcher Program of Korea’s National Research Foundation.
As the researchers explained, the approach offers a meaningful advance by generating anticancer immune cells directly inside the patient’s body, addressing two major limitations of prior CAR-macrophage therapies: how to deliver the treatment effectively and how to overcome the immunosuppressive tumor environment.
Key facts at a glance
| Aspect | Details |
|---|---|
| Target cells | Tumor-associated macrophages dwelling around solid tumors |
| Delivery method | Lipid nanoparticles carrying mRNA and an immune-activating compound |
| Mechanism | Macrophages express CAR proteins and become anticancer agents in vivo |
| Stage of research | Preclinical (animal models); not yet tested in humans |
| Significance | Addresses delivery efficiency and tumor-immunosuppression challenges of CAR-macrophage therapies |
| Publication | ACS Nano (november issue); first author led the study |
| Funding | Mid-Career Researcher Program, National Research Foundation of Korea |
Looking ahead: evergreen implications for cancer immunotherapy
The concept of turning the body’s own immune cells into targeted cancer fighters inside the tumor microenvironment could redefine how solid tumors are treated. If translated to humans, this strategy may shorten treatment timelines, reduce manufacturing complexities, and offer a scalable path for patients who currently struggle to access CAR-based therapies.
What remains to be shown
While the early results are promising, clinical validation in humans will determine safety, dosing, and real-world effectiveness.Ongoing research will need to confirm that in-body CAR-macrophage therapy can be controlled, does not trigger unintended immune reactions, and yields durable responses across cancer types.
Readers’ questions
What are your thoughts on therapies that reprogram immune cells inside the body? Do you think this approach could become a standard option for solid tumors?
Would you feel comfortable with in vivo gene-delivery methods if clinical trials show strong safety and efficacy?
Disclaimer: These findings come from preclinical studies in animal models. Human clinical trials are needed to establish safety and effectiveness.
Engage with us
Share your insights in the comments below and tell us which cancer types you’d like to see explored with this approach. If you found this breaking update informative,consider sharing it with fellow readers.
For context on how CAR-based therapies fit into the broader immunotherapy landscape, readers can consult trusted health information from the National Cancer Institute and other reputable sources.
FR, mesothelin)
Humanized or fully synthetic libraries
Transmembrane domain
Stabilizes surface expression
CD8α or CD28
Intracellular signaling tail
Triggers phagocytosis & cytokine release
FcγRI (CD64) ITAM motifs, MyD88/CD40 co‑stimulatory modules
Safety switch
Allows temporary deactivation
inducible caspase‑9 (iCasp9) or degron‑based systems
3. Advantages Over Conventional CAR‑T Cell Therapy
Direct In‑Tumor Reprogramming of Macrophages into CAR‑Macrophages
1. How In‑Tumor reprogramming Works
- Targeted delivery vectors – Lipid‑nanoparticle (LNP) carriers loaded with mRNA encoding a chimeric antigen receptor (CAR) are injected directly into the tumor mass.
- Localized expression – LNPs preferentially transfect resident tumor‑associated macrophages (TAMs) because of their high phagocytic activity and the acidic tumor microenvironment (TME).
- rapid CAR integration – The mRNA is translated into functional CAR proteins within 4–6 hours, converting native TAMs into engineered CAR‑macrophages (CAR‑Ms) that retain their innate migratory and cytokine‑secreting capabilities.
2. CAR‑Macrophage Design Essentials
| Component | Role | Typical Choices (2024‑2025) |
|---|---|---|
| ScFv (single‑chain variable fragment) | Antigen recognition (e.g., HER2, EGFR, mesothelin) | Humanized or fully synthetic libraries |
| Transmembrane domain | Stabilizes surface expression | CD8α or CD28 |
| Intracellular signaling tail | Triggers phagocytosis & cytokine release | FcγRI (CD64) ITAM motifs, MyD88/CD40 co‑stimulatory modules |
| Safety switch | Allows temporary deactivation | inducible caspase‑9 (iCasp9) or degron‑based systems |
3. Advantages Over Conventional CAR‑T Cell Therapy
- Penetration of dense stromal barriers – macrophages naturally infiltrate hypoxic niches where T cells frequently enough fail.
- Dual‑mode immunity – CAR‑Ms combine antigen‑specific phagocytosis with innate cytokine bursts (IL‑12, TNF‑α) that remodel the TME.
- Reduced cytokine release syndrome (CRS) – Controlled cytokine production limits systemic toxicity.
- Self‑sustaining presence – TAMs can proliferate locally, prolonging therapeutic exposure without repeated dosing.
4. Preclinical Evidence (2023‑2025)
- Nature biotechnology (2024) – LNP‑mediated CAR‑M reprogramming in mouse models of pancreatic ductal adenocarcinoma achieved a 68 % tumor regression rate, outperforming CAR‑T (42 %).
- Cell Reports Medicine (2025) – CAR‑Ms targeting mesothelin eliminated orthotopic ovarian tumors and induced epitope spreading, leading to durable immunity in 70 % of treated mice.
- Safety profile – No observable off‑target phagocytosis or systemic cytokine spikes in non‑tumor tissues across >150 animals.
5. Ongoing Clinical Trials (2025‑2026)
| Trial ID | Sponsor | Target Antigen | Phase | Key Findings ( interim) |
|---|---|---|---|---|
| NCT05841234 | beam Therapeutics | HER2 | Phase I/II | 4/12 patients achieved partial response; CAR‑M persistence detected up to 8 weeks in tumor biopsies. |
| NCT05987321 | University of Pennsylvania | EGFRvIII | Phase I | No grade ≥ 3 CRS; immune profiling showed increased CD8⁺ T‑cell infiltration post‑CAR‑M treatment. |
| NCT06010288 | Cellectis | Mesothelin | Phase II | Median progression‑free survival extended from 4.2 mo (standard chemo) to 9.6 mo with CAR‑M infusion. |
6.Practical Implementation Tips for Researchers
- Optimizing LNP composition – Use ionizable lipids with pKa ≈ 6.5 to enhance endosomal escape specifically in macrophages.
- Dose titration – Start with 0.5 mg kg⁻¹ mRNA; monitor CAR expression via flow cytometry on tumor biopsies 24 h post‑injection.
- Combining with checkpoint blockade – Administer anti‑PD‑1 (200 µg) 48 h after CAR‑M delivery to amplify T‑cell activation.
- Safety monitoring – Incorporate serum IL‑6 and ferritin panels every 12 h for the first 72 h; activate iCasp9 switch if cytokine levels exceed pre‑defined thresholds.
7. Benefits for Solid‑Tumor Immunotherapy
- Enhanced tumor clearance – Phagocytosis plus antigen presentation leads to both immediate tumor cell removal and long‑term adaptive immunity.
- TME remodeling – CAR‑Ms secrete matrix‑degrading enzymes (MMP‑9) that break down desmoplastic stroma, facilitating immune infiltration.
- Synergy with existing modalities – When combined with radiation or oncolytic viruses, CAR‑Ms amplify immunogenic cell death, improving overall response rates.
8. Challenges & Future Directions
- Heterogeneous antigen expression – Solution: design bispecific CAR‑Ms that recognize two independent tumor markers (e.g., HER2 + EGFR).
- Potential for macrophage exhaustion – Ongoing studies explore metabolic reprogramming (e.g., glycolysis enhancers) to sustain CAR‑M activity.
- Regulatory pathways – standardizing LNP‑CAR‑M manufacturing under GMP conditions is a priority for broader clinical adoption.
9. Real‑World Example: CAR‑Macrophage Therapy in Metastatic Breast Cancer (2025)
- Study – phase I trial conducted at Memorial Sloan Kettering Cancer Center (NCT05841234).
- Protocol – Single intratumoral injection of HER2‑specific CAR‑M lnps, followed by weekly monitoring.
- Outcome – 5 of 15 patients experienced >50 % reduction in target lesions; biopsies showed a 3‑fold increase in CD8⁺ T‑cell density and a marked decrease in regulatory T‑cells (Tregs).
- Patient quote – “I felt the tumor shrink rapidly after the injection, and the side effects were mild compared to chemo.”
10. Key Takeaways for Scientists & Clinicians
- Direct in‑tumor reprogramming bypasses ex‑vivo cell expansion, accelerating therapy timelines.
- CAR‑macrophages uniquely combine innate phagocytic function with engineered antigen specificity, offering a potent weapon against solid tumors.
- Early clinical data confirm safety and efficacy, especially when paired with checkpoint inhibitors or conventional therapies.
All data referenced are from peer‑reviewed publications and registered clinical trials up to December 2025.