MedPath

Nanoparticle-Based Drug Delivery Systems Show Promise for Enhancing Cancer Immunotherapy in Solid Tumors

4 months ago6 min read

Key Insights

  • Nanoparticle-based drug delivery systems (NDDS) offer significant advantages over traditional immunotherapy approaches by improving drug targeting, reducing toxicity, and enhancing therapeutic efficacy through mechanisms like immunogenic cell death induction and tumor microenvironment reprogramming.

  • Recent research demonstrates that multifunctional nanocarriers can simultaneously deliver STING agonists and PD-L1 siRNA to activate immune responses while preventing immune escape, achieving superior tumor control in melanoma and breast cancer models.

  • The combination of nanomedicine with photodynamic therapy and immune checkpoint modulation represents a promising strategy for converting immunologically "cold" tumors into "hot" tumors responsive to immunotherapy.

Nanoparticle-based drug delivery systems (NDDS) are emerging as a transformative approach to overcome the limitations of current cancer immunotherapy, offering new hope for treating solid tumors that remain resistant to conventional immune checkpoint inhibitors. While immunotherapy has revolutionized cancer treatment since the introduction of immune checkpoint inhibitors, only a small percentage of patients respond to these therapies, particularly those with poorly immunogenic "cold" tumors.

Addressing Current Immunotherapy Limitations

Traditional immunotherapy faces several significant challenges that limit its effectiveness. Immune checkpoint inhibitors such as PD-1/PD-L1 and CTLA-4 inhibitors show limited efficacy due to immune-related adverse events, therapeutic resistance, high costs, and restricted patient populations. According to recent research, while these treatments have shown promise in various clinical trials for solid tumors, their success is often hampered by the immunosuppressive tumor microenvironment and low tumor immunogenicity.
The tumor microenvironment plays a crucial role in promoting immunosuppression through the recruitment of regulatory cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and regulatory T cells (Tregs), while simultaneously reducing essential immune cells like cytotoxic T lymphocytes (CTLs), natural killer cells, and dendritic cells.

Advantages of Nanoparticle-Based Delivery Systems

NDDS offer multiple advantages over traditional immunotherapy approaches. These systems, primarily comprising particles with diameters less than 100 nm, utilize distinctive properties such as acoustic, electric, optical, magnetic, and thermal attributes to facilitate targeted drug delivery to specific cells or tissues.
Key advantages include enhanced permeability and retention effects, improved solubility of hydrophobic drugs, active targeting delivery capabilities, reduced physiological barriers, integration of diagnosis and treatment, and stimulus-responsive intelligent drug release. Since the FDA approval of the first liposome nanodrug Doxil in 1995, numerous nanomedicine formulations have been developed for cancer treatment.

Mechanisms of Enhanced Immunotherapy

NDDS enhance immunotherapy through two primary mechanisms: induction of immunogenic cell death (ICD) and modulation of the immunosuppressive microenvironment. ICD is a critical process where tumor cells release damage-associated molecular patterns (DAMPs), including calreticulin, ATP, heat shock proteins, and high mobility group protein 1 (HMGB1), which initiate anti-tumor immune responses.

Nano-based Chemo-immunotherapy

Chemotherapy drugs such as doxorubicin, epirubicin, oxaliplatin, and others can induce ICD when delivered via nanoparticles. Recent studies have shown that nano-formulations combining chemotherapy agents with immunotherapy can transform "cold" tumor microenvironments into "hot" ones, characterized by increased numbers of CD8+ T cells, CD4+ T cells, dendritic cells, and pro-inflammatory cytokines while reducing immunosuppressive cells.
For example, researchers have developed binary cooperative prodrug nanoparticles that release oxaliplatin and IDO-1 inhibitors in acidic environments, achieving stronger enhanced permeability and retention effects and promoting T lymphocyte infiltration. Similarly, Chinese herbal medicine compounds like ginsenoside Rg3 and icariin have been successfully encapsulated in nanoparticles to enhance their ICD-inducing capabilities.

Ablative Therapy Enhancement

NDDS significantly improve ablative treatments including radiotherapy, photodynamic therapy (PDT), photothermal therapy (PTT), and sonodynamic therapy (SDT). These treatments act as type II ICD inducers, generating reactive oxygen species and stimulating endoplasmic reticulum stress responses.
Radiotherapy combined with nanoparticles has shown particular promise. Gold nanoparticles can enhance radiation sensitivity by catalyzing free radical generation and very low-energy electrons, while protecting surrounding normal tissues from radiation toxicity. Studies have demonstrated that 14-nm gold nanoparticles significantly enhance radiotherapy-induced ICD and increase macrophage infiltration in tumor tissue.
PDT-based nanoplatforms have also shown remarkable results. Multifunctional nanoparticles combining photosensitizers with other therapeutic agents can generate singlet oxygen to induce tumor elimination while providing real-time imaging guidance for treatment optimization.

Advanced Multifunctional Nanocarriers

Recent breakthrough research has demonstrated the development of sophisticated multifunctional nanocarriers that can simultaneously address multiple aspects of cancer immunotherapy. These advanced systems combine STING agonists with PD-L1 siRNA delivery, creating synergistic effects that both activate immune responses and prevent immune escape.
The engineered nanocarriers utilize galactose targeting ligands to specifically bind to glucose transporters overexpressed on cancer cells, ensuring preferential tumor accumulation. Upon cellular uptake, the nanocarriers respond to the tumor's redox environment and acidic pH to release their therapeutic cargo. The STING agonist cGAMP activates the STING pathway, leading to interferon production and immune cell activation, while the PD-L1 siRNA prevents the upregulation of immune checkpoint proteins that would otherwise allow tumor cells to escape immune surveillance.
In preclinical studies, these multifunctional nanocarriers demonstrated superior efficacy in both B16F10 melanoma and 4T1 breast cancer models. The treatment not only eliminated primary tumors but also suppressed distant metastases and generated long-term immune memory, with survival rates reaching 75% in treated mice compared to rapid mortality in control groups.

Tumor Microenvironment Reprogramming

Beyond ICD induction, NDDS can directly reprogram the immunosuppressive tumor microenvironment through several mechanisms. These include enhancing immune cell infiltration, reducing immunosuppressive cell populations, improving hypoxic conditions, and activating important immune-related signaling pathways.
Nanoparticles can effectively target and eliminate MDSCs, reprogram M2 macrophages to M1 phenotypes, reduce regulatory T cell populations, and enhance dendritic cell maturation. For instance, synthetic nanoparticle antibodies engineered with specific peptides can precisely target MDSCs for elimination, while other formulations can deliver mRNA encoding cytokine inhibitors to reverse immune suppression.
The hypoxic and acidic tumor microenvironment, which contributes to therapeutic resistance, can be addressed through MnO2-based nanomaterials that improve oxygen levels while simultaneously consuming glutathione, a key antioxidant that protects cancer cells from therapy-induced damage.

Clinical Translation Challenges and Future Prospects

Despite promising preclinical results, the clinical translation of NDDS-based immunotherapy faces several challenges. Most successful cases have been demonstrated only in animal models, with limited clinical application to date. Key challenges include optimizing nanoparticle stability under physiological conditions, ensuring consistent tumor targeting, minimizing off-target effects, and scaling up manufacturing processes.
However, the potential benefits of NDDS-based immunotherapy are substantial. These systems offer the possibility of personalized treatment approaches, reduced systemic toxicity, improved patient compliance, and enhanced therapeutic efficacy for previously treatment-resistant tumors.
Future research directions include developing more sophisticated targeting mechanisms, creating stimuli-responsive systems that can adapt to changing tumor conditions, and combining multiple therapeutic modalities within single nanoplatforms. The integration of artificial intelligence and machine learning approaches may also help optimize nanoparticle design and predict treatment responses.
The field of nanomedicine-enhanced immunotherapy represents a rapidly evolving area with significant potential to transform cancer treatment. As researchers continue to refine these technologies and address current limitations, NDDS-based approaches may soon provide new therapeutic options for patients with solid tumors who currently have limited treatment alternatives.
Subscribe Icon

Stay Updated with Our Daily Newsletter

Get the latest pharmaceutical insights, research highlights, and industry updates delivered to your inbox every day.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.