Back to Journals » International Journal of Nanomedicine » Volume 19

Synergistic Potential of Nanomedicine in Prostate Cancer Immunotherapy: Breakthroughs and Prospects

Authors Jiang Y, Wang C, Zu C, Rong X, Yu Q, Jiang J

Received 28 February 2024

Accepted for publication 16 August 2024

Published 2 October 2024 Volume 2024:19 Pages 9459—9486

DOI https://doi.org/10.2147/IJN.S466396

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Dongwoo Khang



Yueyao Jiang,1,* Chengran Wang,2,* Chuancheng Zu,3,* Xin’ao Rong,2 Qian Yu,1 Jinlan Jiang2

1Department of Pharmacy, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China; 2Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China; 3China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Qian Yu, Department of Pharmacy, China–Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin Province, 130033, People’s Republic of China, Email [email protected] Jinlan Jiang, Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, Jilin Province, 130033, People’s Republic of China, Email [email protected]

Abstract: Given the global prevalence of prostate cancer in men, it is crucial to explore more effective treatment strategies. Recently, immunotherapy has emerged as a promising cancer treatment due to its unique mechanism of action and potential long-term effectiveness. However, its limited efficacy in prostate cancer has prompted renewed interest in developing strategies to improve immunotherapy outcomes. Nanomedicine offers a novel perspective on cancer treatment with its unique size effects and surface properties. By employing targeted delivery, controlled release, and enhanced immunogenicity, nanoparticles can be synergized with nanomedicine platforms to amplify the effectiveness of immunotherapy in treating prostate cancer. Simultaneously, nanotechnology can address the limitations of immunotherapy and the challenges of immune escape and tumor microenvironment regulation. Additionally, the synergistic effects of combining nanomedicine with other therapies offer promising clinical outcomes. Innovative applications of nanomedicine include smart nanocarriers, stimulus-responsive systems, and precision medicine approaches to overcome translational obstacles in prostate cancer immunotherapy. This review highlights the transformative potential of nanomedicine in enhancing prostate cancer immunotherapy and emphasizes the need for interdisciplinary collaboration to drive research and clinical applications forward.

Keywords: prostate cancer, nanomedicine, immunotherapy, TME

Graphical Abstract:

Introduction

Prostate cancer is the second most common malignancy in men worldwide and is a leading cause of death among males.1 Androgen deprivation therapy (ADT) is the main treatment for advanced prostate cancer. However, most patients who receive ADT ultimately progress to castration-resistant prostate cancer (CRPC), with a median survival of less than two years.2 Recent advancements in treatment have moderately improved survival rates for CRPC patients. However, the overall prognosis remains poor, and advanced prostate cancer is currently untreatable.3,4 Hence, advanced treatment strategies are required to improve patient survival and prognosis.

The development of cancer immunotherapy has represented a significant milestone in oncology.5,6 Immunotherapy, encompassing immune checkpoint inhibitors, antibody–drug conjugates (ADCs), cell therapy, and vaccine therapy, activate or regulate the immune system to eliminate cancer cells and achieve therapeutic effects.7,8 The 2018 Nobel Prize in Physiology or Medicine was awarded for groundbreaking research on immune checkpoint molecules, specifically cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed death receptor 1 (PD-1), which have significantly advanced the field of immunotherapy.9 Although immunotherapy has become an important tool in prostate cancer treatment, activating the immune system to recognize and destroy cancer cells, it faces several challenges, including immune escape mechanisms that allow cancer cells to evade surveillance and attack by the immune system, immune-related side effects caused by treatment,10 and inconsistent efficacy among patients.11 Therefore, an urgent need exists to develop new treatment strategies to improve immunotherapy outcomes.

Nanomedicine has shown great potential in this context as the application of nanotechnology in tumor therapy.12 Indeed, nanotechnology holds immense promise in advancing the treatment,13,14 prevention,15,16 monitoring,17 and management of biological diseases.18,19 By integrating nano- and drug technologies, nanotechnology facilitates the development of drug delivery systems and enhances immunotherapy effectiveness by influencing the microbiome, enabling photothermal and photogenetic therapy, and facilitating gene therapy.20,21 To specifically address the ineffectiveness of checkpoint blockade in “cold” tumors, nanomedicine can regulate the immune and mechanical properties of the tumor microenvironment (TME) to enhance radiotherapy/radiopharmacology.22 Notably, combining immunotherapy with traditional therapies, such as chemotherapy or radiation therapy, can leverage the strengths and mitigate the weaknesses of each individual treatment. Moreover, by integrating immunotherapeutic modalities and nanomaterials with other treatment modalities, novel opportunities for enhanced cancer therapy may be realized. Therefore, this review discusses the role, synergistic approach, clinical transformation, and future directions of nanotechnology in prostate cancer immunotherapy.

Mechanisms of Nanomedical Applications in Immunotherapy of Prostate Cancer

Traditional ADT exhibits suboptimal efficacy in patients with CRPC, necessitating the development of more efficacious treatment modalities to augment prostate cancer management. Prostate cancer is characterized by its heterogeneity23 and propensity for drug resistance,24 resulting in slow clinical progression and complicating traditional chemotherapy efforts, prone to recurrence and regeneration.25 The integration of nanomedicine into prostate cancer immunotherapy is a promising approach for improving therapeutic efficacy while reducing adverse reactions. This section explores the synergistic potential of nanodrug platforms, such as nanoparticles, liposomes, and dendrimers, in prostate cancer immunotherapy. We discuss their mechanisms of action, including targeted delivery, controlled release, and enhanced immunogenicity, as well as recent advances and key studies demonstrating their effectiveness in the field.

Cancer Immunotherapy Mechanisms

Innate immunity is the body’s first line of defense, comprising innate immune cells and soluble recognition molecules, such as natural antibodies and lectins. This process does not depend on antigen specificity and is not caused by brief induction.26 When the body’s initial barrier is breached, an adaptive immune response is triggered, involving the production of B cells that specifically target antigens and secrete antibodies and the activation of cytotoxic T cells. These cells secrete immune mediators and effector molecules, such as cytokines and chemokines, to eliminate antigens, pathogens, infected and cancer cells.27 In the adaptive immune system, antigen-presenting cells (APCs) constantly eliminate exogenous or endogenous antigens, pathogens, infected and cancer cells. The exogenous antigens are primarily phagocytosed and processed by immature dendritic cells (DCs). During this process, the DCs mature and present target antigens to T cells via major histocompatibility complex (MHC) I or II molecules.28

Three key pathways for T cell activation include MHC I or II complexes binding T cell receptors, surface co-stimulatory molecule expression, and the secretion of cytokines, such as interleukin (IL)-12 and interferon (IFN)-γ. Different populations of T cells, such as helper T cells (CD4+), cytotoxic T cells (CD8+), memory T cells, and regulatory T cells, participate in the immune response. CD4+ T helper cells can differentiate into various subtypes, such as Th1 cells (secrete IFN-γ and tumor necrosis factor (TNF)-α), Th2 cells (secrete IL-4, IL-5, and IL-13), and Th17 cells (secrete IL-17), which help activate cytotoxic CD8+ cells and other innate immune cells to destroy tumor cells.29

Memory T cells form after antigen exposure and circulate through the body to provide long-term protection against foreign antigens. Moreover, antigen cross-presentation is an important aspect of the immune process, allowing extracellular antigens to be presented on MHC I molecules. This is a unique function of DCs that stimulates the immune system to eliminate target antigens such as tumor cells.30

Nanomedicine Platforms in Prostate Cancer Immunotherapy

Nanomedicine emerged as a cutting-edge platform for cancer treatment, offering targeted delivery of nanoparticles that induce superior antitumor responses while mitigating toxicity and associated costs. Various nanoparticles, such as liposomes,31 polymer-based nanoparticles (NPs),32 gold nanoparticles (AuNPs),33 magnetic nanoparticles (MNPs),34 silica particles (SNPs),35 quantum dots (QDs),36 carbon nanotubes (CNTs),37 and mixed particles, are utilized for prostate cancer drug delivery (Figure 1 by Figdraw). These nanoparticles employ passive38,39 or active targeting to enhance immunogenicity.40 As important nanomaterials with high porosity, versatility, and biocompatibility, the ordered porous structure of nanomaterials (nMOFs) can avoid self-quenching of photosensitized agents and promote the diffusion of reactive oxygen species (ROS), improving the effect of photodynamic therapy and eliciting a cytotoxic effect. Its mediated low-dose radiation therapy can be combined with anti-programmed death ligand 1 (PDL1) antibodies to extend the local therapeutic effect of radiation therapy to distant tumors. Additionally, nMOFs can be combined with other forms of immunotherapy (eg, STING agonists or CpG oligonucleotides) to generate systemic anti-tumor immunity.41 Liposomes,42 polymerized nanoparticles, and other drug delivery vehicles effectively encapsulate lipophilic anticancer drugs, creating a protective barrier between the organism and the drug. Upon degradation, these vehicles release the drug contents,43 exhibiting targeted delivery and safeguarding against drug degradation in experimental settings. This mechanism ensures drug accumulation within the tumor, enhancing efficacy.44 The unique properties of nanocarriers in terms of size, surface charge, and ability to functionalize with targeted ligands make them ideal for overcoming the biological barriers that typically limit drug efficacy to solid tumors. The classification of nanocarrier systems is presented in Table 1.

Table 1 Classification of Nanodrug Delivery Systems and Their Application in the Treatment of Prostate Cancer

Figure 1 Optimization strategies of nanoparticles for the treatment of prostate cancer.

Nanoparticle-mediated multimodal therapeutic strategies, such as chemodynamic therapy (CDT), sonodynamic therapy (SDT), photodynamic therapy (PDT), and radiation therapy (RT), provide a target for immune cell attack by inducing ROS production, directly impacting tumor cell survival and enhancing their immunogenicity. The application of these therapeutic tools enhances the direct clearance of tumor cells while helping establish long-term immune memory, improving the durability and effectiveness of treatment (Figure 2 by Figdraw).

Figure 2 Nanomedicines can be utilized to boost tumor immunogenicity in combination with CDT, SDT, PDT, and RT.

The potential of nanoparticles in modulating immune responses through multiple mechanisms in prostate cancer therapy is significant. This highlights the complex interactions of nanoparticles in regulating the TME and provides new insights regarding the mechanisms underlying tumor immune evasion to help guide the development of novel immunotherapies. This multimodal therapeutic approach may be important for overcoming the immunosuppressive microenvironment of prostate cancer and activating a potent antitumor immune response, improving efficacy. In addition, the modulation of the cytokine milieu and its effect on macrophage polarization reveals the possibility of enhancing tumor clearance by precisely modulating the immune response.

Macrophages, key immunoregulatory cells in the TME, differentiate into pro-inflammatory M1-type or anti-inflammatory M2-type depending on the signals received. Nanoparticle-based therapeutics play an important role in the antitumor immune response by modulating cytokine expression patterns within the TME, including upregulating IL-12 and TNF-α levels while reducing IL-10 and TNF-δ expression, influencing macrophage polarization. For example, cyclic RGD peptide-functionalized and manganese-doped eumelanin-like nanocomposites (RMnMels) can be used for high-temperature immunotherapy in PC3 prostate cancer to enhance anti-tumor immune responses by promoting the repolarization of M2 to M1 macrophages by clearing ROS and reshaping the immunosuppressive TME.57

Further investigation into the dynamic in vivo distribution of nanoparticles, their long-term immune effects, and potential systemic immunomodulatory impacts will facilitate the design of safer, more effective nanomedicines, paving the way for innovative approaches to prostate cancer treatment.

Nanomedical Enhancement in Cancer Immunotherapy

Targeted Delivery: Nanodrug platforms can identify and bind specific biomarkers overexpressed in prostate cancer cells, such as prostate-specific membrane antigen (PSMA), ensuring that the therapeutic agent is delivered directly to the tumor site. This can improve treatment specificity and efficacy.58,59

Controlled Release: During targeted delivery of the nanomedicine platform to the tumor site, nanocarriers can encapsulate the immunotherapeutic agent, facilitating its protected transport in the bloodstream and release in a controlled manner at the tumor site. This controlled release mechanism can be designed to respond to specific stimuli within the TME, such as pH changes or enzyme activity, ensuring the spatially and temporally controlled release of therapeutic agents.60

Enhanced Immunogenicity: Certain nanocarriers are designed to deliver drugs that induce immunogenic cell death (ICD) by promoting the release of tumor antigens and stimulating the immune system to recognize and attack danger signals, enhancing tumor cell immunogenicity.61

Recent Advancements

Recent studies have highlighted the effectiveness of nanomedicines in improving the efficacy of immunotherapy for prostate cancer. For example, clinical pilot studies have demonstrated the promise of gold-nanoshell-localized photothermal ablation for focal treatment of prostate tumors, highlighting the ability of nanomedicine to deliver highly targeted and effective treatment options with minimal side effects.62 In addition, the development of PSMA-targeted nanomedicines for treating advanced prostate cancer shows great potential for bridging the gap between nanomedicine research and clinical practice, providing new strategies for disease management.58

Another innovative approach involves using extracellular vesicles from Akkermansia muciniphila, which induce antitumor immunity against prostate cancer by modulating CD8+ T cells and macrophages, demonstrating the potential of nanomedicine to harness microbiota in cancer treatment.20 Additionally, urokinase plasminogen activator receptor (uPAR)-targeted nanocarriers based on exosomes have been explored. Specifically, the Exo-PMA/Fe-HSA@DOX system is loaded with doxorubicin and achieves synergistic therapy via chemodynamic treatment and low-dose chemotherapy in prostate cancer (PCa). This nanosystem can significantly enhance internalization in vitro and block the epidermal growth factor receptor (EGFR)/protein kinase B (AKT)/nuclear factor (NF)-κB signaling pathways.63

The integration of nanomedicine into prostate cancer immunotherapy is at the forefront of current research with the potential to significantly improve patient outcomes. Nanomedical platforms offer promising and feasible solutions to overcome the limitations of current therapeutic modalities by enhancing the targeted delivery, controlled release, and immunogenicity of therapeutic drugs. Ongoing research and clinical trials are essential to determine the full potential of these innovative therapies for treating prostate cancer.

Applying Nanotechnology to Overcome Immunotherapy Limitations

The immunological processes involved in cancer vaccine platforms, PD-1/PD-L1 inhibitors, and CAR-T cells are presented in Figure 3 (By Figdraw). Clinical studies related to immunotherapy are listed in Table 2. Although immunotherapy has achieved some positive results, its efficacy is often hampered by the immunosuppressive nature of the TME and immune escape. Recent advances in nanomedicine have provided innovative strategies to overcome these barriers and enhance the therapeutic potential of prostate cancer immunotherapy. Advanced nanotechnology enables precise targeting of TME components tailored to nanoparticle requirements. This ability to differentiate between healthy and malignant tissues facilitates TME modulation to impede tumor progression. Moreover, these nanoparticles boast prolonged retention, high bioavailability, and low toxicity, further enhancing their therapeutic potential.

Table 2 Immunotherapy Trials for Prostate Cancer

Figure 3 (A). Cancer vaccine platforms and interactions in the immune system; (B). PD-1/PD-L1 are involved in immune checkpoint blockade (ICB) therapy; (C).CAR-T cell immunotherapy process.

Role of Nanomedicine in TME Modulation

Nanomedicine plays a key role in modulating the TME to improve the immune response in prostate cancer. By targeting the unique properties of the TME, such as its acidic pH, hypoxic conditions, and high interstitial pressure, nanomaterials can enhance the penetration and retention of immunotherapeutics. This approach improves therapeutic efficacy and reduces systemic adverse reactions.

The immunosuppressive TME and immune escape mechanisms significantly limit the effectiveness of immunotherapy for prostate cancer. Nanomedical approaches have been developed to specifically target and modulate the TME, enhancing the immune response of tumor cells. For example, the two-pronged strategy using pH-driven membrane anchoring nanophotosensitizers has shown promise in stimulating ICD and isolating immune checkpoints, converting “cold” tumors into “hot” tumors, ultimately enhancing the efficacy of photoimmunotherapy for prostate cancer.64

Supramolecular nanotechnology materials have been developed to modify the immunosuppressive TME, working in tandem with immune checkpoint-blocking therapy to achieve enhanced cancer immunotherapy outcomes.65 After vaccination, DCs play a vital role in capturing, processing, and presenting tumor antigens, ultimately activating T cells that suppress tumor cells. Moreover, activated B cells contribute to tumor cell death through antibody-dependent cell-mediated cytotoxicity. Engineered metal-phenol networks have emerged as a novel strategy to regulate the TME, aiming to enhance cancer treatment by reversing its immunosuppressive nature and rendering tumors sensitive to immunotherapy.

Enhancing Delivery and Efficacy of Immunotherapeutic Agents

The delivery and efficacy of immune checkpoint inhibitors, cancer vaccines, and T-cell therapies are important components of immunotherapy. Nanoparticles can improve the efficacy of these drugs by delivering them directly to the tumor site, minimizing systemic toxicity and increasing their therapeutic efficacy. In mouse prostate tumors that develop in the context of prostate-specific PTEN and p53 deletion, the activation of bovine serum albumin (BSA)-nanoparticles by cabozantinib initiates their uptake by tumor-infiltrating neutrophils (TINs) rather than peripheral neutrophils, avoiding RES uptake in the liver and enabling more efficient intraterritormal payload delivery. Given that this platform minimizes off-target toxicity, it can also be used to test new combination therapies.66

PSMA-targeted nanoparticles have shown great potential in treating prostate cancer. PSMA is highly expressed in prostate cancer cells, providing a precise target for nanoparticles to maximize therapeutic efficacy and minimize side effects in healthy tissue. Meanwhile, a co-delivery system based on stem cell membrane-coated polydopamine nanoparticles significantly improves the targeting and efficacy of doxorubicin and PD-L1 siRNA.67 PSMA-targeted melanin-like nanoparticles combine photothermal therapy and drug delivery functions, achieving up to a 90% apoptosis rate in prostate cancer cells in vitro.68 Glutamate-urea-based PSMA-targeted poly(lactic-co-glycolic) acid (PLGA) nanoparticles deliver docetaxel, effectively doubling the anticancer efficacy of the drug.69 Additionally, PSMA-targeted nanoparticles functionalized with a urea-based inhibitor demonstrate good biocompatibility and high targeting efficiency, reducing tumor viability.70

In radioparticle therapy, PSMA-targeted alpha therapy (TAT) using 225Ac-PSMA-I&T has shown promising antitumor effects in patients with advanced metastatic CRPC (mCRPC). In one study, 11/14 patients experienced a significant decrease in PSA levels, with a ≥ 50% reduction in seven patients, supporting the therapeutic efficacy.71 Hence, PSMA-targeted nanoparticles hold great promise for prostate cancer diagnosis and treatment. However, many technical and clinical challenges must be overcome to maximize their clinical application.72

In addition, approaches such as mRNA- and hydrogel-based CAR-T-cell delivery, photothermal remodeling, and TME-based CAR-T-cell therapy have shown promise in enhancing the efficacy of CAR-T therapy. The integration of nanotechnology into immunotherapy holds the potential to overcome certain limitations of traditional immunotherapy, leading to improved therapeutic outcomes.73

Clinical Implications and Future Prospects

The combination of nanomedicine and immunotherapy for prostate cancer has great potential to improve patient prognosis. Clinical trials and preclinical studies have shown the promise of nanotechnology-based strategies in overcoming the limitations of current immunotherapy approaches. Future research should focus on the development of multifunctional nanosystems that can simultaneously target multiple aspects of the TME and the immune system, providing a comprehensive and novel strategy for combating prostate cancer with excellent clinical performance.

Synergistic Approach and Combination Therapy

In recent decades, single-mode treatment strategies, including chemotherapy, photodynamic therapy, and radiation therapy, have made significant medical advances in tumor suppression and patient survival.74 However, these treatments have encountered many limitations in clinical application. Specifically, the rapid metabolic clearance and non-specific distribution of chemotherapeutic drugs significantly reduce their therapeutic efficiency and may trigger systemic toxicity reactions. Moreover, prolonged or repeated use of a drug can result in tumor cells developing therapeutic resistance.75 Meanwhile, the effectiveness of photodynamic therapy in treating tumors is limited by the potential for irreversible light damage to normal tissues, the inherent heat tolerance of tumors, and the risk of tumor metastasis and recurrence. Moreover, hypoxic tumor cells are less sensitive to ionizing radiation. Monoimmunotherapy also faces off-target toxicity, inadequate immune responses, poorly sustained efficacy, and low immunogenicity, resulting in inadequate treatment outcomes.

However, combining immunotherapy with other treatment modalities, including chemotherapy, photodynamic therapy, radiation therapy, sonodynamic therapy, metabolic therapy, and microwave thermotherapy, has facilitated the development of synergistic treatment strategies. These modalities can achieve a super-additive treatment effect beyond single therapy or simple combination therapy, improving the overall efficacy of cancer treatment.

Recent advances have demonstrated the potential of nanomedicine to work synergistically with existing therapies such as chemotherapy, radiotherapy, and hormone therapy. Thus, combining nanomedicine and immunotherapy to treat prostate cancer is a promising frontier for improving treatment efficacy and overcoming the limitations of conventional treatments.

Combining Nanomedicine with Other Therapies

The rationale for combining nanomedicine with other therapies to treat prostate cancer lies in the multifaceted nature of the disease and its complex TME. Nanomedicine brings about targeted delivery mechanisms, improved bioavailability, and reduced systemic toxicity, which complement the mechanisms of action of traditional therapies. For example, polymerized nanomaterials designed for tumor-targeted combination therapy have demonstrated synergistic genotoxicity in prostate cancer through the combined delivery of chemotherapeutic agents, such as doxorubicin and 5-fluorouracil, enhancing cell cycle arrest, caspase-3 activation, and DNA damage.76

Chemotherapy

Conventional chemotherapy remains a cornerstone in prostate cancer treatment, relying on toxic drugs to eliminate cancer cells.77 Widely used in clinical practice, traditional chemotherapeutic drugs suffer from a short plasma half-life and rapid distribution in healthy tissues and organs, leading to significant side effects. In addition, the limited retention and accumulation of drugs within tumors, coupled with the multidrug resistance (MDR) phenomena induced by chemotherapy, contribute to cancer cells developing resistance against structurally similar drug molecules, increasing the likelihood of treatment failure.78 Therefore, more effective methods must be explored and developed to obtain greater clinical benefits. With the development of nanomedicine, the combination of multifunctional nanocarriers and radiotherapy has shown promising prospects. Strategies to improve the efficacy of nanomaterial-based chemotherapy include chemotherapy targeting specific suborganelles, chemotherapeutic drug enhancement at tumor sites, reversal of drug resistance mechanisms, and combination chemotherapy. Nanocarriers encapsulate chemotherapeutic drugs, shielding them from efflux pumps to increase intracellular drug concentrations.79 By targeting tumor cells or blood vessels, these nanocarriers outperform traditional chemotherapy drugs with lower toxicity, mitigating multidrug resistance and enhancing drug efficacy while reducing side effects. Substances like borletoxins, doxorubicin, and actinomycin are utilized in preparing nanomedical drugs.80 Moreover, some nanocarrier chemotherapeutic drugs, such as paclitaxel- and doxorubicin-containing liposomes, have been applied clinically with relatively mild adverse reactions and high safety.

Radiotherapy

Radiotherapy is a main treatment modality for prostate cancer that is used as a first-line treatment or in combination with surgery and chemotherapy.81 Although effective, it has certain side effects as it cannot distinguish between normal and diseased tissues. Accordingly, many studies have focused on determining the effective dose to kill tumor cells without causing additional damage to healthy tissues. Nanomedicine can also be applied to new drug delivery methods in CRT, and the low toxicity of nanomedicine carriers to normal tissues has been demonstrated experimentally.82 Numerous studies have explored the benefits of nanomedicine delivery in improving chemoradiotherapy (CRT) and enhancing delivery to induce DNA damage directly near tumors to minimize drug off-target effects and the required radiation doses. Nanomaterials labeled with magnetic resonance imaging (MRI), positron emission tomography (PET), and computed tomography (CT) contrast agents can enhance imaging capabilities. Moreover, nanotechnology enables the preparation of nanoparticles to alleviate tumor hypoxia, regulate the immunosuppressive TME, and significantly enhance radiotherapy efficacy.83,84

Combination Therapy

Nanomedicine also plays an important role in combination therapy, with growing evidence supporting its ability to enhance the synergy between immunotherapy and radiotherapy. While most patients undergoing radiotherapy receive chemotherapy concurrently, ie, CRT, this combined approach often fails to eradicate the primary tumor, necessitating improved radiotherapy-based strategies. Emerging immunotherapies can clear tumors by activating the patient’s immune system. In particular, CTLA-4, PD-1, and other immune checkpoints have been shown to improve clinical symptoms85 while combinatorial radiotherapy and immunotherapy further enhance efficacy.86 In preclinical models of metastatic prostate cancer, combining irradiation of metastatic cancer cells and anti-CTLA-4 antibody treatment effectively induced T-cell responses, enhancing local antitumor effects and responses to distant metastases. Thus, this combinatorial therapy has significant systemic immunological effects.87 In the CRPC mouse model, the survival rate was improved by combined radiotherapy and anti-PD-1 or anti-PD-L1 treatment compared with monotherapy.88 Radiotherapy can enhance immunotherapy in clinical practice,89 as evidenced by MHC I up-regulation,90 increased antigen availability,91 and heightened cytokine release. Meanwhile, immune checkpoint inhibitors can enhance CRT.92 Mechanisms underlying this synergy include sensitization of the TME prior to radiotherapy93 or elicitation of an immune response sensitized to radiation.94 Nanoparticles are crucial in this context as they facilitate targeted drug delivery and enhance antigen presentation by APCs.95 In addition to enhancing the immunotherapeutic effect to improve treatments,96 nanoparticles can also transport photosensitizers as complex delivery carriers of antibodies or radioisotopes for direct radiation delivery,96 which can be activated by photothermal or photodynamic therapy.

The Au/Mn nanoparticle-Luteinizing Hormone-Releasing Hormone (AMNDs-LHRH) nanosystem is a sophisticated targeted therapeutic platform designed for multimodal imaging-guided photothermal therapy of prostate cancer. It boasts excellent targeting capabilities and efficient photothermal conversion, significantly enhancing the therapeutic effects of photothermal treatment for metastatic prostate cancer. Specifically, this system targets gonadotropin-releasing hormone receptor (GnRH-R)-positive prostate cancer cells and their metastases, facilitating accurate preoperative diagnosis via CT/MR imaging. Additionally, the system features fluorescence visualization for surgical navigation, minimal invasiveness, lack of drug resistance or side effects, and the potential to improve patients’ quality of life. Consequently, the Au/Mn nanoparticle-LHRH nanosystem holds significant potential for clinical diagnosis and treatment of metastatic prostate cancer.97 Alternatively, up-conversion can mark and stimulate DCs.98 In addition, nanotechnology may synergistically enhance these two therapies by improving the responses of NK and B cells. In conclusion, nanotechnology exhibits promising synergy in combining immunotherapy and radiotherapy.

Multifunctional nanosystems not only improve drug bioavailability, enhance tumor-targeting capabilities, and reduce drug side effects but also offer a potential platform for treating and diagnosing metastatic prostate cancer. While prostate cancer and its metastases are typically detected using MRI, challenges arise due to discrepancies in detection methods, such as PET-CT and ECT.99 Conventional contrast media are limited to single-mode imaging, whereas multifunctional nanosystems improve the accuracy of multimode imaging detection of metastatic prostate cancer, effectively guiding cancer treatment. Furthermore, multifunctional nanosystems demonstrate potent photothermal treatment effects, inducing tumor cell apoptosis by elevating the temperature of tumor cells through near-infrared light absorption and heat conversion, facilitating thermal ablation of tumor cells100 with minimal invasiveness, no drug resistance, few side effects, and low toxicity.101 Hence, multifunctional nanosystems present new possibilities for the diagnosis and treatment of prostate cancer, offering powerful targeting capabilities.

Case Studies and Clinical Trials Highlighting Synergistic Effects

Several case studies and clinical trials have demonstrated the synergistic potential of nanomedicines in combination with other cancer treatments. For example, nanodrug-driven PDT has been shown to trigger ICD and regulate the TME, improving the efficacy of cancer immunotherapy.102 In addition, ongoing studies on vaccine therapy, CTLA-4 inhibitors, PD-1/PD-L1 inhibitors, and PSMA-targeted therapies have reported promising results as prostate cancer immunotherapy modalities and the indispensable role of nanomedicine in facilitating these approaches.8 Recent studies have reported the utilization of combined immunotherapy and nanomedicine in clinical trials for cancer treatment (Table 3).

Table 3 Immunotherapy Combined with Nanomedicine in Cancer

Future Prospects for Designing Multifunctional Nanosystems for Immunotherapy

Looking to the future, the design of multifunctional nanosystems has the potential to advance the development of prostate cancer immunotherapy. The development of vaccine-based immunotherapy regimens, such as PF-06753512, which targets prostate-specific antigens and uses immune checkpoint inhibitors, exemplifies the innovative approaches being explored. PF-06753512 is a vaccine-based immunotherapy regimen (VBIR) under development for treating patients with prostate cancer across various clinical stages. This regimen combines a vaccine approach with immune checkpoint inhibitors, utilizing novel administration methods, including electroporation of plasmid DNA (pDNA) encoding antigens and subcutaneous (SC) delivery of immune checkpoint inhibitors. In a Phase I open-label study, this strategy exhibited safety signals comparable to other immune checkpoint inhibitor combination trials in mCRPC, stimulating antigen-specific immunity across all cohorts and demonstrating modest antitumor activity in patients with biochemical recurrence (BCR) without the use of androgen deprivation therapy (ADT).121 These strategies are designed to enhance antigen specificity, modulate immune responses, and achieve more effective and long-lasting antitumor effects.121

The synergy and fusion of nanomedicine and other therapeutic methods provide a promising avenue for improving the efficacy of immunotherapy for prostate cancer. By harnessing nanomedicine’s unique properties, researchers and clinicians can more effectively target the TME, overcome drug resistance, and minimize adverse reactions. Continued exploration and clinical validation of these combination therapies will ensure that they reach their full potential in prostate cancer treatment.

Clinical Transformation and Challenges

Overview of Nanomedicine Products in Clinical Use or Trials

Products such as PSMA-targeted nanomedicines, nanoparticle-based siRNA, and chemotherapeutic drug delivery systems have shown potential in preclinical and early-stage clinical trials. These nanomedicines aim to improve targeted drug delivery through innovative strategies, such as photothermal therapy and targeted delivery of immunomodulators, to reduce adverse reactions and improve therapeutic efficacy.122–125

Nanoparticles have shown considerable promise in the diagnosis and treatment of prostate cancer. In diagnosis, nanoparticles can be used for biomarker detection, encompassing quantitative fluorescence nanosensors,126 superparamagnetic iron oxide nanoparticles modified by chitosan, and sarcosine oxidase gold nanoparticles.127 Additionally, nanoparticles have been applied in nuclear medicine, including in the assembly of aptamers from fluorophores128 and utilizing Raman optical inspection platforms based on nanocolumns.129 They may also contribute to imaging through gene amplification of nanoparticle tumor homing strategies.130

Nanomedicine products have also undergone a series of clinical transformations. In chemotherapy, nanoparticles can be used to deliver docetaxel,131,132 cabatasel,133 carbataxel combined with hyaluronic acid,134 and other drugs. In radiotherapy, nanoparticles can facilitate X-ray source radiation by significantly increasing the radiosensitivity of cancer cells135 or inhibiting the cloning potential of hypoxic prostate cancer cells.136 They also have applications in image-guided surgery, PSMA receptor-targeting quantum dots,137 and double infrared-near-infrared spectroscopy fluorescent and radio-guided probes.138 In genetic and epigenetic therapies, nanotechnology facilitates the delivery of gold nanoparticles,139 micelles,140 and microRNA-197 inhibitors.141 In pH-based strategies, nanotechnology can be used to coat calcium dioxide with polyacrylic acid142 and cathepsin,143 among other agents. Nanomedicines can also be used with natural compounds, such as doxorubicin, tanshinone,144 and goniothalamin.145 Additionally, nanotechnology can be applied for bionic phosphatidylserine,146 lentinan,147 graphene oxide–peptide complexes,148 and various nanomaterials to treat prostate cancer, including overcoming drug resistance of CRPC cells,149 inhibiting tumor growth by targeting abnormal expression of protein kinase N3 (PKN3) expression, and using hyperthermia to kill prostate cancer cells.150 Herein, we focus on the application of nanoparticles in immunotherapy.

Studies have been conducted on the clinical transformation of nanomedicine products in prostate cancer immunotherapy. Guo et al used PSMA aptamer (Apt)-functionalized putamen nanoparticles in paclitaxel (PTX)-resistant LNCaP (LNCaP/PTX) cells. The nanoparticles were found to inhibit epithelial–mesenchymal transition and re-sensitize cancer cells to PTX.151 Mangiferin-functional gold nanoparticles (MGF-AuNPs) designed by Khoobchandani et al increase the expression of antitumor cytokines IL-12 and TNF-⍺ by regulating the balance between pro-tumor M2 and antitumor M1 macrophages in mice with prostate cancer. It also reduces the expression of the tumor-promoting cytokines IL-10 and IL-6.139 Meanwhile, Cole et al used cationic RALA/pDNA NPs combined with dissolvable microneedle patches to create a two-layer delivery system. This system effectively delivers a prostate cancer DNA vaccine to the dermal and epidermal APCs, enhancing the antitumor immune response and delaying tumor growth in mice with prostate cancer, ultimately prolonging survival.152 Similarly, Islam et al activated the CD8+ T-cell-mediated antitumor response using nanoparticles containing antigen-coding mRNA and TLR7/8 agonists.153

Translational Barriers

Although nanomedicine has considerable potential in prostate cancer immunotherapy, several translational barriers have hindered its clinical application.

  1. Safety concerns: Nanomedicines have unique properties that, although beneficial, pose potential safety concerns. Understanding the long-term effects and toxicity of nanoparticles remains a major challenge.154
  2. Scalability: Producing nanomedicines on a scale suitable for widespread clinical use is complex and expensive. Therefore, it is important to ensure consistency in quality and efficacy across batches.155
  3. Regulatory approval: The novelty of nanomedicines complicates the regulatory approval process. Demonstrating their safety and efficacy, as well as establishing their superiority over existing therapies, are critical factors for regulatory approval.9
  4. Patient stratification: It is important to determine which patients will benefit the most from nanomedicine-based treatments. This requires the selection of patients using biomarkers and monitoring their treatment response.156

Strategies to Overcome Translational Barriers

Several strategies have been proposed to address translational barriers and promote the clinical application of nanomedicine in prostate cancer immunotherapy.

  1. Increased safety through design: The development of nanoparticles with biocompatible and biodegradable materials can minimize toxicity and improve safety. A targeting component can also be added to reduce off-target effects.157
  2. Improve scalability using advanced manufacturing technology: Advances in manufacturing technology using nanotechnology can improve scalability and reduce costs. Continuous manufacturing processes and automation can provide this solution.158
  3. Navigating the regulatory pathway: Engaging with regulators early in the development process can address regulatory challenges. Establishing clear guidelines for evaluating nanomedicines could streamline this process.62
  4. Advancing patient stratification: Investing in research to identify and validate patient-selected biomarkers is critical. Combining nanomedicine with precision medicine can help improve patient outcomes.159

Nanomedicine presents a groundbreaking opportunity for prostate cancer immunotherapy by offering targeted, effective, and less toxic treatment options. Overcoming translational barriers through innovative design, manufacturing application, regulatory strategies, and precision medicine approaches will be pivotal to realizing nanomedicine’s full potential in clinical settings.

Future Directions and Innovations

The advancement of nanomedicine in the field of prostate cancer immunotherapy has been marked by the development of intelligent nanocarriers and stimulus-response systems. These innovative platforms are designed to improve the delivery and efficacy of immunotherapeutics by targeting the TME with high precision. Smart nanocarriers can be designed to identify specific tumor markers to ensure that therapeutic agents are delivered directly to cancer cells, minimizing systemic toxicity and improving patient outcomes.58,59 The stimulus-response system further complements this approach by releasing therapeutic payloads in response to specific physiological triggers within the TME, such as pH changes or enzyme activity, providing a controllable, targeted therapy strategy.60

Smart Nanocarriers and Stimulus-Responsive Systems

Despite significant advancements in nanoscale drug delivery technology, the drug delivery efficacy of most conventional carriers remains limited by their single release curve, which remains unchanged over time and fails to adapt to the specific needs of patients or physiological environments.160,161 Moreover, the effectiveness of nanoscale drug carriers is often compromised due to immune system defenses.162 Therefore, the development of intelligent and controllable nanocarriers is imperative. These nanocarriers should not only exhibit flexible drug release in diverse environments but also deliver immunomodulators locally and effectively while minimizing side effects to enhance therapeutic outcomes. In addition, nanomaterials have diverse structures and can easily undergo functional modifications, providing opportunities for developing intelligent stimulus-responsive nanodrug delivery systems.163 These smart nanocarriers flexibly release drugs in different environments and locally and effectively release immunomodulators while reducing side effects, improving therapeutic effectiveness.164,165

Future directions and innovations in prostate cancer immunotherapy should focus on the development of smart nanomedicine delivery systems that respond to specific physiological and pathological stimuli. Stimulus-responsive nanomaterials have been engineered to construct intelligent drug delivery systems that recognize distinct features of the TME, such as acidic pH, peroxide levels, and the presence of specific enzymes, enabling more precise drug delivery. In this way, the local concentration of immunotherapy drugs will be enhanced without corresponding increases in adverse effects on normal tissues. Moreover, the therapeutic effect by will be enhanced by modulating the immune response, providing a more personalized and effective treatment plan for patients with prostate cancer.

The responsiveness of intelligent nanomaterials to stimuli can be categorized into endogenous and exogenous stimuli, with the stimulus-response system further classified into single-stimulus-response and multi-stimulus-response nanosystems. Stimulus-responsive nanosystems can accurately release drugs upon exposure to specific stimuli, exhibiting exceptional specificity in response to various stimuli to modulate the immune system by releasing immunomodulators, thus improving cancer treatment.

Smart Nanocarrier Mechanisms

Smart nanosystems can regulate the immune system to improve cancer treatment through the following mechanisms:

  1. Endogenous stimulus-response: Smart nanosystems can recognize specific markers in the TME, such as low pH and highly expressed enzymes or peroxide levels. For example, pH-sensitive nanocarriers dissociate in an acidic TME, releasing immune activators that directly activate the surrounding immune cells to enhance tumor attack.
  2. Exogenous stimulus-response: Using external stimuli, such as light, magnetic fields, or ultrasound, smart nanosystems can release immunomodulators at specific times and locations. For example, light-sensitive nanocarriers offer a targeted approach to delivering immune enhancers directly to tumors through irradiation with near-infrared light, minimizing systemic side effects and enhancing treatment specificity.
  3. Multi-stimulus-response systems: By integrating endogenous and exogenous stimulus responses, multi-stimulus-response nanosystems enable precise drug delivery under more complex regulatory conditions. Such systems can simultaneously respond to changes in the TME and external stimuli, facilitating the fine control of immunomodulator release to more effectively modulate the immune system and improve cancer treatment outcomes. Through these mechanisms, smart nanomaterials can improve the efficiency and specificity of drug delivery while also effectively activating or suppressing immune responses by precisely controlling the release of immunomodulators. This innovative treatment is expected to significantly improve treatment outcomes for cancers, including prostate cancer, providing patients with more personalized and effective treatment options.

These stimulus-response systems can specifically react to various stimuli, including the redox environment,166 pH,167 heat,168 light stimulation,169 magnetic fields,170 enzymes,171 ultrasonic stimulation,170 etc., and can release immunomodulators at specific sites. However, stimulus-response systems with synergistic effects between multiple stimuli are more likely to deliver and release immunomodulators effectively.172,173 Immunomodulators encompass various substances capable of modulating immune responses, including cytokines, chemical factors, small-molecule drugs, and specific proteins. In prostate cancer immunotherapy, the precise release of immunomodulators is essential for activating or inhibiting specific immune pathways to enhance the therapeutic effect or reduce adverse reactions. For example, pH-sensitive nanocarriers can release immune activators such as cancer vaccines or long-acting cytokines in the acidic TME, directly activating T cells and natural killer cells to generate a strong immune response against tumor cells. Simultaneously, nanosystems that respond to heat or light stimulation can be used to remotely control the release of anti-inflammatory cytokines or immunosuppressants to mitigate immune-related side effects that may occur during treatment. Furthermore, multi-stimulus-response nanosystems can detect multiple environmental signals, such as simultaneous responses to the properties of drugs and pH changes in the TME, enabling more precise and selective immunomodulator release strategies. This system ensures that immunomodulators are released when and where they are most needed, maximizing the effectiveness of the treatment and minimizing the impact on normal tissues. Therefore, the development of intelligent nanocarriers capable of responding to single or multiple physiological and pathological stimuli for the precise delivery and release of immunomodulators provides new strategies and directions for the design of immunotherapeutics for complex diseases such as prostate cancer.

Various novel multi-stimulus-response nanosystems have been designed. Researchers have designed smart size/shape convertible nanomedicines that can respond to near-infrared laser irradiation and an acidic TME, effectively ablating tumors and inhibiting metastasis. Nanomaterials can inhibit the mobility of tumor cells and significantly prolong the residence time of MEL/Cypate@HA in tumor tissues, achieving effective tumor clearance.174 Furthermore, an HA-functionalized nanoparticle platform based on molybdenum disulfide responds to near-infrared laser irradiation and a reoxidation environment, achieving targeted delivery of CPT. This platform not only prevents random leakage of encapsulated CPT into the bloodstream but also accelerates drug release in tumor-associated environments rich in glutathione (GSH). MoS2-SS-HA-CPT effectively inhibits the proliferation of lung cancer cells and tumor growth under near-infrared irradiation.175 Meanwhile, tunable nanocapsules have been developed that are able to respond to near-infrared laser irradiation and the TME. These nanocapsules are coated with Fe/FeO core-shell nanocrystals in a PLGA-polymer matrix and co-loaded with chemotherapeutic drugs and photothermic agents. These cleverly designed nanocapsules not only shrink and decompose into small-sized nanoscale drugs upon drug release but also regulate the TME to produce excess ROS, enhancing the synergistic treatment of tumors.176 Other studies have used near-infrared dyes (eg, IR820) as carriers to induce the supramolecular assembly of chemical drugs (eg, Docetaxel, DTX) to form nanoparticles with enhanced drug-coating properties. These nanoparticles exhibit a dual response to MMP and GSH. NP-coated NIR dyes can be used as photothermal conversion agents for effective photothermal therapy. In addition, a CF27 peptide containing 12 D-amino acid units was designed as a PD-L1 agonist to block the immune checkpoint PD-1/PD-L1 interaction.177 This peptide was encapsulated within cancer cell membrane mesoporous organosilica nanoparticles (MONs) that possess X-ray and room-responsive diselenide bonds. Additionally, these MONs were loaded with doxorubicin within pinhole diselenide-bridged structures and coated with membrane segments from cancer cells to facilitate tumor targeting and evasion of the immune system.178 Additionally, pH/ROS cascade prodrug micelles have been developed with size-shrinking and charge-reversal properties. These micelles can deliver siTGF-β, leading to synergistic TME remodeling.179 Similarly, novel nanomaterials with dual pH and redox responsiveness have been designed to enhance therapeutic efficacy in prostate cancer treatment.

Through their unique immunomodulatory mechanisms, nanomaterials can activate or regulate the immune system’s response to tumors, enhancing the immunotherapeutic effect. For example, DNA-based nanomaterials are widely used in innovative and effective cancer immunotherapy, including for the delivery of ICD inducers, adjuvants, vaccines, and immune checkpoint blockers, as well as applications in immune cell engineering and adoptive cell therapy. These nanoplatforms can precisely deliver immunomodulators capable of triggering specific responses in immune cells, such as enhancing T-cell activity or modulating the immunosuppressive environment. In the field of prostate cancer treatment, pegylated manganese-zinc ferrite nanocrystals combined with tumor-implanted micromagnets enable synergistic prostate cancer therapy through the activation of iron death and ICD, a strategy that further amplifies the effects of ICD by stimulating the cGAS–STING pathway. Therefore, combining the immunomodulatory mechanisms of these nanomaterials with specific applications in prostate cancer treatment has the potential to improve treatment outcomes while also providing patients with more personalized and effective treatment options. Future research should focus on exploring the specific mechanisms of these nanomaterials in prostate cancer immunotherapy and on how to achieve personalized cancer immunotherapy by designing smarter nanomedicines.

New Strategies for Prostate Cancer Immunotherapy

ZIF-8@DOX @organosilica nanoparticles (ZDOS NPs) demonstrate significant potential for exploring new strategies in prostate cancer immunotherapy. These nanocores, with nanoscale ZIF-8 cores and silicone shells containing disulfide bonds, provide a good structure and up to 41.2% doxorubicin loading capacity also helping to activate immune responses.180

ZIF-8 nanoparticles can induce endogenous pyroptosis via a caspase-1/gasdermin D (GSDMD)-dependent pathway, accompanied by necrosis and ICD, providing a new pathway for effective in situ immune initiation. Moreover, delivering PD-1 inhibitors via ZIF-8 nanoparticles demonstrates a precise and targeted approach to cancer therapy, reducing non-targeted effects and enhancing therapeutic efficacy. This combination strategy addresses the existing challenges and limitations of current immunotherapy technologies, with the ultimate goal of improving patient outcomes in cancer treatment. Notably, in the context of prostate cancer treatment, the application of ZDOS NPs extends beyond simply increasing drug loading. More importantly, these nanoparticles hold the potential to regulate the TME, facilitate immune cell penetration, and stimulate the immune response. For example, by altering the pore size and surface properties of ZIF-8 nanoparticles, controlled drug release can be achieved to activate an immune response against prostate cancer cells while reducing damage to healthy tissues. Therefore, the application of ZDOS NPs in prostate cancer immunotherapy not only reflects their function as drug carriers but also their unique value in activating and regulating the immune system.

Personalized Nanomedicine: A Vision for Prostate Cancer Immunotherapy

The ultimate goal of advancements in nanomedicine is to establish a personalized approach for prostate cancer immunotherapy that tailors treatments to the genetic makeup, tumor characteristics, and immune profile of an individual patient. This approach fully exploits the potential of nanotechnology, artificial intelligence, and machine learning to design highly specific nanocarriers that can effectively target and regulate the TME, enhance immune responses against cancer cells, and overcome resistance mechanisms. The promise of personalized nanomedicine lies in its ability to provide more effective, less toxic, and highly specific treatment regimens, thereby changing the landscape of prostate cancer immunotherapy.181–183

Recently, a concept known as personalized nanomedicine has emerged, combining precision medicine and nanomedicine. This approach involves integrating a patient’s omics information, including their genome, metabolome, and proteomics data, with nanotechnology.184 This integration enables the development of more precise treatment plans tailored to individual patients. Compared with a one-size-fits-all approach, a precision medicine approach may help more accurately predict which treatment and prevention strategies will be effective for a particular patient population. Precision medicine refers to the medical diagnosis, treatment, and products tailored to individual patients.184,185 However, designing personalized nanomedicine presents challenges due to the overexpression of specific metabolic enzymes in patients, which can prematurely degrade the matrix and targeted ligands. Additionally, many targeted ligands do not correspond to the most overexpressed cell surface receptors in cancer.

Recent research demonstrates that combining nanotechnology platforms with cancer immunotherapy strategies enables precise regulation of prostate cancer treatment. For example, the use of ZIF-8 nanoparticles as carriers of PD-1 inhibitors improves the efficiency of targeted drug delivery and enhances the effect of immunotherapy by regulating the TME. In addition, nanotechnology can activate the immune system to attack tumors by regulating the immunosuppressive state in the TME, improving therapeutic effects.

Artificial Intelligence (AI) and Machine Learning in Nanomedicine Design

The integration of AI and machine learning (ML) in the design and optimization of nanomedicines for cancer immunotherapy is a frontier research area (Figure 4 by Figdraw). These technologies offer an unprecedented ability to analyze complex biological data, predict nanocarrier behavior, and optimize treatment outcomes. AI and ML algorithms have been successfully used to identify new biomarkers, design personalized nanomedicine formulations, predict patient responses to immunotherapy, and create more effective and personalized treatment plans.186,187

Figure 4 Using databases and AI to design nanomaterials for immune response and immunotherapy.

Through AI, it is possible to optimize the drug and dose parameters of nanomedicine drugs. Indeed, AI has been utilized in diagnostics, therapeutics, personalized drug targeting and dosing, and the assessment of nanotoxicity in nanomaterials (Table 4).

Table 4 Application of AI/MI in Nanomedicine

This involves determining the appropriate degree of drug exposure required to achieve the optimal therapeutic effect, reducing time and dose dependence. Additionally, AI can facilitate personalized treatment by adapting to the specific needs of each patient.209 The application of AI is not limited to the optimal combination of a single drug but also includes real-time adjustments during treatment to respond to changes in the TME and dynamic changes in the patient’s immune status.

The application of AI in nanomedicine facilitates drug dose optimization and combination therapy strategies by identifying powerful drug combinations within a vast parameter space. Clinical treatment studies across various cancers have demonstrated the effectiveness of this approach. Patients exhibit considerable variability in the required drug doses to achieve synergistic effects and optimal treatment outcomes. Moreover, these parameters can fluctuate over time within the same patient. The emergence of AI is poised to play a pivotal role in reconciling these complexities, enabling the development of more effective and safer treatments in nanomedicine.210,211

Conclusion

Nanomedicine represents a pivotal advancement for improving prostate cancer immunotherapy. It encompasses diverse delivery platforms, each offering unique advantages in this context. Nanotechnology is expected to overcome many limitations of immunotherapy, such as immune escape and the challenges of TME regulation. Nanotechnology can also enhance the delivery efficiency and therapeutic effectiveness of checkpoint inhibitors, cancer vaccines, and T-cell therapies to overcome various limitations of immunotherapy. Integrating nanomedicine with traditional treatments like chemotherapy and radiotherapy has shown promise in achieving superior outcomes compared to standalone therapies, fostering synergistic effects in combination therapy. This multifaceted approach improves drug bioavailability, enhances tumor targeting, and minimizes side effects, thereby advancing prostate cancer diagnosis and treatment. While clinical translation may face hurdles such as safety, scalability, regulatory approval, and patient stratification of nanomaterials, ongoing research and the integration of advanced technologies such as AI and ML in both research and clinical settings offer promising avenues for addressing these challenges. We advocate for continued innovation and exploration in this dynamic field to unlock the full potential of nanomedicine in prostate cancer immunotherapy.

Funding

This work was supported by the WU JIEPING MEDICAL FOUNDATION (No. 320.6750.2023-11-34).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA: A Cancer j Clin. 2023;73(1):17–48. doi:10.3322/caac.21763

2. Chowdhury S, Bjartell A, Lumen N, et al. Real-World Outcomes in First-Line Treatment of Metastatic Castration-Resistant Prostate Cancer: the Prostate Cancer Registry. Targ Oncol. 2020;15(3):301–315. doi:10.1007/s11523-020-00720-2

3. Gillessen S, Attard G, Beer TM, et al. Management of Patients with Advanced Prostate Cancer: report of the Advanced Prostate Cancer Consensus Conference 2019. Europ urol. 2020;77(4):508–547. doi:10.1016/j.eururo.2020.01.012

4. Nuhn P, De Bono JS, Fizazi K, et al. Update on Systemic Prostate Cancer Therapies: management of Metastatic Castration-resistant Prostate Cancer in the Era of Precision Oncology. Europ urol. 2019;75(1):88–99. doi:10.1016/j.eururo.2018.03.028

5. Cancer Immunotherapy.

6. Zhang Y, Zhang Z. The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell Mol Immunol. 2020;17(8):807–821. doi:10.1038/s41423-020-0488-6

7. Saxena M, Der Burg SH V, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21(6):360–378. doi:10.1038/s41568-021-00346-0

8. Mitsogiannis I, Tzelves L, Dellis A, Issa H, Papatsoris A, Moussa M. Prostate cancer immunotherapy. Expert opin biol ther. 2022;22(5):577–590. doi:10.1080/14712598.2022.2027904

9. Irvine DJ, Dane EL. Enhancing cancer immunotherapy with nanomedicine. Nat Rev Immunol. 2020;20(5):321–334. doi:10.1038/s41577-019-0269-6

10. Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. J Med. 2018;378(2):158–168. doi:10.1056/NEJMra1703481

11. Pfirschke C, Engblom C, Rickelt S, et al. Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy. Immunity. 2016;44(2):343–354. doi:10.1016/j.immuni.2015.11.024

12. Thakor AS, Gambhir SS. Nanooncology: the future of cancer diagnosis and therapy. CA a Cancer J Clinicians. 2013;63(6):395–418. doi:10.3322/caac.21199

13. Dai W, Lu S, Zeng W, Lee D. Lipid Coated and Chlorin e6 Loaded Calcium Carbonate for Effective In Situ Immunotheraphy of Colorectal Cancer. j biomed nanotechnol. 2020;16(8):1196–1204. doi:10.1166/jbn.2020.2965

14. Parvani JG, Gujrati MD, Mack MA, Schiemann WP, Lu ZR. Silencing β3 Integrin by Targeted ECO/siRNA Nanoparticles Inhibits EMT and Metastasis of Triple-Negative Breast Cancer. Cancer Res. 2015;75(11):2316–2325. doi:10.1158/0008-5472.CAN-14-3485

15. Meng L, Teng Z, Yang S, et al. Biomimetic nanoparticles for DC vaccination: a versatile approach to boost cancer immunotherapy. Nanoscale. 2023;15(14):6432–6455. doi:10.1039/D2NR07071E

16. Buschmann MD, Carrasco MJ, Alishetty S, Paige M, Alameh MG, Weissman D. Nanomaterial Delivery Systems for mRNA Vaccines. Vaccines. 2021;9(1):65. doi:10.3390/vaccines9010065

17. Wang X, Fu J, Jiang C, et al. Specific and Long‐Term Luminescent Monitoring of Hydrogen Peroxide in Tumor Metastasis. Adv Mater. 2023;35(20):2210948. doi:10.1002/adma.202210948

18. Adekiya TA, Kondiah PPD, Choonara YE, Kumar P, Pillay V. A Review of Nanotechnology for Targeted Anti-schistosomal Therapy. Front Bioeng Biotechnol. 2020;8:32. doi:10.3389/fbioe.2020.00032

19. Xie W, Deng WW, Zan M, et al. Cancer Cell Membrane Camouflaged Nanoparticles to Realize Starvation Therapy Together with Checkpoint Blockades for Enhancing Cancer Therapy. ACS Nano. 2019;13(3):2849–2857. doi:10.1021/acsnano.8b03788

20. Shi Y, Lammers T. Combining Nanomedicine and Immunotherapy. Acc Chem Res. 2019;52(6):1543–1554. doi:10.1021/acs.accounts.9b00148

21. Wong KH, Yang D, Chen S, He C, Chen M. Development of nanoscale drug delivery systems of dihydroartemisinin for cancer therapy: a review. Asian J. Pharm Sci. 2022;17(4):475–490. doi:10.1016/j.ajps.2022.04.005

22. Ni K, Xu Z, Culbert A, et al. Synergistic checkpoint-blockade and radiotherapy–radiodynamic therapy via an immunomodulatory nanoscale metal–organic framework. Nat Biomed Eng. 2022;6(2):144–156. doi:10.1038/s41551-022-00846-w

23. Boyd LK, Mao X, Lu YJ. The complexity of prostate cancer: genomic alterations and heterogeneity. Nat Rev Urol. 2012;9(11):652–664. doi:10.1038/nrurol.2012.185

24. Cai M, Song XL, Li XA, et al. Current therapy and drug resistance in metastatic castration-resistant prostate cancer. Drug Resist Updates. 2023;68:100962. doi:10.1016/j.drup.2023.100962

25. Marín-Aguilera M, Codony-Servat J, Reig Ò, et al. Epithelial-to-Mesenchymal Transition Mediates Docetaxel Resistance and High Risk of Relapse in Prostate Cancer. Mol Cancer Ther. 2014;13(5):1270–1284. doi:10.1158/1535-7163.MCT-13-0775

26. Ayoubi‐Joshaghani MH, Seidi K, Azizi M, et al. Potential Applications of Advanced Nano/Hydrogels in Biomedicine: static, Dynamic, Multi‐Stage, and Bioinspired. Adv Funct Mater. 2020;30(45):2004098. doi:10.1002/adfm.202004098

27. Bailey BA, Ochyl LJ, Schwendeman SP, Moon JJ. Toward a Single‐Dose Vaccination Strategy with Self‐Encapsulating PLGA Microspheres. Adv Healthcare Materials. 2017;6(12):1601418. doi:10.1002/adhm.201601418

28. Abramson J, Dobeš J, Lyu M, Sonnenberg GF. The emerging family of RORγt+ antigen-presenting cells. Nat Rev Immunol. 2024;24(1):64–77. doi:10.1038/s41577-023-00906-5

29. Rosalia RA, Silva AL, Camps M, et al. Efficient ex vivo induction of T cells with potent anti-tumor activity by protein antigen encapsulated in nanoparticles. Cancer Immunol Immunother. 2013;62(7):1161–1173. doi:10.1007/s00262-013-1411-0

30. Blank C, Kuball J, Voelkl S, et al. Blockade of PD‐L1 (B7‐H1) augments human tumor‐specific T cell responses in vitro. Intl Journal of Cancer. 2006;119(2):317–327. doi:10.1002/ijc.21775

31. Kroon J, Metselaar JM, Storm G, Van Der Pluijm G. Liposomal nanomedicines in the treatment of prostate cancer. Cancer Treat Rev. 2014;40(4):578–584. doi:10.1016/j.ctrv.2013.10.005

32. Saneja A, Kumar R, Mintoo MJ, et al. Gemcitabine and betulinic acid co-encapsulated PLGA−PEG polymer nanoparticles for improved efficacy of cancer chemotherapy. Mater Sci Eng C. 2019;98:764–771. doi:10.1016/j.msec.2019.01.026

33. Jiang Y, Huo S, Hardie J, Liang XJ, Rotello VM. Progress and perspective of inorganic nanoparticle-based siRNA delivery systems. Expert Opin Drug Delivery. 2016;13(4):547–559. doi:10.1517/17425247.2016.1134486

34. Yao Y, Zhou Y, Liu L, et al. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front Mol Biosci. 2020;7:193. doi:10.3389/fmolb.2020.00193

35. Mohamed Isa ED, Ahmad H, Abdul Rahman MB, Gill MR. Progress in Mesoporous Silica Nanoparticles as Drug Delivery Agents for Cancer Treatment. Pharmaceutics. 2021;13(2):152. doi:10.3390/pharmaceutics13020152

36. Pilch J, Matysiak-Brynda E, Kowalczyk A, et al. New Unsymmetrical Bisacridine Derivatives Noncovalently Attached to Quaternary Quantum Dots Improve Cancer Therapy by Enhancing Cytotoxicity toward Cancer Cells and Protecting Normal Cells. ACS Appl Mater Interfaces. 2020;12(15):17276–17289. doi:10.1021/acsami.0c02621

37. Murugesan R, Raman S. Recent Trends in Carbon Nanotubes Based Prostate Cancer Therapy: aBiomedical Hybrid for Diagnosis and Treatment. CDD. 2022;19(2):229–237. doi:10.2174/1567201818666210224101456

38. Fang J, Nakamura H, Maeda H. The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv Drug Delivery Rev. 2011;63(3):136–151. doi:10.1016/j.addr.2010.04.009

39. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv Drug Delivery Rev. 2015;91:3–6. doi:10.1016/j.addr.2015.01.002

40. Clemons TD, Singh R, Sorolla A, Chaudhari N, Hubbard A, Iyer KS. Distinction Between Active and Passive Targeting of Nanoparticles Dictate Their Overall Therapeutic Efficacy. Langmuir. 2018;34(50):15343–15349. doi:10.1021/acs.langmuir.8b02946

41. Ni K, Lan G, Chan C, et al. Nanoscale metal-organic frameworks enhance radiotherapy to potentiate checkpoint blockade immunotherapy. Nat Commun. 2018;9(1):2351. doi:10.1038/s41467-018-04703-w

42. Kroon J, Buijs JT, Van Der Horst G, et al. Liposomal delivery of dexamethasone attenuates prostate cancer bone metastatic tumor growth In Vivo. Prostate. 2015;75(8):815–824. doi:10.1002/pros.22963

43. Jain AK, Thareja S. In vitro and in vivo characterization of pharmaceutical nanocarriers used for drug delivery. Artif Cells Nanomed Biotechnol. 2019;47(1):524–539. doi:10.1080/21691401.2018.1561457

44. Yoo J, Won YY. Phenomenology of the Initial Burst Release of Drugs from PLGA Microparticles. ACS Biomater Sci Eng. 2020;6(11):6053–6062. doi:10.1021/acsbiomaterials.0c01228

45. Yan L, Crayton SH, Thawani JP, Amirshaghaghi A, Tsourkas A, Cheng Z. A pH‐Responsive Drug‐Delivery Platform Based on Glycol Chitosan–Coated Liposomes. Small. 2015;11(37):4870–4874. doi:10.1002/smll.201501412

46. Nassir AM, Shahzad N, Ibrahim IAA, Ahmad I, Md S, Ain MR. Resveratrol-loaded PLGA nanoparticles mediated programmed cell death in prostate cancer cells. Saudi Pharm J. 2018;26(6):876–885. doi:10.1016/j.jsps.2018.03.009

47. Abbasi E, Aval SF, Akbarzadeh A, et al. Dendrimers: synthesis, applications, and properties. Nanoscale Res Lett. 2014;9(1):247. doi:10.1186/1556-276X-9-247

48. Shalmani AA, Wang A, Ahmed Z, et al. Tunable polymeric micelles for taxane and corticosteroid co-delivery. Drug Deliv Transl Res. doi:10.1007/s13346-023-01465-x

49. Thambiraj S, Vijayalakshmi R, Ravi Shankaran D. An effective strategy for development of docetaxel encapsulated gold nanoformulations for treatment of prostate cancer. Sci Rep. 2021;11(1):2808. doi:10.1038/s41598-020-80529-1

50. Thipe VC, Amiri KP, Bloebaum P, et al. Development of resveratrol-conjugated gold nanoparticles: interrelationship of increased resveratrol Corona on anti-tumor efficacy against breast, pancreatic and prostate cancers. IJN. 2019;14:4413–4428. doi:10.2147/IJN.S204443

51. Butterworth KT, Nicol JR, Ghita M, et al. Preclinical evaluation of gold-DTDTPA nanoparticles as theranostic agents in prostate cancer radiotherapy. Nanomedicine. 2016;11(16):2035–2047. doi:10.2217/nnm-2016-0062

52. Mandriota G, Di Corato R, Benedetti M, De Castro F, Fanizzi FP, Rinaldi R. Design and Application of Cisplatin-Loaded Magnetic Nanoparticle Clusters for Smart Chemotherapy. ACS Appl Mater Interfaces. 2019;11(2):1864–1875. doi:10.1021/acsami.8b18717

53. Shen L, Pan S, Niu D, et al. Facile synthesis of organosilica-capped mesoporous silica nanocarriers with selective redox-triggered drug release properties for safe tumor chemotherapy. Biomater Sci. 2019;7(5):1825–1832. doi:10.1039/C8BM01669K

54. Chen C, Ma T, Tang W, et al. Reversibly-regulated drug release using poly(tannic acid) fabricated nanocarriers for reduced secondary side effects in tumor therapy. Nanoscale Horiz. 2020;5(6):986–998. doi:10.1039/D0NH00032A

55. Tan RS. Glycosylated and non-glycosylated quantum dot-displayed peptides trafficked indiscriminately inside lung cancer cells but discriminately sorted in normal lung cells: an indispensable part in nanoparticle-based intracellular drug delivery. Asian J. Pharm Sci. 2018;13(3):197–211. doi:10.1016/j.ajps.2017.12.002

56. Sahoo NG, Bao H, Pan Y, et al. Functionalized carbon nanomaterials as nanocarriers for loading and delivery of a poorly water-soluble anticancer drug: a comparative study. Chem Commun. 2011;47(18):5235. doi:10.1039/c1cc00075f

57. Liu Y, Shang W, Liu H, et al. Biomimetic manganese-eumelanin nanocomposites for combined hyperthermia-immunotherapy against prostate cancer. J Nanobiotechnol. 2022;20(1):48. doi:10.1186/s12951-022-01248-5

58. He M, Cao Y, Chi C, et al. Unleashing novel horizons in advanced prostate cancer treatment: investigating the potential of prostate specific membrane antigen-targeted nanomedicine-based combination therapy. Front Immunol. 2023;14:1265751. doi:10.3389/fimmu.2023.1265751

59. Tian Y, Liu Z, Wang J, et al. Nanomedicine for Combination Urologic Cancer Immunotherapy. Pharmaceutics. 2023;15(2):546. doi:10.3390/pharmaceutics15020546

60. Vicente‐Ruiz S, Serrano‐Martí A, Armiñán A, Vicent MJ. Nanomedicine for the Treatment of Advanced Prostate Cancer. Adv Ther. 2021;4(1):2000136. doi:10.1002/adtp.202000136

61. Luo ZW, Xia K, Liu YW, et al. Extracellular Vesicles from Akkermansia muciniphila Elicit Antitumor Immunity Against Prostate Cancer via Modulation of CD8+ T Cells and Macrophages. IJN. 2021;16:2949–2963. doi:10.2147/IJN.S304515

62. Rastinehad AR, Anastos H, Wajswol E, et al. Gold nanoshell-localized photothermal ablation of prostate tumors in a clinical pilot device study. Proc Natl Acad Sci USA. 2019;116(37):18590–18596. doi:10.1073/pnas.1906929116

63. Pan S, Zhang Y, Huang M, et al. Urinary exosomes-based Engineered Nanovectors for Homologously Targeted Chemo-Chemodynamic Prostate Cancer Therapy via abrogating EGFR/AKT/NF-kB/IkB signaling. Biomaterials. 2021;275:120946. doi:10.1016/j.biomaterials.2021.120946

64. Wang H, He Z, Gao Y, et al. Dual‐Pronged Attack: pH‐Driven Membrane‐Anchored NIR Dual‐Type Nano‐Photosensitizer Excites Immunogenic Pyroptosis and Sequester Immune Checkpoint for Enhanced Prostate Cancer Photo‐Immunotherapy. Adv. Sci. 2023;10(28):2302422. doi:10.1002/advs.202302422

65. Guo S, Feng J, Li Z, et al. Improved cancer immunotherapy strategies by nanomedicine. WIREs Nanomed Nanobiotechnol. 2023;15(3):e1873. doi:10.1002/wnan.1873

66. Chaudagar KK, Landon-Brace N, Solanki A, et al. Cabozantinib Unlocks Efficient In Vivo Targeted Delivery of Neutrophil-Loaded Nanoparticles into Murine Prostate Tumors. Mol Cancer Ther. 2021;20(2):438–449. doi:10.1158/1535-7163.MCT-20-0167

67. Mu X, Zhang M, Wei A, et al. Doxorubicin and PD-L1 siRNA co-delivery with stem cell membrane-coated polydopamine nanoparticles for the targeted chemoimmunotherapy of PCa bone metastases. Nanoscale. 2021;13(19):8998–9008. doi:10.1039/D0NR08024A

68. Dai L, Shen G, Wang Y, Yang P, Wang H, Liu Z. PSMA-targeted melanin-like nanoparticles as a multifunctional nanoplatform for prostate cancer theranostics. J Mater Chem B. 2021;9(4):1151–1161. doi:10.1039/D0TB02576C

69. Saniee F, Shabani Ravari N, Goodarzi N, et al. Glutamate-urea-based PSMA-targeted PLGA nanoparticles for prostate cancer delivery of docetaxel. Pharm Developm Techn. 2021;26(4):381–389. doi:10.1080/10837450.2021.1875238

70. Chandran SS, Banerjee R, S CM, R PMG, Denmeade R. Characterization of a targeted nanoparticle functionalized with a urea-based inhibitor of prostate-specific membrane antigen (PSMA). Cancer Biol Ther. 2008;7(6):974–982. doi:10.4161/cbt.7.6.5968

71. Zacherl MJ, Gildehaus FJ, Mittlmeier L, et al. First Clinical Results for PSMA-Targeted α-Therapy Using 225 Ac-PSMA-I&T in Advanced-mCRPC Patients. J Nucl Med. 2021;62(5):669–674. doi:10.2967/jnumed.120.251017

72. An S, Huang G, Liu J, Wei W. PSMA-targeted theranostics of solid tumors: applications beyond prostate cancers. Eur J Nucl Med Mol Imaging. 2022;49(12):3973–3976. doi:10.1007/s00259-022-05905-7

73. Xie L, Li J, Wang L, Dai Y. Engineering metal‐phenolic networks for enhancing cancer therapy by tumor microenvironment modulation. WIREs Nanomed Nanobiotechnol. 2023;15(3):e1864. doi:10.1002/wnan.1864

74. Liu F, Lin L, Zhang Y, et al. A Tumor‐Microenvironment‐Activated Nanozyme‐Mediated Theranostic Nanoreactor for Imaging‐Guided Combined Tumor Therapy. Adv Mater. 2019;31(40):1902885. doi:10.1002/adma.201902885

75. Liu Q, Chen F, Hou L, et al. Nanocarrier-Mediated Chemo-Immunotherapy Arrested Cancer Progression and Induced Tumor Dormancy in Desmoplastic Melanoma. ACS Nano. 2018;12(8):7812–7825. doi:10.1021/acsnano.8b01890

76. Yang Q, Yang Y, Li L, Sun W, Zhu X, Huang Y. Polymeric Nanomedicine for Tumor-Targeted Combination Therapy to Elicit Synergistic Genotoxicity against Prostate Cancer. ACS Appl Mater Interfaces. 2015;7(12):6661–6673. doi:10.1021/am509204u

77. Gilman A. The initial clinical trial of nitrogen mustard. Am J Surg. 1963;105(5):574–578. doi:10.1016/0002-9610(63)90232-0

78. Qin SY, Cheng YJ, Lei Q, Zhang AQ, Zhang XZ. Combinational strategy for high-performance cancer chemotherapy. Biomaterials. 2018;171:178–197. doi:10.1016/j.biomaterials.2018.04.027

79. Veiseh O, Gunn JW, Zhang M. Design and fabrication of magnetic nanoparticles for targeted drug delivery and imaging. Adv Drug Delivery Rev. 2010;62(3):284–304. doi:10.1016/j.addr.2009.11.002

80. Ali I, Rahis-Uddin S, K AR, M AW, Haque A W. Advances in Nano Drugs for Cancer Chemotherapy. CCDT. 2011;11(2):135–146. doi:10.2174/156800911794328493

81. Palacios Eito A, Cabezas SG, Ugalde PF, et al. Characterization and adequacy of the use of radiotherapy and its trend in time. Radiother Oncol. 2013;106(2):260–265. doi:10.1016/j.radonc.2012.10.008

82. Lawrence TS, Haffty BG, Harris JR. Milestones in the Use of Combined-Modality Radiation Therapy and Chemotherapy. JCO. 2014;32(12):1173–1179. doi:10.1200/JCO.2014.55.2281

83. Chen Q, Chen J, Yang Z, et al. Nanoparticle‐Enhanced Radiotherapy to Trigger Robust Cancer Immunotherapy. Adv Mater. 2019;31(10):1802228. doi:10.1002/adma.201802228

84. Guan X, Sun L, Shen Y, et al. Nanoparticle-enhanced radiotherapy synergizes with PD-L1 blockade to limit post-surgical cancer recurrence and metastasis. Nat Commun. 2022;13(1):2834. doi:10.1038/s41467-022-30543-w

85. Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, Phase 2 trial. Lancet. 2017;389(10064):67–76. doi:10.1016/S0140-6736(16)32455-2

86. Runnels J, Bloom JR, Hsieh K, et al. Combining Radiotherapy and Immunotherapy in Head and Neck Cancer. Biomedicines. 2023;11(8):2097. doi:10.3390/biomedicines11082097

87. Hurwitz AA, Foster BA, Kwon ED, et al. Combination Immunotherapy of Primary Prostate Cancer in a Transgenic Mouse Model Using CTLA-4 Blockade.

88. Dudzinski SO, Cameron BD, Wang J, Rathmell JC, Giorgio TD, Kirschner AN. Combination immunotherapy and radiotherapy causes an abscopal treatment response in a mouse model of castration resistant prostate cancer. J Immunother Cancer. 2019;7(1):218. doi:10.1186/s40425-019-0704-z

89. Postow MA, Callahan MK, Barker CA, et al. Immunologic Correlates of the Abscopal Effect in a Patient with Melanoma. N Engl J Med. 2012;366(10):925–931. doi:10.1056/NEJMoa1112824

90. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203(5):1259–1271. doi:10.1084/jem.20052494

91. Mouw KW, Goldberg MS, Konstantinopoulos PA, D’Andrea AD. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer Discovery. 2017;7(7):675–693. doi:10.1158/2159-8290.CD-17-0226

92. Gong X, Li X, Jiang T, et al. Combined Radiotherapy and Anti–PD-L1 Antibody Synergistically Enhances Antitumor Effect in Non–Small Cell Lung Cancer. J Thorac Oncol. 2017;12(7):1085–1097. doi:10.1016/j.jtho.2017.04.014

93. Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015;16(13):e498–e509. doi:10.1016/S1470-2045(15)00007-8

94. Victor C T-S, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520 (7547):373–377. doi:10.1038/nature14292

95. Wang M, Song J, Zhou F, et al. NIR‐Triggered Phototherapy and Immunotherapy via an Antigen‐Capturing Nanoplatform for Metastatic Cancer Treatment. Adv. Sci. 2019;6(10):1802157. doi:10.1002/advs.201802157

96. Au KM, Tripathy A, Lin CPI, et al. Bespoke Pretargeted Nanoradioimmunotherapy for the Treatment of Non-Hodgkin Lymphoma. ACS Nano. 2018;12(2):1544–1563. doi:10.1021/acsnano.7b08122

97. Wang Z, Xing H, Liu A, et al. Multifunctional nano-system for multi-mode targeted imaging and enhanced photothermal therapy of metastatic prostate cancer. Acta Biomater. 2023;166:581–592. doi:10.1016/j.actbio.2023.05.014

98. Deng G, Sun Z, Li S, et al. Cell-Membrane Immunotherapy Based on Natural Killer Cell Membrane Coated Nanoparticles for the Effective Inhibition of Primary and Abscopal Tumor Growth. ACS Nano. 2018;12(12):12096–12108. doi:10.1021/acsnano.8b05292

99. Liu F, Dong J, Shen Y, et al. Comparison of PET/CT and MRI in the Diagnosis of Bone Metastasis in Prostate Cancer Patients: a Network Analysis of Diagnostic Studies. Front Oncol. 2021;11:736654. doi:10.3389/fonc.2021.736654

100. Huang H-C, Barua S, Sharma G, Dey SK, Rege K. Inorganic nanoparticles for cancer imaging and therapy. J Control Release. 2011;155(3):344–357. doi:10.1016/j.jconrel.2011.06.004

101. Zhao L, Liu Y, Chang R, Xing R, Yan X. Supramolecular Photothermal Nanomaterials as an Emerging Paradigm toward Precision Cancer Therapy. advanced Functional Materials. 2019;29(4):1806877. doi:10.1002/adfm.201806877

102. Ji B, Wei M, Yang B. Recent advances in nanomedicines for photodynamic therapy (PDT)-driven cancer immunotherapy. Theranostics. 2022;12(1):434–458. doi:10.7150/thno.67300

103. Bhatia SN, Chen X, Dobrovolskaia MA, Lammers T. Cancer nanomedicine. Nat Rev Cancer. 2022;22(10):550–556. doi:10.1038/s41568-022-00496-9

104. Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17(1):20–37. doi:10.1038/nrc.2016.108

105. Yang Z, Gao D, Zhao J, et al. Thermal immuno-nanomedicine in cancer. Nat Rev Clin Oncol. 2023;20(2):116–134. doi:10.1038/s41571-022-00717-y

106. Garbayo E, Pascual‐Gil S, Rodríguez‐Nogales C, Saludas L, Estella‐Hermoso De Mendoza A, Blanco‐Prieto MJ. Nanomedicine and drug delivery systems in cancer and regenerative medicine. WIREs Nanomed Nanobiotechnol. 2020;12(5):e1637. doi:10.1002/wnan.1637

107. McGranahan N, Furness AJS, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351(6280):1463–1469. doi:10.1126/science.aaf1490

108. Chakraborty DD, Chakraborty P. Cancer-Specific Nanomedicine Delivery Systems and the Role of the Tumor Microenvironment: a Critical Linkage. Current Nanomed. 2023.

109. Arrieta VA, Dmello C, McGrail DJ, et al. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Investig. 2023;133(2):e163447. doi:10.1172/JCI163447

110. Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discovery. 2018;8(9):1069–1086. doi:10.1158/2159-8290.CD-18-0367

111. Ou B Saponin Nanoparticle Adjuvants Incorporating Toll-Like Receptor Agonists Improve Vaccine Immunomodulation.

112. Zhou P, Yan B, Wei B, et al. Quercetin-solid lipid nanoparticle-embedded hyaluronic acid functionalized hydrogel for immunomodulation to promote bone reconstruction. Regen Biomaterials. 2023;10:rbad025. doi:10.1093/rb/rbad025

113. Janic B, Neff R, Brown SL, et al. Radiation and Gold Nanoparticle Immunomodulation in MDA MB 231 Mouse Breast Cancer Model. Intern J Radiation Oncol Biol Physics. 2020;108(3):e545–e546. doi:10.1016/j.ijrobp.2020.07.1699

114. Shaw J, Pearson RM. Nanoparticle personalized biomolecular Corona: implications of pre-existing conditions for immunomodulation and cancer. Biomater Sci. 2022;10(10):2540–2549. doi:10.1039/D2BM00315E

115. Khezrian A, Ahmadi M, Mokarram P, Afshar S, Parvizi Y. A Review of Recent Advances in Peptide-Based Anticancer Therapeutic Vaccines and Nanovaccines in Prostate Cancer. Int J Pept Res Ther. 2023;29(5):70. doi:10.1007/s10989-023-10542-1

116. Bakhshi P, Nourizadeh M, Sharifi L, Farajollahi MM, Mohsenzadegan M. Development of dendritic cell loaded MAGE-A2 long peptide; a potential target for tumor-specific T cell-mediated prostate cancer immunotherapy. Cancer Cell Int. 2023;23(1):270. doi:10.1186/s12935-023-03108-0

117. Lin W, Li C, Xu N, et al. Dual-Functional PLGA Nanoparticles Co-Loaded with Indocyanine Green and Resiquimod for Prostate Cancer Treatment. IJN. 2021;16:2775–2787. doi:10.2147/IJN.S301552

118. Zhou L, Murphy KC, Snyder J, DeMarco KD, Ma B, Ruscetti M. Abstract B035: leveraging therapy-induced senescence for prostate cancer immunotherapy. Cancer Res. 2023;83(11_Supplement):B035–B035. doi:10.1158/1538-7445.PRCA2023-B035

119. Shen X, Zhou S, Yang Y, et al. TAM-targeted reeducation for enhanced cancer immunotherapy: mechanism and recent progress. Front Oncol. 2022;12:1034842. doi:10.3389/fonc.2022.1034842

120. Bhatia V, Kamat NV, Pariva TE, et al. Targeting advanced prostate cancer with STEAP1 chimeric antigen receptor T cell and tumor-localized IL-12 immunotherapy. Nat Commun. 2023;14(1):2041. doi:10.1038/s41467-023-37874-2

121. Autio KA, Higano CS, Nordquist L, et al. First-in-human, Phase 1 study of PF-06753512, a vaccine-based immunotherapy regimen (VBIR), in non-metastatic hormone-sensitive biochemical recurrence and metastatic castration-resistant prostate cancer (mCRPC). J Immunother Cancer. 2023;11(3):e005702. doi:10.1136/jitc-2022-005702

122. Kaittanis C, Bolaender A, Yoo B, Shah N, Ouerfelli O, Grimm J. Targetable Clinical Nanoparticles for Precision Cancer Therapy Based on Disease-Specific Molecular Inflection Points. Nano Lett. 2017;17(11):7160–7168. doi:10.1021/acs.nanolett.7b04209

123. Tieu T, Irani S, Bremert KL, et al. Patient‐Derived Prostate Cancer Explants: a Clinically Relevant Model to Assess siRNA‐Based Nanomedicines. Adv Healthcare Materials. 2021;10(6):2001594. doi:10.1002/adhm.202001594

124. Zhang C, Pu K. Molecular and nanoengineering approaches towards activatable cancer immunotherapy. Chem Soc Rev. 2020;49(13):4234–4253. doi:10.1039/C9CS00773C

125. Hare JI, Lammers T, Ashford MB, Puri S, Storm G, Barry ST. Challenges and strategies in anti-cancer nanomedicine development: an industry perspective. Adv Drug Delivery Rev. 2017;108:25–38. doi:10.1016/j.addr.2016.04.025

126. Williams RM, Lee C, Heller DA. A Fluorescent Carbon Nanotube Sensor Detects the Metastatic Prostate Cancer Biomarker uPA. ACS Sens. 2018;3(9):1838–1845. doi:10.1021/acssensors.8b00631

127. Uhlirova D, Stankova M, Docekalova M, et al. A Rapid Method for the Detection of Sarcosine Using SPIONs/Au/CS/SOX/NPs for Prostate Cancer Sensing. IJMS. 2018;19(12):3722. doi:10.3390/ijms19123722

128. Omer M, Andersen VL, Nielsen JS, Wengel J, Kjems J. Improved Cancer Targeting by Multimerizing Aptamers on Nanoscaffolds. Mol Ther Nucleic Acids. 2020;22:994–1003. doi:10.1016/j.omtn.2020.10.013

129. Zhang C, Siddhanta S, Paria D, Li Y, Zheng C, Barman I. Spectroscopy‐Assisted Label‐free Molecular Analysis of Live Cell Surface with Vertically Aligned Plasmonic Nanopillars. Small. 2021;17(21):2100161. doi:10.1002/smll.202100161

130. Schilham MGM, Zamecnik P, Privé BM, et al. Head-to-Head Comparison of 68 Ga-Prostate-Specific Membrane Antigen PET/CT and Ferumoxtran-10–Enhanced MRI for the Diagnosis of Lymph Node Metastases in Prostate Cancer Patients. J Nucl Med. 2021;62(9):1258–1263. doi:10.2967/jnumed.120.258541

131. Repp L, Unterberger CJ, Ye Z, et al. Oligo(Lactic Acid)8-Docetaxel Prodrug-Loaded PEG-b-PLA Micelles for Prostate Cancer. Nanomaterials. 2021;11(10):2745. doi:10.3390/nano11102745

132. Loiseau A, Boudon J, Mirjolet C, Morgand V, Millot N. About the Influence of PEG Spacers on the Cytotoxicity of Titanate Nanotubes-Docetaxel Nanohybrids against a Prostate Cancer Cell Line. Nanomaterials. 2021;11(10):2733. doi:10.3390/nano11102733

133. Cohen L, Assaraf YG, Livney YD. Novel Selectively Targeted Multifunctional Nanostructured Lipid Carriers for Prostate Cancer Treatment. Pharmaceutics. 2021;14(1):88. doi:10.3390/pharmaceutics14010088

134. Qu Z, Ren Y, Shen H, Wang H, Shi L, Tong D. Combination Therapy of Metastatic Castration-Recurrent Prostate Cancer: hyaluronic Acid Decorated, Cabazitaxel-Prodrug and Orlistat Co-Loaded Nano-System. DDDT. 2021;15:3605–3616. doi:10.2147/DDDT.S306684

135. Bennie LA, Feng J, Emmerson C, et al. Formulating RALA/Au nanocomplexes to enhance nanoparticle internalisation efficiency, sensitising prostate tumour models to radiation treatment. J Nanobiotechnol. 2021;19(1):279. doi:10.1186/s12951-021-01019-8

136. Lip H, Amini MA, Zetrini A, et al. Redox-responsive nanoparticles enhance radiation therapy by altering multifaceted radio-resistance mechanisms in human castration-resistant prostate cancer cells and xenografts. Radiother Oncol. 2022;170:213–223. doi:10.1016/j.radonc.2022.02.026

137. Asha Krishnan M, Yadav K, Roach P, Chelvam V. A targeted near-infrared nanoprobe for deep-tissue penetration and imaging of prostate cancer. Biomater Sci. 2021;9(6):2295–2312. doi:10.1039/D0BM01970D

138. Cordonnier A, Boyer D, Besse S, et al. Synthesis and in vitro preliminary evaluation of prostate-specific membrane antigen targeted upconversion nanoparticles as a first step towards radio/fluorescence-guided surgery of prostate cancer. J Mater Chem B. 2021;9(36):7423–7434. doi:10.1039/D1TB00777G

139. Khoobchandani M, Khan A, Katti KK, et al. Green nanotechnology of MGF-AuNPs for immunomodulatory intervention in prostate cancer therapy. Sci Rep. 2021;11(1):16797. doi:10.1038/s41598-021-96224-8

140. Hu C, Gu F, Gong C, Xia Q, Gao Y, Gao S. Co-delivery of the autophagy inhibitor si-Beclin1 and the doxorubicin nano-delivery system for advanced prostate cancer treatment. J Biomater Appl. 2022;36(7):1317–1331. doi:10.1177/08853282211060252

141. Wang Z, Xu D. MiR-197 Inhibitor Loaded AbCD133@MSNs@GNR Affects the Development of Prostate Cancer Through Targeting ITGAV. Front Cell Develop Biol. 2021;9.

142. Wu D, Zhu ZQ, Tang HX, et al. Efficacy-shaping nanomedicine by loading Calcium Peroxide into Tumor Microenvironment-responsive Nanoparticles for the Antitumor Therapy of Prostate Cancer. Theranostics. 2020;10(21):9808–9829. doi:10.7150/thno.43631

143. Hadi MM, Nesbitt H, Masood H, et al. Investigating the performance of a novel pH and cathepsin B sensitive, stimulus-responsive nanoparticle for optimised sonodynamic therapy in prostate cancer. J Control Release. 2021;329:76–86. doi:10.1016/j.jconrel.2020.11.040

144. Sun G, Sun K, Sun J. Combination prostate cancer therapy: prostate-specific membranes antigen targeted, pH-sensitive nanoparticles loaded with doxorubicin and tanshinone. Drug Delivery. 2021;28(1):1132–1140. doi:10.1080/10717544.2021.1931559

145. Braga CB, Kido LA, Lima EN, et al. Enhancing the Anticancer Activity and Selectivity of Goniothalamin Using pH-Sensitive Acetalated Dextran (Ac-Dex) Nanoparticles: a Promising Platform for Delivery of Natural Compounds. ACS Biomater Sci Eng. 2020;6(5):2929–2942. doi:10.1021/acsbiomaterials.0c00057

146. Radaic A, Joo NE, Jeong SH, Yoo SI, Kotov N, Kapila YL. Phosphatidylserine-Gold Nanoparticles (PS-AuNP) Induce Prostate and Breast Cancer Cell Apoptosis. Pharmaceutics. 2021;13(7):1094. doi:10.3390/pharmaceutics13071094

147. An Y, Zhao J. Functionalized Selenium Nanotherapeutics Synergizes With Zoledronic Acid to Suppress Prostate Cancer Cell Growth Through Induction of Mitochondria-Mediated Apoptosis and Cell Cycle S Phase Arrest. Front Oncol. 2021;11:685784. doi:10.3389/fonc.2021.685784

148. Verde V, Longo A, Cucci LM, et al. Anti-Angiogenic and Anti-Proliferative Graphene Oxide Nanosheets for Tumor Cell Therapy. IJMS. 2020;21(15):5571. doi:10.3390/ijms21155571

149. Tanaudommongkon I, Tanaudommongkon A, Prathipati P, Nguyen JT, Keller ET, Dong X. Curcumin Nanoparticles and Their Cytotoxicity in Docetaxel-Resistant Castration-Resistant Prostate Cancer Cells. Biomedicines. 2020;8(8):253. doi:10.3390/biomedicines8080253

150. Güney Akkurt M, Gülsoy M. Polylactide nanoparticles encapsulating indocyanine green for photothermal therapy of prostate cancer cells. Photodiagn Photodyn Ther. 2022;37:102693. doi:10.1016/j.pdpdt.2021.102693

151. Guo Q, Dong Y, Zhang Y, et al. Sequential Release of Pooled siRNAs and Paclitaxel by Aptamer-Functionalized Shell–Core Nanoparticles to Overcome Paclitaxel Resistance of Prostate Cancer. ACS Appl Mater Interfaces. 2021;13(12):13990–14003. doi:10.1021/acsami.1c00852

152. Cole G, Ali AA, McErlean E, et al. DNA vaccination via RALA nanoparticles in a microneedle delivery system induces a potent immune response against the endogenous prostate cancer stem cell antigen. Acta Biomater. 2019;96:480–490. doi:10.1016/j.actbio.2019.07.003

153. Islam MA, Rice J, Reesor E, et al. Adjuvant-pulsed mRNA vaccine nanoparticle for immunoprophylactic and therapeutic tumor suppression in mice. Biomaterials. 2021;266:120431. doi:10.1016/j.biomaterials.2020.120431

154. Fan Y, Moon J. Nanoparticle Drug Delivery Systems Designed to Improve Cancer Vaccines and Immunotherapy. Vaccines. 2015;3(3):662–685. doi:10.3390/vaccines3030662

155. Van Der Meel R, Sulheim E, Shi Y, Kiessling F, Mulder WJM, Lammers T. Smart cancer nanomedicine. Nat Nanotechnol. 2019;14(11):1007–1017. doi:10.1038/s41565-019-0567-y

156. Sasikumar A, Kamalasanan K. Nanomedicine for prostate cancer using nanoemulsion: a review. J Control Release. 2017;260:111–123. doi:10.1016/j.jconrel.2017.06.001

157. Pugazhendhi A, TNJI E, Karuppusamy I, Kathirvel B. Inorganic nanoparticles: a potential cancer therapy for human welfare. Int J Pharm. 2018;539(1–2):104–111. doi:10.1016/j.ijpharm.2018.01.034

158. Guo J, Schlich M, Cryan JF, O’Driscoll CM. Targeted Drug Delivery via Folate Receptors for the Treatment of Brain Cancer: can the Promise Deliver? J Pharmaceut Sci. 2017;106(12):3413–3420. doi:10.1016/j.xphs.2017.08.009

159. Zhao Z, Zheng L, Chen W, Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: recent advances and future perspectives. J Hematol Oncol. 2019;12(1):126. doi:10.1186/s13045-019-0817-3

160. Davoodi P, Lee LY, Xu Q, et al. Drug delivery systems for programmed and on-demand release. Adv Drug Delivery Rev. 2018;132:104–138. doi:10.1016/j.addr.2018.07.002

161. Zhang A, Jung K, Li A, Liu J, Boyer C. Recent advances in stimuli-responsive polymer systems for remotely controlled drug release. Prog Polym Sci. 2019;99:101164. doi:10.1016/j.progpolymsci.2019.101164

162. Fenton OS, Olafson KN, Pillai PS, Mitchell MJ, Langer R. Advances in Biomaterials for Drug Delivery. Adv Mater. 2018;30(29):1705328. doi:10.1002/adma.201705328

163. Cheng Z, Li M, Dey R, Chen Y. Nanomaterials for cancer therapy: current progress and perspectives. J Hematol Oncol. 2021;14(1):85. doi:10.1186/s13045-021-01096-0

164. Nie W, Wu G, Zhang J, et al. Responsive Exosome Nano‐bioconjugates for Synergistic Cancer Therapy. Angew Chem Int Ed. 2020;59(5):2018–2022. doi:10.1002/anie.201912524

165. Jeevarathinam AS, Lemaster JE, Chen F, Zhao E, Jokerst JV. Photoacoustic Imaging Quantifies Drug Release from Nanocarriers via Redox Chemistry of Dye‐Labeled Cargo. Angew Chem Int Ed. 2020;59(12):4678–4683. doi:10.1002/anie.201914120

166. Bansal KK, Özliseli E, Rosling A, Rosenholm JM. Synthesis and Evaluation of Novel Functional Polymers Derived from Renewable Jasmine Lactone for Stimuli‐Responsive Drug Delivery. Adv Funct Mater. 2021;31(33):2101998. doi:10.1002/adfm.202101998

167. Liu F, Niko Y, Bouchaala R, et al. Drug‐Sponge Lipid Nanocarrier for in Situ Cargo Loading and Release Using Dynamic Covalent Chemistry. Angew Chem Int Ed. 2021;60(12):6573–6580. doi:10.1002/anie.202014259

168. Ruan L, Chen J, Du C, et al. Mitochondrial temperature-responsive drug delivery reverses drug resistance in lung cancer. Bioact Mater. 2022;13:191–199. doi:10.1016/j.bioactmat.2021.10.045

169. Cai X, Mao D, Wang C, Kong D, Cheng X, Liu B. Multifunctional Liposome: a Bright AIEgen–Lipid Conjugate with Strong Photosensitization. Angew Chem Int Ed. 2018;57(50):16396–16400. doi:10.1002/anie.201809641

170. Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics. 2020;10(10):4557–4588. doi:10.7150/thno.38069

171. Ma Y, Mao G, Wu G, Cui Z, Zhang XE, Huang W. CRISPR-dCas9-Guided and Telomerase-Responsive Nanosystem for Precise Anti-Cancer Drug Delivery. ACS Appl Mater Interfaces. 2021;13(7):7890–7896. doi:10.1021/acsami.0c19217

172. Jeong C, Uthaman S, Bagheri B, et al. Self-assembled heptamethine cyanine dye dimer as a novel theranostic drug delivery carrier for effective image-guided chemo-photothermal cancer therapy. J Control Release. 2021;329:50–62. doi:10.1016/j.jconrel.2020.11.046

173. Zhou Z, Vázquez-González M, Willner I. Stimuli-responsive metal–organic framework nanoparticles for controlled drug delivery and medical applications. Chem Soc Rev. 2021;50(7):4541–4563. doi:10.1039/D0CS01030H

174. Jia HR, Zhu YX, Liu X, et al. Construction of Dually Responsive Nanotransformers with Nanosphere–Nanofiber–Nanosphere Transition for Overcoming the Size Paradox of Anticancer Nanodrugs. ACS Nano. 2019;13(10):11781–11792. doi:10.1021/acsnano.9b05749

175. Liu J, Li F, Zheng J, Li B, Zhang D, Jia L. Redox/NIR dual-responsive MoS2 for synergetic chemo-photothermal therapy of cancer. J Nanobiotechnol. 2019;17(1):78. doi:10.1186/s12951-019-0510-2

176. Wang Z, Ju Y, Ali Z, et al. Near-infrared light and tumor microenvironment dual responsive size-switchable nanocapsules for multimodal tumor theranostics. Nat Commun. 2019;10(1):4418. doi:10.1038/s41467-019-12142-4

177. Peng J, Yang Q, Xiao Y, et al. Tumor Microenvironment Responsive Drug‐Dye‐Peptide Nanoassembly for Enhanced Tumor‐Targeting, Penetration, and Photo‐Chemo‐Immunotherapy. Adv Funct Mater. 2019;29(19):1900004. doi:10.1002/adfm.201900004

178. Shao D, Zhang F, Chen F, et al. Biomimetic Diselenide‐Bridged Mesoporous Organosilica Nanoparticles as an X‐ray‐Responsive Biodegradable Carrier for Chemo‐Immunotherapy. Adv Mater. 2020;32(50):2004385. doi:10.1002/adma.202004385

179. Dai L, Li X, Zheng X, et al. TGF-β blockade-improved chemo-immunotherapy with pH/ROS cascade-responsive micelle via tumor microenvironment remodeling. Biomaterials. 2021;276:121010. doi:10.1016/j.biomaterials.2021.121010

180. Ren SZ, Zhu D, Zhu XH, et al. Nanoscale Metal–Organic-Frameworks Coated by Biodegradable Organosilica for pH and Redox Dual Responsive Drug Release and High-Performance Anticancer Therapy. ACS Appl Mater Interfaces. 2019;11(23):20678–20688. doi:10.1021/acsami.9b04236

181. Huang S, Yuan J, Xie Y, et al. Targeting nano-regulator based on metal–organic frameworks for enhanced immunotherapy of bone metastatic prostate cancer. Cancer Nano. 2023;14(1):43. doi:10.1186/s12645-023-00200-y

182. Filippi L, Frantellizzi V, Chiaravalloti A, et al. Prognostic and Theranostic Applications of Positron Emission Tomography for a Personalized Approach to Metastatic Castration-Resistant Prostate Cancer. IJMS. 2021;22(6):3036. doi:10.3390/ijms22063036

183. Bou-Dargham MJ, Sha L, Sang QXA, Zhang J. Immune landscape of human prostate cancer: immune evasion mechanisms and biomarkers for personalized immunotherapy. BMC Cancer. 2020;20(1):572. doi:10.1186/s12885-020-07058-y

184. Hood L, Friend SH. Predictive, personalized, preventive, participatory (P4) cancer medicine. Nat Rev Clin Oncol. 2011;8(3):184–187. doi:10.1038/nrclinonc.2010.227

185. Bragazzi N. From P0 to P6 medicine, a model of highly participatory, narrative, interactive, and “augmented” medicine: some considerations on Salvatore Iaconesi’s clinical story. PPA. 2013;353. doi:10.2147/PPA.S38578

186. Guo Y, Wang R, Shi J, et al. Machine learning-based integration develops a metabolism-derived consensus model for improving immunotherapy in pancreatic cancer. J Immunother Cancer. 2023;11(9):e007466. doi:10.1136/jitc-2023-007466

187. Mueller AN, Morrisey S, Miller HA, et al. Prediction of lung cancer immunotherapy response via machine learning analysis of immune cell lineage and surface markers. CBM. 2022;34(4):681–692. doi:10.3233/CBM-210529

188. Uemura S, Aitken CE, Korlach J, Flusberg BA, Turner SW, Puglisi JD. Real-time tRNA transit on single translating ribosomes at codon resolution. Nature. 2010;464(7291):1012–1017. doi:10.1038/nature08925

189. Eid J, Fehr A, Gray J, et al. Real-Time DNA Sequencing from Single Polymerase Molecules; 2009.

190. Zhang Q, Wang W, Huang S, et al. Capture and selective release of multiple types of circulating tumor cells using smart DNAzyme probes. Chem Sci. 2020;11(7):1948–1956. doi:10.1039/C9SC04309H

191. Loeian MS, Mehdi Aghaei S, Farhadi F, et al. Liquid biopsy using the nanotube-CTC-chip: capture of invasive CTCs with high purity using preferential adherence in breast cancer patients. Lab Chip. 2019;19(11):1899–1915. doi:10.1039/C9LC00274J

192. Zhang CY, Yeh HC, Kuroki MT, Wang TH. Single-quantum-dot-based DNA nanosensor. Nat Mater. 2005;4(11):826–831. doi:10.1038/nmat1508

193. Iv CW S, Wang JL, Ohiri KA, et al. Magnetic separation of acoustically focused cancer cells from blood for magnetographic templating and analysis. Lab Chip. 2016;16(19):3833–3844. doi:10.1039/C6LC00719H

194. Huang J, Zhuang C, Chen J, et al. Targeted Drug/Gene/Photodynamic Therapy via a Stimuli‐Responsive Dendritic‐Polymer‐Based Nanococktail for Treatment of EGFR‐TKI‐Resistant Non‐Small‐Cell Lung Cancer. Adv Mater. 2022;34(27):2201516. doi:10.1002/adma.202201516

195. Rashid MBMA, Toh TB, Hooi L, et al. Optimizing drug combinations against multiple myeloma using a quadratic phenotypic optimization platform (QPOP). Sci Transl Med. 2018;10(453):eaan0941. doi:10.1126/scitranslmed.aan0941

196. Zarrinpar A, Lee DK, Silva A, et al. Individualizing liver transplant immunosuppression using a phenotypic personalized medicine platform. Sci Transl Med. 2016;8(333). doi:10.1126/scitranslmed.aac5954

197. Blasiak A, Khong J. CURATE.AI: optimizing Personalized Medicine with Artificial Intelligence. SLAS Technology. 2020;25(2):95–105. doi:10.1177/2472630319890316

198. Chen Z, Huo J, Hao L, Zhou J. Multiscale modeling and simulations of responsive polymers. Curr. Opin. Chem. Eng. 2019;23:21–33. doi:10.1016/j.coche.2019.02.004

199. Wang YL, Lu ZY, Laaksonen A. Specific binding structures of dendrimers on lipid bilayer membranes. Phys Chem Chem Phys. 2012;14(23):8348. doi:10.1039/c2cp40700k

200. Shinoda W, DeVane R, Klein ML. Computer simulation studies of self-assembling macromolecules. Curr Opin Struct Biol. 2012;22(2):175–186. doi:10.1016/j.sbi.2012.01.011

201. Vuković L, Madriaga A, Kuzmis A, et al. Solubilization of Therapeutic Agents in Micellar Nanomedicines. Langmuir. 2013;29(51):15747–15754. doi:10.1021/la403264w

202. Vácha R, Martinez-Veracoechea FJ, Frenkel D. Intracellular Release of Endocytosed Nanoparticles Upon a Change of Ligand–Receptor Interaction. ACS Nano. 2012;6(12):10598–10605. doi:10.1021/nn303508c

203. Liu J, Weller GER, Zern B, et al. Computational model for nanocarrier binding to endothelium validated using in vivo, in vitro, and atomic force microscopy experiments. Proc Natl Acad Sci USA. 2010;107(38):16530–16535. doi:10.1073/pnas.1006611107

204. Gao Z, Chen Y, Cai X, Xu R. Predict drug permeability to blood–brain-barrier from clinical phenotypes: drug side effects and drug indications. Bioinformatics. 2017;33(6):901–908. doi:10.1093/bioinformatics/btw713

205. Yaari Z, Da Silva D, Zinger A, et al. Theranostic barcoded nanoparticles for personalized cancer medicine. Nat Commun. 2016;7(1):13325. doi:10.1038/ncomms13325

206. Dahlman JE, Kauffman KJ, Xing Y, et al. Barcoded nanoparticles for high throughput in vivo discovery of targeted therapeutics. Proc Natl Acad Sci USA. 2017;114(8):2060–2065. doi:10.1073/pnas.1620874114

207. Oladele OT. Nanomaterials Characterization Using Hybrid Genetic Algorithm Based Support Vector Machines. IJMSE. doi:10.12720/ijmse.2.2.107-114

208. Liu J, Hopfinger AJ. Identification of Possible Sources of Nanotoxicity from Carbon Nanotubes Inserted into Membrane Bilayers Using Membrane Interaction Quantitative Structure−Activity Relationship Analysis. Chem Res Toxicol. 2008;21(2):459–466. doi:10.1021/tx700392b

209. Soltani M, Moradi Kashkooli F, Souri M, et al. Enhancing Clinical Translation of Cancer Using Nanoinformatics. Cancers. 2021;13(10):2481. doi:10.3390/cancers13102481

210. Weerarathna IN, Kamble AR, Luharia A. Artificial Intelligence Applications for Biomedical Cancer Research: a Review. Cureus. doi:10.7759/cureus.48307

211. Bajpai S, Shreyash N, Sonker M, Gupta V, Tiwary SK, Biswas S. Concept of Artificial Intelligence in Discovering and Re-Purposing of Drugs. Med Pharm. 2021. doi:10.20944/preprints202105.0726.v1

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.