For what indications are CAR-NK being investigated?

17 March 2025
Introduction to CAR-NK Cell Therapy

Definition and Basic Concepts
Chimeric antigen receptor–engineered natural killer (CAR‐NK) cell therapy is an emerging immunotherapeutic strategy that seeks to harness the innate cytotoxic capacity of NK cells by genetically modifying them to express CARs. These engineered receptors are designed to specifically recognize tumor‐associated antigens (TAAs) on cancer cells and trigger an antitumor response. Unlike conventional T cells, NK cells can be used in an allogeneic “off‐the‐shelf” setting because they do not require strict HLA matching. The concept originated from the tremendous success of CAR‐T cells in hematological malignancies; however, mistakes that have been observed with CAR‐T therapy – such as high production costs, complex manufacturing requirements, and severe toxicities – have led researchers to investigate CAR‐NK cells as a potentially safer and more scalable option. CAR‐NK cells maintain their innate cytotoxicity, can be activated both via the engineered CAR‐signal and their natural repertoire of activating receptors, and even provide additional killing mechanisms such as antibody‐dependent cellular cytotoxicity (ADCC). In short, CAR‐NK therapy is defined by genetically modifying NK cells with a synthetic receptor that redirects their cytotoxicity toward target cancer cells, while also preserving their innate safety profile and making them ubiquitous for rapid clinical application.

Differences between CAR-NK and CAR-T Therapies
Although both CAR‐NK and CAR‐T cells use the CAR technology to recognize tumor antigens, there are several important differences that underpin their translational potential:
- Safety Profile: CAR‐NK cells have a shorter lifespan in vivo and secrete a distinct cytokine spectrum (e.g., predominantly IFN-γ and GM-CSF rather than IL-1, IL-6, or TNF-α), thereby reducing the incidence and severity of CRS, neurotoxicity, and graft-versus-host disease (GVHD).
- Allogeneic Utility: Unlike CAR‐T cells, which generally require autologous production to limit the risk of immune rejection, CAR‐NK cells can be produced from donor sources such as peripheral blood, umbilical cord blood, NK-92 cell lines, or even induced pluripotent stem cells (iPSCs). This lends itself to an “off‐the‐shelf” product that is less time‐ and cost-intensive.
- Mechanisms of Cytotoxicity: While CAR‐T cells rely mainly on CAR-mediated killing, NK cells have innate mechanisms such as recognition of “missing self” and natural cytotoxicity receptor (NCR)-mediated killing. As a result, CAR‐NK cells are equipped to target cancer cells even if the expression of the CAR-targeted antigen is heterogeneous or partially lost, often leading to more robust tumor cell clearance.
- Manufacturing and Expansion: CAR‐NK cells present unique challenges in genetic modification due to their transduction difficulty; however, advancements in viral and nonviral methodologies—as well as feeder systems and cytokine approaches—have enabled effective expanding and engineering.

Current Research on CAR-NK Indications

The preclinical and clinical investigations into CAR‐NK cell therapy have broadened their indications to include a wide array of cancers. Indications are largely grouped into two major categories: hematological malignancies and solid tumors.

Hematological Malignancies
CAR‐NK cell therapy was initially propelled by the clinical success of CAR‐T cells, and many studies have targeted blood cancers. The following are several key hematological indications:

1. B-Cell Malignancies:
Much of the early research in CAR‐NK therapy has focused on B-cell neoplasms. Preclinical studies demonstrated that CAR‐NK cells engineered to target CD19 on B-cell malignancies can induce significant cytotoxicity. Clinical trials have shown encouraging response rates with complete remissions being observed, albeit in early-phase studies. Moreover, these CAR‐NK cells appear to overcome some of the issues of antigen escape that can limit CAR‐T cell efficacy. For instance, engineered NK cells have effectively targeted CD19, while also leveraging their natural cytotoxic functions.

2. Acute Myeloid Leukemia (AML):
In addition to B-cell targets, AML has been a major focus. CAR‐NK cells engineered to target myeloid-specific antigens (such as CD33) have been under investigation, offering potential advantages by eliminating residual blasts without severe off-target toxicities. The inherent safety of NK cells makes them well suited for AML patients, where aggressive immune responses are a concern.

3. Other Hematologic Malignancies:
Beyond CD19+ malignancies and AML, indications also include other forms such as multiple myeloma, where targets like BCMA (B-cell maturation antigen) are of interest. Additional targets include ligands for the natural killer group 2, member D (NKG2D) receptor, where the engineered CAR can direct NK cells toward a broader range of myeloid and even lymphoid lineages. Some preclinical strategies have also looked at targeting T-cell malignancies by using CAR constructs against T-cell antigens (e.g., CD3, CD5, or CD7) so that CAR‐NK cells can be deployed against otherwise hard-to-treat T-cell neoplasms.

4. Combination and Universal Approaches:
Several studies also indicate the potential of integrating CAR‐NK cells with other immune therapies (such as checkpoint inhibitors) to treat hematological malignancies that may be resistant to conventional therapies. The design of universal CAR-NK cells from iPSC sources, for example, offers the possibility of broad application across patient cohorts without HLA matching. This approach is particularly promising given the observed durable responses and decreased toxicity profiles in early trials.

Solid Tumors
Although the initial clinical successes of CAR therapies were in hematologic cancers, a significant research focus in CAR‐NK cell therapy is addressing the challenges of targeting solid tumors. Solid tumor indications being investigated include:

1. Glioblastoma and Other Brain Tumors:
CAR‐NK cells engineered to target antigens overexpressed in glioblastoma (e.g., EGFR variant III or other glioma-associated markers) have shown promising preclinical results. Early-phase clinical trials have begun exploring intracranial administration to overcome the blood–brain barrier, with preclinical models demonstrating improved tumor infiltration and cytotoxicity.

2. Breast Cancer:
There are multiple studies where CAR‐NK cells are being engineered to target HER2/neu overexpressed in several types of breast cancers. These CAR‐NK cells have displayed enhanced cytotoxicity against HER2-expressing tumor cells, and preclinical work suggests that they can overcome the immunosuppressive microenvironment typically observed in solid tumors. In one detailed preclinical study, HER2 CAR‐NK cells demonstrated higher killing efficiency of breast cancer cells compared to donor-matched HER2 CAR‐T cells, while avoiding damage to healthy cells with low HER2 expression.

3. Ovarian and Pancreatic Cancers:
Ovarian cancer studies have investigated CAR‐NK cells targeting mesothelin or folate receptor α (FRα), two antigens that are frequently overexpressed in ovarian and pancreatic ductal adenocarcinoma (PDAC) tumors. Preclinical data indicate that CAR‐NK cells can localize to tumor sites and mediate effective tumor cell lysis in xenograft models, with additional evidence showing that combination strategies with oncolytic viruses or radiotherapy may synergistically improve outcomes.

4. Colorectal Cancer and Gastrointestinal Malignancies:
Investigations into gastrointestinal cancers, including colorectal carcinoma, have seen the development of CAR‐NK cells targeting antigens such as B7-H3 and PDL-1. These approaches aim to exploit the ability of CAR‐NK cells to counteract the immunosuppressive tumor microenvironment. Early-phase clinical trials, although scarce, are beginning to report on safety and biodistribution data for such solid tumor indications.

5. Lung Cancer:
Advanced preclinical models have also focused on lung cancers. The engineered NK cells target markers that are prevalent on non-small cell lung cancer (NSCLC) cells, and their ability to migrate, persist, and overcome inhibitory signals in the tumor microenvironment are being rigorously evaluated. The combination of CAR‐NK cells with radiotherapy is particularly promising in lung cancer by modulating the tumor milieu to favor NK cell activity.

6. Other Solid Malignancies:
Additional investigations include melanoma, ovarian carcinoma beyond PDAC, and even advanced head and neck squamous cell carcinoma (HNSCC), where the diverse target antigen repertoire of CAR‐NK cells—including both CAR-mediated and native receptor-mediated killing—is seen as advantageous. Furthermore, multiplex targeting strategies that incorporate dual or tandem CAR constructs are being explored to address tumor heterogeneity and the risk of antigen escape.

Mechanisms and Effectiveness

Mechanisms of Action
CAR‐NK cells are designed to utilize both CAR‐mediated and innate cytotoxic pathways to target cancer cells. Their mechanisms of action include:
- CAR-Dependent Cytotoxicity:
Upon engagement of the CAR binding domain with its target antigen, a signal transduction cascade is triggered that leads to the release of perforins, granzymes, and death ligands like FasL or TRAIL. This mechanism is similar to that observed in CAR‐T cells but tends to be less associated with a cytokine storm because NK cells secrete a different cytokine profile (mainly IFN-γ and GM-CSF).
- CAR-Independent Killing:
NK cells naturally express activating receptors such as NKG2D, NKp30, and DNAM-1, which enable them to recognize and kill tumor cells even in the absence of CAR engagement. This dual functionality is particularly useful when the target antigen is downregulated by tumor cells, a phenomenon known as antigen escape.
- ADCC:
Additionally, the presence of Fc receptors (such as CD16) on NK cells allows them to mediate antibody-dependent cellular cytotoxicity, which is potentiated when used in combination with monoclonal antibodies.
- Modulation of the Tumor Microenvironment (TME):
Several studies have demonstrated that engineered NK cells can secrete cytokines that remodel the TME to be more permissive for immune cell infiltration and tumor cell killing. This “armoring” of CAR‐NK cells by adding cytokine transgenes (e.g., IL-15) or using additional gene switches is a key mechanism to improve persistence and tumor targeting in hostile microenvironments.

Preclinical and Clinical Trial Results
Preclinical studies across a variety of hematological and solid tumor models have illustrated several promising features of CAR‐NK cells:
- In Hematological Models:
Preclinical models using CAR‐NK cells targeting CD19 demonstrated effective killing of B-cell malignancies and were associated with complete remissions in animal models. Early-phase clinical trials (for example, in patients with relapsed or refractory B-cell malignancies) indicated that CAR‐NK cells could induce rapid responses with low incidences of toxicity.
- In Solid Tumor Models:
In solid tumor models (such as breast, ovarian, and glioblastoma), CAR‐NK cells were observed to have superior tumor infiltration, higher cytotoxicity, and the ability to eradicate tumor cells in both in vitro and in vivo settings. Studies showed that HER2-targeting CAR‐NK cells could significantly restrain tumor growth without damaging normal tissues.
- Clinical Observations:
Phase I clinical trials have reported that CAR‐NK cells, particularly those derived from cord blood and engineered with multiple functional domains (e.g., a membrane-bound IL-15 and safety switches), can induce complete responses in patients with hematological malignancies without the severe adverse events typical of CAR‐T therapies. Additionally, clinical studies using CAR‐NK cells in the context of solid tumors are in early phases, but preclinical and early clinical evidence is mounting which supports their further development.

Challenges and Future Prospects

Current Challenges
Despite the promising indications and early successes, several challenges remain in the clinical development of CAR‐NK cell therapies:
- Genetic Engineering and Transduction Efficiency:
NK cells historically pose challenges in gene transduction due to lower viral transduction rates relative to T cells. While innovative methods such as lentiviral transduction with cytokine cocktails, electroporation, and even improved non-viral gene editing (e.g., CRISPR/Cas9) have increased efficiency, robust and standardized protocols are still needed to ensure clinical-scale manufacturing.
- Persistence and In Vivo Expansion:
One of the major hurdles in CAR‐NK cell therapy is their relatively short lifespan in vivo post-infusion, which may limit their long-term efficacy. Approaches such as engineering NK cells to express cytokines like IL-15 or utilizing memory-like NK cell technology are being explored to prolong persistence and enhance antitumor activity.
- Tumor Microenvironment (TME):
Solid tumors represent a hostile microenvironment with immunosuppressive factors, hypoxia, and physical barriers that limit infiltration. Although CAR‐NK cells have been engineered to withstand such conditions, further improvements are needed to ensure effective trafficking, persistence, and activity within the TME.
- Antigen Escape and Heterogeneity:
Tumor cells can lose or downregulate the target antigens, leading to therapy resistance. The inherent dual killing mechanism of NK cells helps address this, but further strategies, including multispecific CAR constructs or combined therapies with checkpoint inhibitors, are needed to minimize relapse risk.
- Scalability and Allogeneic Product Safety:
While the promise of an “off-the‐shelf” product is a major advantage, ensuring a reliable and cost-effective manufacturing process while maintaining product consistency remains a challenge. This includes addressing issues such as cell expansion, standardization of feeder-cell methods, and ensuring safety regarding allogeneic use.

Future Research Directions
Looking forward, research is focusing on multiple strategies to overcome current challenges and expand the indications for CAR‐NK cells:
- Optimizing CAR Designs:
Next-generation CAR constructs tailored specifically to NK cell biology are under development. These include modifications to costimulatory domains tailored for NK cells and the integration of safety switches and cytokine support systems. For example, dual or tandem CAR designs aimed at targeting multiple antigens at once could improve efficacy in heterogeneous tumors.
- Enhanced Persistence Strategies:
Future approaches include the use of armored CAR‐NK cells that can secrete immune-stimulatory cytokines (e.g., IL-15 or IL-12) in a controlled manner, as well as strategies to induce a memory-like phenotype that confers long-term antitumor activity, particularly in the treatment of chronic or aggressive malignancies.
- Combination Therapies:
A promising area for future research is combining CAR‐NK cell therapy with other treatment modalities. Radiotherapy, chemotherapy, monoclonal antibodies, and immune checkpoint blockade are all being evaluated as adjuncts that could synergistically improve the efficacy of CAR‐NK cells in treating both hematological and solid tumors. For example, combining radiotherapy with CAR‐NK cell therapy has shown potential in modulating the TME, increasing tumor antigen presentation, and enhancing NK cell infiltration.
- Expanding the Range of Target Antigens:
In hematological malignancies, novel targets beyond CD19, CD33, and BCMA are being explored. Similarly, in solid tumors, expanding the repertoire to include antigens such as HER2, mesothelin, FRα, GD2, and B7-H3 is under active investigation. Research is also moving toward developing “universal” CAR platforms derived from iPSCs that can be easily retargeted to different antigens with minimal re-engineering.
- Leveraging Novel Sources and Manufacturing Approaches:
The translation of CAR‐NK therapy into off‐the‐shelf products is being facilitated by advances in deriving NK cells from iPSCs, cord blood, and NK cell lines such as NK‐92. Continued innovation in bioreactor technologies and genetic manipulation platforms (e.g., CRISPR/Cas9 gene editing, nanochannel electroporation) will further streamline the production process, improve consistency, and reduce manufacturing costs.
- Rigorous Clinical Trials and Regulatory Pathways:
As more clinical data emerge from early-phase trials, further research should prioritize the establishment of standardized endpoints, toxicity profiles, and long-term safety studies. This will pave the way for later-stage trials and eventual regulatory approvals, thereby broadening the clinical indications for CAR-NK cells across a range of malignancies.

Conclusion
In summary, CAR-NK cell therapy is being investigated for a broad spectrum of indications that span both hematological malignancies and solid tumors. In the context of hematologic cancers, CAR-NK cells have been primarily explored in B-cell neoplasms, AML, multiple myeloma, and even T-cell malignancies, leveraging their dual mechanisms of CAR-dependent and innate cytotoxicity while demonstrating a favorable safety profile and lower risk of adverse events. In parallel, the treatment of solid tumors such as glioblastoma, breast cancer, ovarian and pancreatic cancers, colorectal carcinoma, lung cancer, and other advanced malignancies is emerging as a promising field. Preclinical models and early-phase clinical trials have provided encouraging evidence that engineered CAR-NK cells can effectively infiltrate tumors, execute target-specific killing, and potentially overcome the challenges posed by a suppressive tumor microenvironment.

From a mechanistic standpoint, CAR-NK cells operate by merging the precision of genetically encoded receptors with their native ability to eliminate tumor cells through various cytotoxic pathways such as granule exocytosis, death ligand interactions, and ADCC. Although challenges persist—particularly in the realms of efficient gene transduction, limited in vivo persistence, tumor antigen heterogeneity, and manufacturing scalability—ongoing research is addressing these hurdles through improved CAR designs, combination therapies, and innovative cell sources.

The future of CAR-NK cell therapy holds promise not only for the treatment of refractory hematological diseases but also as a transformative approach for solid tumors where conventional immunotherapies have fallen short. As research advances, the refinement of CAR constructs specific to NK cell biology, strategies to boost persistence and trafficking into the tumor microenvironment, and integrated combination approaches will be key to unlocking the full therapeutic potential of CAR-NK cells. The integration of these novel strategies indicates that CAR-NK cells may soon complement or even surpass existing CAR-T cell therapies in certain contexts, offering a safer, more accessible, and effective immunotherapy for a wide range of cancers.

Overall, while many obstacles remain, the current trajectory of research—ranging from early mechanistic studies to advanced clinical trials—demonstrates remarkable progress and underscores the robust potential of CAR-NK cell therapy to revolutionize treatment protocols for both hematological malignancies and solid tumors. Continued interdisciplinary collaboration, technological refinement, and clinical validation are essential steps to achieve widespread clinical adoption and improve patient outcomes across diverse cancer indications.

In conclusion, CAR-NK cells, with their unique safety profile, dual mechanism of action, and potential for mass production from allogeneic sources, are being investigated for indications spanning aggressive blood cancers and challenging solid tumors. Their ongoing development and the broadening of target antigens along with combination strategies reflect a dynamic field poised to overcome persistent challenges and deliver next-generation immunotherapies. The future research directions aiming at enhanced persistence, optimized CAR engineering, and synergistic treatment combinations hold the promise of translating CAR-NK cell research into therapeutic breakthroughs that may redefine the standards of cancer care across multiple indications.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成