Next Article in Journal
Thermoresponsive Azithromycin-Loaded Niosome Gel Based on Poloxamer 407 and Hyaluronic Interactions for Periodontitis Treatment
Next Article in Special Issue
Disialoganglioside GD2-Targeted Near-Infrared Photoimmunotherapy (NIR-PIT) in Tumors of Neuroectodermal Origin
Previous Article in Journal
Methylene Blue-Based Combination Therapy with Amodiaquine Prevents Severe Malaria in an Experimental Rodent Model
Previous Article in Special Issue
Nanoparticle-Based Drug Delivery Systems Targeting Tumor Microenvironment for Cancer Immunotherapy Resistance: Current Advances and Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanoparticle-Based Therapeutic Strategies for Enhanced Pancreatic Ductal Adenocarcinoma Immunotherapy

1
Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Chengdu 610017, China
2
Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610017, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2022, 14(10), 2033; https://doi.org/10.3390/pharmaceutics14102033
Submission received: 22 July 2022 / Revised: 13 September 2022 / Accepted: 19 September 2022 / Published: 24 September 2022
(This article belongs to the Special Issue Drug Repurposing and Delivery Systems for Immunotherapy)

Abstract

:
Immunotherapy has dramatically changed prognosis for patients with malignant tumors. However, as a non-immunogenic tumor, pancreatic ductal adenocarcinoma (PDAC) has a low response to immunotherapy. Factors that contribute to the inefficiency of PDAC immunotherapy include the tumor microenvironment (TME) and its dense stroma, which acts as a barrier for drug delivery and immune cell infiltration. Recent studies have shown that nanoparticle-based therapeutic strategies have more promising applications in improving drug delivery and reversing the immunosuppressive TME for PDAC. Therefore, nanomaterial-based therapeutic approaches are expected to enhance the effectiveness of immunotherapy and improve prognosis of patients with PDAC. Here, we outline the status and dilemma of PDAC immunotherapy, and summarize the latest advances in nanoparticle-based treatment strategies to enhance the efficacy of PDAC immunotherapy.

1. Introduction

Pancreatic cancer is a lethal tumor with an increasing incidence. With the advancements in tumor diagnosis and therapy, several malignant tumor-associated mortality rates have decreased; however, the pancreatic-cancer-associated mortality rate remains high. The reasons behind the high mortality rates for pancreatic cancer include difficulty in achieving early diagnosis and lack of effective treatment options [1]. Pancreatic ductal adenocarcinoma (PDAC) is the main type of pathological pancreatic cancer, accounting over 90% of all pancreatic cancers. Moreover, it has the worst response to treatment and highest mortality rate of all pathological types of pancreatic cancer [2].
Radical resection is a relatively effective treatment option for patients with PDAC. However, the lack of specific clinical symptoms leads to late diagnosis and dismal prognosis. Over 50% of patients presenting with liver or peritoneal metastases are unresectable at the time of diagnosis [3]. Even for patients who have their tumor lesions removed, recurrence is difficult to avoid due to the presence of microscopic lesions. Hence, following surgery, patients require adjuvant treatment. The current therapies have a limited effect on the survival of patients with PDAC [4]. The main treatment methods include chemotherapy, radiotherapy, molecular targeted therapy, and immunotherapy. Of these, chemotherapy is the most effective. Standard chemotherapy regimens for PDAC present modest survival benefits, with resistance ultimately unavoidable [1]. Even for patients with advanced and distant metastatic PDAC, who have received the standard chemotherapy, the five-year survival rate is still less than 5% [4]. Thus, developing more effective treatment options is important for improving the prognosis and survival of patients with PDAC.
Immunotherapy, especially immune checkpoint inhibitors (ICIs), has shown remarkable efficacy in many malignant tumors; however, the current immunotherapy response is dismal in patients with PDAC [5]. The reason for this is complex. High desmoplasia plays an important role in the poor immunotherapy efficacy for PDAC. The PDAC stroma accounts for over 80% of tumor mass. The tumor comprises various cells, including cancer-associated fibroblasts (CAFs), pancreatic stellate cells (PSCs), tumor cells, immune cells, and endothelial cells, that secrete scores of cytokines and induce the formation of the extracellular matrix (ECM). This dense stroma acts as a “physical barrier” to obstruct the antitumor agent delivery and as an immunosuppressive biological barrier to limit immune active cell infiltration [6]. Stromal cells can also induce immunosuppressive tumor cells [7]. Moreover, the infiltration of immunosuppressive cells, including regulatory T (Treg) cells, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), was reportedly rich, while immunosupportive cells, such as intratumoral CD8+ T cells, were relatively scarce in the PDAC tumor microenvironment [8,9]. Additionally, PDAC is a hypovascular tumor accompanied by hypoxia and a high interstitial fluid pressure (IFP) in the tumor microenvironment (TME), which impairs drug delivery and leads to immunosuppression [10].
The current immunotherapeutic approaches have shown a limited improvement in the prognosis for patients with PDAC [11]. However, several studies reported that a positive response to the therapy and improved prognosis in patients with PDAC are associated with immune activation [12,13,14,15,16]. Therefore, immunotherapy remains a promising treatment option for patients with PDAC. New therapeutic strategies for improving effectiveness of immunotherapy for PDAC include combining existing strategies and exploring new therapeutic targets [17,18,19]. Multiple molecules (including non-coding RNAs) have been identified as the potential research targets to enhance the immunotherapy efficacy for PDAC [20,21]. However, delivery of these small molecules or co-delivery of antitumor agents to the tumor’s interior has become an important obstacle to overcome. Additionally, improved approaches are required to enable the drugs to cross the dense stroma of PDAC to reach the deeper part of the tumor. Recently, nanoparticles have shown promise as carriers for antitumor agent delivery. Complex molecules, such as non-coding RNAs (e.g., siRNA and microRNA), can also be delivered using nanoparticles as the carriers [22,23]. The advantages of these nanoparticles include controllable size and drug release, prolonged circulation, reduced side effects, co-delivery of multiple therapeutic agents, and site-specific drug delivery [23]. Two nano-formulations have been approved by the U.S. Food and Drug Administration (FDA) for PDAC therapy. The first is an albumin-bound paclitaxel called nab-paclitaxel. Compared to the traditional paclitaxel, nab-paclitaxel shows a higher plasma clearance, better distribution, lower toxicity, and better therapeutic results [24]. Based on results from a phase 3 MPACT trial, the FDA approved nab-paclitaxel as a delivery agent for the chemotherapy drug gemcitabine as the first-line treatment for metastatic PDAC [25]. Another nano-formulation approved by the FDA for PDAC treatment is the liposomal formulation of irinotecan. Liposomal irinotecan plus 5-fluorouracil (5-FU) and leucovorin was approved by the FDA and used as second-line treatment option for metastatic patients with PDAC who were resistant to gemcitabine. Compared to free irinotecan, the liposomal formulations of irinotecan presented lower toxicity and better therapeutic results [26].
Several preclinical studies have presented data on the benefits of nanoparticle-based therapeutic strategies for PDAC immunotherapy. Nanoparticles can remodel the tumor stroma, improve drug delivery efficiency, regulate the immunosuppressive microenvironment, activate antitumor immunity, and induce a stronger antitumor effect [27,28]. In this review, we summarize the current status of PDAC immunotherapy and discuss obstacles in PDAC research. Additionally, we outline the potential role of nanoparticle-based therapeutic strategies in improving drug delivery and regulating immunosuppressive TME of PDAC.

2. Status and Dilemma of Immunotherapy for PDAC

Tumor immunotherapy involves killing tumor cells by activating the immune response of the body. The current immunotherapy approaches include cancer vaccines, ICIs, adoptive cell transfer (ACT), monoclonal antibodies, and cytokine-mediated therapies [29]. Of these therapeutic modalities, ICIs are currently the only immunotherapy strategy approved for clinical use in multiple solid tumors (e.g., non-small cell lung cancer, melanoma, renal cell cancer, and colorectal cancer) [30]. The main targets of the ICIs include cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein-1 (PD-1), and programmed cell death ligand-1 (PD-L1). Monoclonal antibodies, including anti-PD-L1 inhibitors (e.g., atezolizumab, durvalumab, and avelumab), anti-PD-1 inhibitors (e.g., pembrolizumab and nivolumab), and CTLA4 receptor inhibitors (e.g., ipilimumab and tremelimumab), have been approved by the FDA for several solid tumor treatments [30,31]. ICI monoclonal antibodies, as single drugs or combination strategies, were studied in PDAC clinical trials [32,33,34,35,36,37,38,39,40,41,42,43]. Other immunotherapeutic modalities (e.g., cancer vaccines, ACT, and cytokine-mediated therapies) were also studied in pancreatic cancer clinical trials; however, their clinical application remains to be demonstrated [44,45,46]. This section summarizes the key clinical trial results for PDAC immunotherapy (especially ICI and ICI combined with chemotherapy/radiotherapy) and outlines the influence of the tumor immune microenvironment (TIME) on the efficacy of PDAC immunotherapy.

2.1. Status of Immunotherapy for PDAC

Reported clinical trials on the immunotherapy strategies for PDAC include ICIs, cancer vaccines, adoptive cellular immunotherapy (ACI), and cytokine-mediated therapies. The results for key clinical trials on PDAC immunotherapy are summarized in Table 1 and Supplementary Table S1.
Studies on ICIs and their use in a combined strategy account for the majority of the PDAC clinical research. ICIs release the inhibitory brakes in T cells, which strongly activates the immune system and induces antitumor immune reactions [30,31]. The first reported ICI in a PDAC clinical trial was ipilimumab, a CTLA-4 inhibitor. In this phase 2 clinical trial, 27 patients with locally advanced or metastatic PDAC were treated with the ipilimumab for two cycles. Although one case with a delayed reaction was reported, no objective response was reported in this study [32]. In 2012, the safety and activity of anti-PD-L1 antibody BMS-936559 were evaluated in patients with advanced cancer. In this multicenter phase 1 trial, 14 patients with PDAC were recruited and treated with the BMS-936559. Similar to the ipilimumab trial result, no objective response was observed in patients with PDAC after the anti-PD-L1 inhibitor treatment [33]. These results indicate that PDAC does not respond well to ICI monotherapy. By 2020, KEYNOTE-158 showed that patients with solid tumors with high microsatellite instability (MSI-H)/mismatch repair-deficient (dMMR) can benefit from anti-PD-L1 monotherapy. Here, the overall response rate (ORR) of 22 patients with PDAC with MSI-H was 18.2%. This led the FDA to approve pembrolizumab for treatment in patients with solid tumors with MSI-H/dMMR, including PDAC [34]. However, it is worth noting that the ORR for PDAC was much lower than that of other tumors in the study, and only a very small proportion of patients with PDAC (below 2%) had MSI-H [35]. Efficacy of the dual ICI combination therapy was also poor in patients with PDAC. In a phase II clinical study, 65 metastatic patients with PDAC received combination therapy with the anti-PD-L1 antibody durvalumab and anti-CTLA-4 antibody tremelimumab. This combination treatment did not improve the prognosis of patients with PDAC. Moreover, its adverse effects increased significantly [36].
Following the failure of single- and double-ICI combination therapies, researchers started exploring whether combination therapy strategies could improve the efficacy of PDAC immunotherapy. It was suggested that chemotherapy agents may boost the immune response and potentially overcome PDAC resistance to ICIs [37]. Therefore, a combination therapy strategy of chemotherapy with ICI was attempted in patients with PDAC. Gemcitabine combined with anti-CTLA-4 antibodies (i.e., tremelimumab and ipilimumab) was evaluated in two phase I trials. In one clinical trial, tremelimumab combined with gemcitabine was used to treat metastatic PDAC. The median overall survival (OS) was 7.4 months. At the end of the treatment, 2 of the 34 patients achieved partial response (PR) [38]. In the second trial, ipilimumab was combined with gemcitabine for the treatment of advanced PDAC. The first reported response (PR and stable disease (SD)) was at 43% [39]. The final result from this study was reported in 2020 with an ORR of 14%. The median progression-free survival (PFS) and median OS were 2.78 and 6.90 months, respectively [19]. Gemcitabine combined with anti-PD-1 and anti-PD-L1 inhibitors has also been administered to patients with advanced PDAC. A phase Ib/II trial reported on the use of the anti-PD-1 inhibitor pembrolizumab, in combination with gemcitabine and nab-paclitaxel, for treatment of advanced solid tumors. Among the PDAC arms, disease control rates (PR and SD) were at 100%. The median PFS and OS were 9.1 and 15.0 months, respectively [40]. In another phase I study, the anti-PD-1 inhibitor nivolumab was combined with nab-paclitaxel and gemcitabine as treatment for patients with advanced pancreatic cancer. A response rate was observed in 18% of the patients. The median PFS and OS were 5.5 and 9.9 months, respectively [41]. Recently, a randomized phase III trial tested a new PD-1 antibody called sintilimab, in combination with modified FOLFIRINOX versus FOLFIRINOX alone as the first- or second-line therapy for metastatic and recurrent PDAC. In this study, an improved ORR (50% vs. 23.9%) was observed in the sintilimab and modified FOLFIRINOX combination groups, but OS and PFS did not improve [42]. Additionally, the results of a phase II study showed that there was no significant difference in the OS and PFS between the dual ICI and chemotherapy combined groups and chemotherapy alone [43]. These results suggest that, although the combination of chemotherapy and immunotherapy slightly improved the response rate in patients with PDAC relative to immunotherapy alone, there was still no significant improvement in their OS.
Radiation therapy presents another promising strategy for enhancing the therapeutic effects of PDAC immunotherapy. Radiation therapy may increase the response to immunotherapy through an abscopal effect [47]. In a preclinical study, radiation promoted the release of tumor-specific antigens and enhanced priming of tumor-specific T cells in PDAC mouse models [48]. However, the current clinical trial data show that the combination of radiotherapy and ICI only has moderate therapeutic benefits for patients with PDAC, and does not improve their survival [49,50,51].
Other monotherapies and combination therapies were widely explored in PDAC clinical trials (e.g., tumor vaccines, oncolytic viruses, ACI, and monoclonal antibodies; Supplementary Table S1); however, these trials only reported slight benefits in the response rates in patients with PDAC. Overall, the existing immunotherapy strategies did not significantly improve survival of patients with PDAC, and were limited by the small number of participants; hence, these therapies need to be tested within a larger subject population.
Table 1. Published clinical trials of ICIs in PDAC.
Table 1. Published clinical trials of ICIs in PDAC.
Therapeutic StrategyPhaseStageNumber of PatientsObjective Response RateMedian PFS (Months)Median OS (Months)Immune-Related Adverse Events (≥Grade 3)Publication YearNCT NumberRef.
Sigle ICIipilimumab (anti-CTLA-4)IIPre-treated
LAPC/mPDAC
270NANA11.1% (3/27)2010NCT00112580[32]
BMS-936559 (anti-PD-L1)Iadvanced PDAC140NANANA2012NCT00729664[33]
pembrolizumab (anti-PD-1)IIadvanced PDAC2218.2 (all dMMR patients)2.14NA2020NCT01876511[34]
Double ICIsdurvalumab (anti-PD-L1)
+ tremelimumab (anti-CTLA-4)
vs.
durvalumab
IImPDAC32 vs. 333.1% vs. 0%1.5 vs. 1.53.1 vs. 3.622% vs. 6%2019NCT02558894[36]
ICIs + chemotherapy tremelimumab (anti-CTLA-4) + GEMIchemotherapy-naive mPDAC342 PRNA7.4NA2014NCT00556023[38]
ipilimumab (anti-CTLA-4) + GEMIbadvanced PDAC1643% PR + SD2.58.5NA2016NCT01473940[39]
ipilimumab (anti-CTLA-4) + GEMIbadvanced PDAC2114%2.786.919%2020NCT01473940[19]
GEM + Nab-P + pembrolizumab (anti-PD-1)Ib/IImPDAC17100% PR + SD 9.11553%2018NCT02331251[40]
nivolumab (anti-PD-1) + GEM + Nab-P Iadvanced PDAC509%5.59.996%2020NCT02309177[41]
sintilimab (anti-PD-1) + FOLFIRINOX
vs.
FOLFIRINOX
IIImetastatic and recurrent PDAC55 vs. 5550% vs. 23.9%5.9 vs. 5.7310.9 vs. 10.85.7% (sintilimab + FOLFIRINOX)2022NCT03977272[42]
GEM + Nab-P + durvalumab (anti-PD-L1) + tremelimumab (anti-CTLA-4)
vs.
GEM + Nab-P
IImPDAC119 vs. 6130.3% vs. 23.0% 5.5 vs. 5.49.8 vs. 8.838% vs. 20%2020NCT02879318[43]
ICIs + radiotherapySBRT + nivolumab (anti-PD-1)
vs.
SBRT + nivolumab (anti-PD-1) + ipilimumab (anti-CTLA-4)
IIRefractory mPDAC41 vs. 43 clinical benefit rate 17.1% vs. 37.2%1.7 vs. 1.63.8 vs. 3.824.4% vs. 30.2%2022NCT02866383[49]
radiation + nivolumab (anti-PD-1) + ipilimumab (anti-CTLA-4)IImetastatic MSS PDAC2518%2.76.156%2021NCT03104439[50]
durvalumab (anti-PD-L1) + SBRT 8 Gy
vs.
durvalumab (anti-PD-L1) + SBRT 25 Gy
vs.
durvalumab (anti-PD-L1)/tremelimumab(anti-CTLA-4) + SBRT 8 Gy
vs.
durvalumab (anti-PD-L1)/tremelimumab(anti-CTLA-4) + SBRT 25 Gy
ImPDAC14 vs. 10 vs. 19 vs. 165.1%1.7 vs. 2.5 vs. 0.9 vs. 2.33.3 vs. 9.0 vs. 2.1 vs. 4.27.1% vs. 33.3% vs. 21.1% vs. 62.5%2020NCT02311361[51]
GEM: gemcitabine; Nab-P: nab-paclitaxel; LAPC: locally advanced pancreatic cancer; mPDAC: metastatic pancreatic ductal adenocarcinoma; SBRT: stereotactic body radiation therapy; NA: not available.

2.2. Dilemma in Immunotherapy for PDAC

The difficulty in immunotherapy for PDAC is mainly due to its unique TIME (Figure 1). Tumor, immune, and stromal cells, and extracellular components constitute the TIME. The TIME is crucial for the induction and maintenance of malignant tumor phenotypes. Moreover, it can directly determine the patient prognosis and response to immunotherapy [52]. According to the infiltration of immune cells in the TIME, tumors can be classified as “hot” (immunologically active) and “cold” (immunologically inactive). “Hot tumors” are characterized by an abundant immune infiltration and a better response to immunotherapy. PDAC is generally considered a “cold tumor” with poor immunogenicity [53]. Abundant immunosuppressive cells exist in the TIME of the PDAC, whereas immunosupportive cells (e.g., CD8+ T cells) are limited. The immunosuppressive cells in the TIME of PDAC include MDSCs, TAMs, and Tregs. These cells secrete cytokines to sustain the immunosuppressive TIME in PDAC cells. Moreover, PDAC has a low mutational burden, resulting in low neoantigen levels and a reduced ability in T cells to recognize tumors [54]. Therefore, successful induction and activation of effector T cells in the TIME are required for an effective PDAC antitumor immune response [55].
PDAC is characterized by its abundant desmoplastic stroma that contains a variety of cellular components. These components include immune cells, PSCs, CAFs, and endothelial cells, with CAFs representing the main cells in the PDAC stroma [6]. However, the origin and functional characteristics of CAFs remains unclear. PSCs are generally considered the main source of CAFs [56]. Heterogeneous CAFs play a variety of roles in the modeling of TIME for the PDAC. For example, myofibroblastic CAFs can induce a dense fibrous tissue formation to create a barrier that protects cancer cells from being recognized by the immune system. Inflammatory CAFs can produce cytokines that induce TAM differentiation and recruit immunosuppressive cells, such as Tregs and MDSCs [57]. Together, the cellular and ECM form an immunosuppressive biological barrier, which acts as a drug delivery barrier. Breaking this barrier would vastly improve drug delivery, and consequently, improve the immune response of PDAC. However, the tumor stroma is an important barrier that limits the tumor metastasis [58]. An indiscriminate targeting of the stroma may promote PDAC invasion [59]. Thus, rational stromal remodeling would be an ideal research direction for improving PDAC immunotherapy.
Additionally, PDAC is characterized as a hypovascular tumor. The hypovascularity and poor perfusion of PDAC not only limits the delivery of antitumor agents, but also induces hypoxia in the TIME [10]. Hypoxia is another important obstacle for PDAC immunotherapy. Hypoxia can induce and maintain an immunosuppressive TME and activate PSCs to mediate fibrosis [60]. Therapeutic strategies targeting optimized hypoxia have shown synergistic improvements in immunotherapy in the preclinical studies for pancreatic cancer [61,62]. In a phase I clinical trial, evofosfamide (prodrug that alleviates hypoxia) combined with ipilimumab was tested in advanced solid malignancies. Here, seven patients with pancreatic cancer were enrolled, and two achieved SD [63]. Further clinical trials are required to confirm this result. Tumor vascular normalization is an emerging strategy for alleviating hypoxia and enhancing cancer immunotherapy [64]. Some studies proposed strategies to normalize tumor vasculature in pancreatic cancer, and their potential role in PDAC immunotherapy was proven in preclinical models [65,66]. These results are yet to be demonstrated in clinical trials.
Overall, the specific immunosuppressive TIME, high levels of desmoplasia, hypovascularity, and extreme hypoxia are typical pathological features of PDAC, and are some of the main reasons for its low response to immunotherapy. A more comprehensive investigation of the PDAC TIME is needed to find the effective immunotherapy targets.

3. Nanoparticle-Based Therapeutic Strategies for Enhanced PDAC Immunotherapy

Two key problems hinder the therapeutic effects of PDAC immunotherapy: the inadequate response caused by the immunosuppressive TIME and the delivery obstacle presented by the dense tumor stroma and hypovascularization [8]. Recent studies show that nanoparticles could help solve these problems. Nanoparticles can not only be applied to regulate the immune system, but, more importantly, can enable, modulate, and penetrate the dense stroma of PDAC [23,27]. For example, nano-albumin-bound (Nab)-paclitaxel, a nano-based drug commonly used in PDAC treatment, was proven to deplete the tumor stroma through the interaction between the albumin and secreted protein acid, and is rich in cysteine [67]. Additionally, the size of the nanoparticles is adjustable. Small-sized nanoparticles can pass through the tumor stroma where molecular drugs cannot penetrate, which allows nanoparticles to co-deliver stroma depletion and anti-tumor/immunomodulatory agents [68]. Recently, the use of nano drug delivery devices coupled with stroma depletion has emerged as a promising treatment for PDAC [69]. Furthermore, several studies have tested nanoparticle-based photothermal therapy (PTT), photodynamic therapy (PDT), chemodynamic therapy (CDT), and sonodynamic therapy (SDT) for treatment of PDAC, and the results were promising in the preclinical models [70,71,72,73,74,75,76,77,78]. Thus, nanoparticle-based therapeutic strategies have provided new avenues for the PDAC immunotherapy. Here, we summarize the reported nanoparticle-based therapeutic strategies for the PDAC immunotherapy (Table 2) and discuss their prospects and challenges.

3.1. Nanoparticle-Based Immunogenic Cell Death Strategies for Enhanced PDAC Immunotherapy

Immunogenic cell death (ICD) is a specific type of cancer cell death. When the tumor cells are stimulated by external stimuli, the dying tumor cells release damage-associated molecular patterns (DAMPs) that activate tumor-specific immune responses and boost antitumor responses. Additionally, ICD induces the continuous release of tumor antigens, which can help turn cold tumors into hot tumors. Moreover, ICD leads to the activation and recruitment of cytotoxic T cell lymphocytes (CTLs) [108]. Therefore, ICDs could work synergistically with ICIs to increase the patient immune response rates. Some chemotherapeutic drugs, oncolytic viruses, physicochemical therapies, photodynamic therapies, and radiotherapies can induce ICD [108]. Nanomedicines can reportedly enhance the ICD-inducible agents to exert stronger immune effects, owing to their proven advantages in drug delivery. This is especially suitable for solid tumors with poor drug delivery, such as PDAC [109].
Oxaliplatin is a commonly used chemotherapeutic agent for PDAC treatment and is an ICD-inducible agent [110]. Compared to free oxaliplatin treatment, pancreatic tumor cells treated with nanoparticles encapsulating oxaliplatin release more DAMPs and induce stronger dendritic cell immune responses and a higher percentage of tumor-infiltrating activated cytotoxic T lymphocytes [79]. Additionally, nanoparticle capsules can help reduce the oxaliplatin toxicity. Liu et al. developed a mesoporous silica nanoparticle (MSNP)-based platform for the high-dose loading of platinum chemotherapeutic agents. Using an orthotopic Kras-derived PDAC model, they proved that MSNP not only improved the intratumoral administration and pharmacokinetics of the oxaliplatin, but also maintained stability of the colloid after intravenous injection and reduced its toxicity [80]. Nanoparticles can also realize an effective transportation of molecular entities with antitumor and ICD effects. Shen et al. proposed a sequential receptor-mediated mixed-charge targeted drug delivery system to administer effective components from the Euphorbia plant for the treatment of pancreatic cancer. Here, the modified nanomicelles induced ICD and penetrated the tumor vessel walls [81]. Therefore, nanoparticle-based therapy strategies could maximize the role of the ICD drugs, enhance their immune enhancement and antitumor effects, and have a potential application value in PDAC therapy.

3.2. Nanoparticle-Based Immune Modulation Strategies for Enhanced PDAC Immunotherapy

Regulation of the immune microenvironment is key to improving the effectiveness of PDAC immunotherapy. The TIME of the PDAC is frequently characterized by a low number of tumor-infiltrating lymphocytes (TILs) and a high number of Tregs, M2 TAMs, and MDSCs. The latter are related to the poor immune response for PDAC. Repolarization of the TAM phenotype from M2 to M1 is reportedly a promising strategy for PDAC treatment [111]. miR-125b can affect TAM repolarization. Parayath et al. conjugated the M2 peptides to hyaluronic acid-polyethyleneglycol (HA-PEG)/HA-polyethylenimine (HA-PEI) polymers to form self-assembled nanoparticles with miR-125b. This delivery system has a dual functionality: the HA polymer can target CD44 receptors on the surface of macrophages, and the M2 peptide can specifically target TAMs. The synthesized nanoparticles can effectively reprogram the M2 TAMs to the antitumoral M1 phenotype [82]. Moreover, PI3K-γ and colony-stimulating factor-1 (CSF-1) or CSF-1 receptor (CSF-1R) pathways are also involved in the infiltration and polarization of the M2 TAMs in pancreatic cancer [112]. Li et al. developed nanomicelles that co-deliver a PI3K-γ inhibitor and CSF-1R-siRNA. They proved that this nanomicelle not only reduced the M2 TAMs level and increased the M1 TAMs level, but also inhibited the tumor infiltration of MDSCs and effectively modulated the TIME of PDAC [83]. Cao et al. developed a nanoplatform that can co-deliver monoacylglycerol lipase siRNA (suppresses free fatty acid generation) and CB-2 siRNA (regulates macrophage phenotype). This nanoplatform can inhibit production of free fatty acids in the PDAC tumor cells and cut off nutritional supply to the tumor. Additionally, it can repolarize the TAMs into the tumor-inhibiting M1-like phenotype, reverse the immunosuppressive microenvironment, and produce synergistic antitumor effects [84]. MDSCs are another promising target for PDAC treatment [113]. However, therapeutic abrogation of MDSCs usually causes a compensatory recruitment; therefore, it cannot effectively improve the immunosuppressive TIME of PDAC [114]. Reportedly, inhibiting MDSC recruitment may weaken the immunosuppression [115]. Based on low-molecular-weight heparin (LMWH) inhibiting MDSC recruitment, Lu et al. developed LMWH-based nanoparticles loaded with paclitaxel. These nanoparticles not only improved the pancreatic TIME, but also inhibited spontaneous metastasis of the tumors [85]. In addition to the immunosuppressive cells, dysfunction and immunosuppressive antigen-presenting cells (APCs) are abundant in the immunosuppressive TIME of PDAC, which is also related to its low response to immunotherapy. Lorkowski et al. developed an immunostimulatory nanoparticle (immuno-NP), and co-loaded two immune agonists. The first agonist was a stimulator for the interferon pathway, and the second was an agonist for the Toll-like receptor 4 pathway. The combined introduction of these two agonists proved that immuno-NP can trigger robust activation and expansion of the APCs, and strengthen the immune response in PDAC [86].
Targeting immunomodulatory cytokines can also regulate the TIME. Hence, nanoparticle delivery of these cytokines can potentially achieve a more effective immunomodulation. Miao and Shen separately designed nanoparticles that co-load chemokine C-X-C motif ligand 12 (CXCL12) traps with Interleukin-10 (IL-10) or PD-L1 traps to synergistically change the immunosuppressive microenvironment of PDAC and modify the immunosuppressive TIME to allow the host’s immune system to kill the tumor cells [87,88]. Moreover, retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) induce secretion of IFN-α/β and other pro-inflammatory cytokines [116]. Das et al. developed an NP-mediated delivery for 5′-triphosphate double-stranded RNA, which is specific to Bcl2 and can bind to RLRs. These therapeutic nanoparticles modulated the TIME and inhibited tumor growth in the PDAC model [89]. Systemic administration of immunomodulatory drugs rarely reaches the tumor-draining lymph nodes (TDLNs); however, this is overcome when using the nanoparticles because they effectively deliver drugs to deep tumors and TDLNs. Han et al. combined Interleukin-12 (IL-12) microspheres with stereotactic body radiotherapy (SBRT) to achieve repolarization of the TIME for pancreatic cancer. This biodegradable microsphere not only enables local continuous administration, but can also deeply penetrate the TDLN of PDAC, thus achieving complete tumor inhibition [90].
Using nanoparticles as carriers can protect physically or chemically unstable molecules from exerting immunotherapeutic effects. A previous study found that the RNAi-based drugs have a high sequence specificity to target molecules and are considered more effective than antibodies for blocking immune checkpoints [117]. However, RNA is easily attacked by RNase in circulation. Jung et al. proposed the use of poly-based siRNA nanoparticles to target PD-L1. They showed that the modified PD-L1 siRNA nanodrug had better targeting than the traditional PD-L1 antibodies. This siPD-L1 nanoparticle not only showed an effective knockout of PD-L1 in the cancer cells, but also showed a strong antitumor immunity in a PDAC preclinical model [91]. Recently, microcapsules for oxygen delivery were proposed. Here, the oxygen microcapsules provided a stable oxygen delivery deep into the tumor, reversed hypoxia in the TME, and improved the performance of ICB in PDAC [62].
Nanoparticles can also be used as immune adjuvants in vaccines. Dong et al. used aluminum hydroxide nanoparticles together with PEI as an immune adjuvant for dendritic cell (DC) vaccines. This nano-adjuvant could enhance antigen transport and cross-presentation of DCs in a pancreatic cancer model, thus further improving the DC vaccine efficacy [92].

3.3. Nanoparticle-Based Chemoimmunotherapy for Enhanced PDAC Immunotherapy

Chemotherapy and immunotherapy can synergistically exert antitumor effects. Some chemotherapy drugs can induce ICD, which can then be enhanced in combination with ICIs or other immunomodulators. Therefore, the chemoimmunotherapy combination is rational [118]. However, during clinical studies, chemotherapy combined with ICI only presented a modest efficacy and did not improve prognosis for patients with PDAC [37,38,39,40,41,42,43]. This may be related to the inherently strong immunosuppressive TIME, lack of new antigens with low ICI response, or the dense stroma of PDAC. Nanoparticles can enhance the ICD of chemotherapeutic drugs, be used as immunomodulators, and act as carriers for co-delivery of immunomodulators and chemotherapeutic agents to achieve effective drug delivery to deep layers of the tumors [27,28]. Thus, nanoparticle-based chemoimmunotherapy presents a promising approach for enhancing the efficacy of PDAC immunotherapy.
Lu et al. first used MSNP to co-deliver oxaliplatin and an indoleamine 2,3-dioxygenase (IDO) inhibitor to treat PDAC. IDO is an enzyme that induces immunosuppressive activity. Therefore, IDO inhibitors can reverse the immunosuppressive TME. Additionally, oxaliplatin can induce ICD. Nanocarriers can improve the drug delivery and, consequently, the intratumoral concentration of IDO inhibitors and oxaliplatin. This synergistic approach boosted the antitumor immune response in a PDAC mouse model [93]. Based on a similar combination, Huang et al. developed cationic lipid-assisted polymeric nanoparticles to deliver the IDO1 siRNA. Using colorectal and pancreatic cancer mouse models, they proved that simultaneous administration of oxaliplatin and siIDO1 nanoparticles could achieve synergistic antitumor effects by promoting maturation of DCs, increasing TILs, and reducing the number of Tregs, which could then prevent tumor recurrence by provoking long-term antitumor immune memory [94].
Gemcitabine is a chemotherapeutic drug commonly used for PDAC treatment. Although gemcitabine cannot induce ICD [119], when modified with nanoparticles, it can be employed as a tumor-penetrating nanocarrier. In a previous study, a redox-responsive gemcitabine-conjugated polymer was used as the nanocarrier to co-load an IDO1 inhibitor (NLG919) and paclitaxel. The co-loaded micelles deeply penetrated the pancreatic tumor to induce immune-active antitumor activity in PDAC models [95]. Tong et al. developed a tumor-pH-sensitive polymer whose cavity was a hydrophobic gemcitabine prodrug. Nanopolymers can respond to the pH of the TME and transform into small particles to promote the in-depth delivery of gemcitabine. Additionally, these nanoparticles can modulate the TME, upregulate PD-L1 expression levels in tumor cells, and act synergistically with anti-PD-1 therapy [96]. Chen et al. designed TME-activatable charge-conversional micelles to co-load two prodrugs, GEM-C18 and NI-HJC0152. Once the lowered pH of the outer layer of PDAC is sensed, the micelle surface charge changes from negative to positive, releasing the prodrugs across the rich stroma of the PDAC. Additionally, NI-HJC0152 responds to hypoxia and releases HJC0152, inhibiting signal transducer and activator of transcription 3 (STAT3) and inducing immune activation. Thus, micelles play a dual role by reversing both the immunosuppressive TME and drug resistance in PDAC [97]. In addition to the pH response, special nanoparticle modifications can achieve effective delivery of antitumor drugs deep into the tumor. Hu et al. developed a negatively charged HA, which can achieve graded nanostructures and effectively deliver celastrol (a pentacyclic triterpenoid extracted from traditional Chinese medicine) and 1-methyltryptophan (IDO inhibitor) to a deep tumor site of the pancreas. The combination of celastrol with an IDO inhibitor, administered using nanoparticles, showed significantly enhanced tumor inhibition and downregulation of the immunosuppressive TIME [98]. Liposomal irinotecan is a nano-formulation currently approved for PDAC treatment. Liu et al. encapsulated irinotecan in a silicasome instead of a liposome and proved that it could induce a more powerful ICD response. Additionally, when combined with anti-PD-1 treatment, it led to a more pronounced survival improvement compared to anti-PD-1 combination therapy with free or liposomal irinotecan. Moreover, silicasome irinotecan is less leaky and toxic than liposomal irinotecan [99].
Targeting TAMs combined with chemotherapeutic drugs is another promising strategy for PDAC chemoimmunotherapy. Zhou et al. developed an exosome-based dual delivery biosystem for the galectin-9 siRNA and oxaliplatin prodrugs. Disruption of the galectin-9/dectin1 axis could reverse the PDAC immunosuppression of M2-TAMs, whereas oxaliplatin could trigger ICD. Thus, use of this biological material not only significantly increases accumulation of the drugs at the tumor site, but also enhances antitumor immunity in PDAC [22]. Similarly, Wang et al. developed a biomimetic dual-targeting nanomedicine based on gemcitabine and M2pep. Nano-formation enables the simultaneous targeted delivery of gemcitabine to the pancreatic tumor sites and TAMs repolarization to potentiate its therapeutic effects. This nano-formation also synergistically enhances the antitumor effect of PD-L1 antibodies in the PDAC [100].
These studies suggest that nanoparticles may not only serve as vehicles to improve the permeability and immunogenicity of existing chemotherapeutic agents, but, more importantly, may provide a flexible carrier for multiple chemoimmunotherapy combination options. Nanoparticle-based chemoimmunotherapy offers more possibilities for enhancing the efficacy of immunotherapy for PDAC.

3.4. Nanoparticle-Based Stroma Modulation for Enhanced PDAC Immunotherapy

The dense stroma in PDAC is a major obstacle for immunotherapy. It hinders the delivery of immunotherapeutic agents and infiltration of effector T lymphocytes. However, the dense stroma in PDAC is associated with the formation of an immunosuppressive TME (e.g., stromal cells like CAFs and PSCs), which can induce and recruit immunosuppressive lymphocytes as well as secrete immunosuppressive cytokines [120]. The ECM is the main non-cellular component of the PDAC stroma and is an important obstacle that hinders an effective drug delivery and effector T lymphocyte infiltration. The stroma cells and ECM are the main targets for PDAC tumor stroma regulation. Currently, stromal modulation combined with antitumor agents (e.g., chemotherapeutic or immunotherapeutic drugs) is regarded as an effective treatment option for PDAC management [121]. Several nanoparticle-based stoma modulation therapies have also been tested to enhance the PDAC immunotherapy.
Collagen and fibroblasts constitute the main components of the ECM and are the main targets for PDAC stroma modulation. Huang et al. proposed a nano-formation that co-loads an antifibrosis phosphate-modified α-mangostin (MP) and a plasmid encoding the immune-enhancing cytokine LIGHT (tumor necrosis factor superfamily 14, TNFSF14, CD258). In this nano-formation, called nanosapper, MP reversed the activation of CAFs, decreased collagen deposition, and relieved compressed vessels, while LIGHT normalized the tumor vessels, stimulated secretion of cytokines, recruited lymphocytes, and inhibited Tregs. Furthermore, this nanosapper can remodel TME and synergistically provide therapy with anti-PD-1 in PDAC models [101]. An acidic tumor extracellular pH-responsive clustered nanoparticle was developed to co-deliver a transforming growth factor-beta (TGF-β) receptor inhibitor and PD-L1 siRNA. The TGF-β receptor inhibitors can effectively inhibit activation of PSCs, reduce production of type I collagen, and remodel dense ECM. The siPD-L1 can silence the expression of PD-L1 in tumor cells. Therefore, this nano-formation can synergistically inhibit the growth of tumors in the PDAC model by activating the antitumor immune response [102]. The sonic hedgehog (SHH) signaling pathway plays an important role in the PDAC ECM synthesis. SHH signaling can activate the CAFs to produce a highly fibrotic stroma [122] and has been proposed as a promising target for the PDAC stroma remodeling [123]. However, SHH deletion reportedly caused tumors to become more aggressive in a PDAC mouse model [59]. Zhao et al. developed a polymeric micelle-based nano-formulation to co-deliver an SHH inhibitor and paclitaxel. This nano-formulation can increase the intratumoral vasculature density and promote CD8+ T cell infiltration through SHH inhibition, while paclitaxel can restrain tumor cell proliferation. They further proved that this nano-formulation could enhance the antitumor effect of anti-PD-1 monotherapy in a PDAC mouse model [103].
HA is another key component of the PDAC stroma and plays a role in increasing IFP, compressing tumor vessels, causing insufficient perfusion in the tumor, and hindering the delivery of antitumor agents [124]. PEGPH20 is a PEGylated recombinant human hyaluronidase that can reduce HA in the PDAC stroma [125]. PEGPH20 combined with chemotherapeutic agents showed promising antitumor effects in a PDAC preclinical model [126]. Blair et al. proposed a therapeutic strategy that combines PEGPH20 with GVAX (a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine), and proved that the single agent GVAX upregulates myeloid C-X-C chemokine receptor 4 (CXCR4) expression, while combined PEGPH20 modifying HA can decrease the CXCL12/CXCR4/chemokine receptor 7 (CCR7) immunosuppressive signaling axis, thus regulating myeloid cells, increasing T cell infiltration, and inducing memory effector T cells in PDAC. This combination can boost the antitumor immune response of GVAX [104]. Recently, researchers proposed a PEGPH20 combination with a focal adhesion kinase (FAK) inhibitor and an anti-PD-1 antibody to treat metastatic PDAC. In this combination strategy, PEGPH20 targets extracellular stromal HA, whereas FAK can regulate intracellular signaling in stromal and myeloid cells. The results proved that this dual stromal targeting combination can increase T cell infiltration and alter their phenotype towards effector memory T cells, thus sensitizing PDAC to anti-PD-1 therapy [105].
However, in clinical trials, PEGPH20 with chemotherapy did not improve the OS of patients with PDAC, and the incidence of adverse events was significantly higher in the combination therapy group [127,128]. Some researchers have proposed that the failure in PEGPH20 is related to the stroma, that it has protective effects in restraining PDAC tumor growth and progression, and that the stromal depletion may potentiate their metastatic capacity [129]. Thus, selective stroma modulation is the direction to enhance the efficacy of PDAC immunotherapy, instead of stroma deletion without discrimination. Chen et al. designed a nanoparticle co-loaded with paclitaxel and phosphorylated gemcitabine that selectively disrupted the central stroma and preserved the external stroma in a PDAC model. Thus, absence of the central stroma can effectively induce effector T lymphocyte infiltration, whereas the remaining external stroma can effectively prevent tumor metastasis. Additionally, these nanoparticles can modulate the immunosuppressive TIME of PDAC by augmenting the number of CTLs and restraining the percentage of Tregs [106]. Moreover, cancer–stroma interaction is an important factor in maintaining PDAC malignancy and affecting the PDAC response to immunotherapy [57]. Xie et al. designed cholesterol-modified polymeric CXCR4 antagonist nanoparticles for co-delivery of anti-miR-210 and siKRASG12D. This nano-formation has a triple-action for inhibiting PDAC: CXCR4 antagonists can block cancer–stroma interactions, anti-miR-210 can modulate the PDAC stroma through PSC inactivation, and siKRASG12D can kill pancreatic cancer cells. Since nanomedicines achieve a tumor-targeted delivery, mainly through the enhanced permeability and retention (EPR) effect following intravenous administration, this EPR effect is severely compromised in PDAC. Thus, they used an intraperitoneal administration to improve delivery and effective tumor penetration of nanoparticles. Combined therapy displayed an improved therapeutic effect in a PDAC model [107].

3.5. Nanoparticle-Based Photothermal/Photodynamic/Chemodynamic/Sonodynamic Therapy for Enhanced PDAC Immunotherapy

PTT converts light energy into heat energy to eliminate tumors [130]. PTT can induce ICD and elicit an immune response against cancer. Thus, PTT combined with immunotherapy presents a promising treatment for tumors. However, the dense stroma of PDAC impedes both laser penetration and the release of immune drugs deep into tumor tissue [130]. Yu et al. designed size-adjustable nanoparticles loaded with an immune checkpoint blocker, called BMS-202, to synergize with PTT. In this nanoplatform, the size-adjustable nanoparticles responded to fibroblast activation protein-α (specifically expressed on the fibrotic matrix) and a near-infrared (NIR) laser releasing small-sized nanoparticles. Small-sized nanoparticles loaded with BMS-202 were widely distributed within the tumor, and the immunosuppressive TME was alleviated. Meanwhile, the NIR laser-induced mild hyperthermia, reduced tumor hypoxia, and increased endogenous immune cell recruitment. Therefore, combination therapy inhibited not only tumor growth, but also tumor metastasis in the PDAC model [70]. Li et al. used a local tumor NIR photothermal immunotherapy to overcome the dense stroma of PDAC. They proposed supramolecular nanofibrils to co-load the clinically approved immunomodulatory thymopentin and photosensitizer indocyanine green for tumor photothermal immunotherapy. Using an orthotopic pancreatic tumor model, they showed that a local injection of the nanofibrils combined with NIR laser irradiation can effectively terminate tumor tissues while protecting normal tissues. Moreover, they demonstrated that this combination can promote proliferation and differentiation of antitumor immune cells, thus boosting tumor immunotherapy, while simultaneously eliminating tumor metastasis [71]. Sun et al. developed photosensitive dopamine-coated nanoparticles co-loaded with gemcitabine and NLG919 (IDO inhibitor) to construct nanoparticles for early- and late-stage metastatic cancer immunochemo-photothermal therapy. Using a PANC02 tumor-bearing mouse model, they found that the combination of the nanoparticles and laser irradiation enhanced the tumor inhibition effect. However, the abscopal effect of PTT combined with nanoparticles occurred mainly in late-stage tumor metastasis (large distal tumors), whereas in early-stage tumor metastasis (small distal tumors), no abscopal effect was observed [72]. Recently, Wang et al. proposed an interventional PTT (IPTT)-synergized immunotherapy guided by magnetic resonance imaging (MRI) for pancreatic cancer. They used amorphous iron oxide nanoparticles (IONs) loaded with photothermal agent’s indocyanine green (ICG) and immunoadjuvant imiquimod (IMQ) to construct a nanoplatform. This unique nanoplatform has multiple functions. First, it can serve as a contrast agent for the MRI, whereby an ION-assisted MRI is used to guide implementation of IPTT and monitor temperature distribution within the tumor and surrounding tissues during treatment, thus minimizing damage to surrounding healthy tissues. Second, it could act as a drug delivery carrier for IMQ and ICG, triggering a strong antitumor immunity. Third, it can act as a catalyst for the TME immune activation. Additionally, appropriately sized nanoparticles can penetrate the dense stroma in pancreatic cancer. Thus, IPTT-induced immune activation is synergistically enhanced by IMQ, while the IONs modulate the suppressive TME, thereby amplifying the antitumor immune effects. Together, they trigger a powerful antitumor systemic immunity to terminate both in situ tumors and metastasis [73].
PDT is another type of phototherapy that utilizes a specific wavelength to irradiate the tumor site, activate a photosensitive drug, and/or generate reactive oxygen species (ROS) to terminate the tumor. PDT was effective and promising in clinical trials for malignant tumors [131,132]. In phase I and I/II clinical trials, the safety of EUS-guided and verteporfin-PDT for locally advanced pancreatic cancer was confirmed [133,134]. Additionally, PDT can induce ICD and, consequently, be used in synergistic immunotherapy. However, there are still some obstacles in the use of PDT synergistic immunotherapy for PDAC. For example, PDT induces ICD while driving oxygen consumption and microvascular damage, further exacerbating hypoxia and glycolysis, which leads to lactate accumulation and aggravated immunosuppression in the TME. To solve this problem, Sun et al. designed HA-based supramolecular nanoparticles for co-delivery of photosensitizers, bromodomains, and extraterminal protein 4 inhibitors (BRD4i). In this nanosystem, BRD4i antagonizes the oncogene c-Myc and activate CTL, while HA contributes to long-term retention and deep tumor penetration for nanoparticles. They proved that this nanosystem combined with PDT could overcome the PDT-induced glycolysis and provoke a durable antitumor immunity in a pancreatic model [74]. Jang et al. used a tumor-derived reassembled exosome (R-Exo) as a delivery vehicle loaded with the photosensitizer chlorin e6 (Ce6-R-Exo). These modified exosomes retain their membrane proteins and have an average size, which allows for targeting of tumor and immune cells. The Ce6-R-Exo efficiently generated ROS-inducing cytokines that released under laser irradiation. Furthermore, Ce6-R-Exo can be captured by innate immune cells as antigens to boost immune responses, which can induce a long-term immune response and inhibit tumor growth, recurrence, and metastasis. Additionally, Ce6-R-Exo can be visualized through photoacoustic imaging. Therefore, Ce6-R-Exo provides a new strategy for an effective combination of PDT and immunotherapy [75].
SDT uses a low-intensity ultrasound that activates sonosensitizers to generate ROS and kill the tumor cells. Ultrasound penetrates deeper in the tissue than a laser; thus, SDT is more suitable for deep tumor treatment than the PDT/PTT [135]. Intratumoral oxygen is an important factor affecting SDT. The PDAC is an inherently hypoxic tumor. Hence, Nicholas et al. prepared pH-sensitive polymethacrylate-coated CaO2 nanoparticles, which could alleviate the tumor hypoxia. Here, Rose Bengal functioned as a sonosensitizer. A potent abscopal effect was observed in the SDT-treated PDAC animal models [76]. SDT can also induce ICD, thereby boosting immunotherapy. However, considering the dense stroma and poor blood perfusion in PDAC, the effectiveness of SDT may be limited. Chen et al. designed cavitation-assisted endoplasmic reticulum (ER)-targeted sonodynamic nanodroplets, which can achieve deep penetration as aided by ultrasound. Under the ultrasound irradiation, the modified sonosensitizer accumulates in the ER to generate large amounts of ROS in situ, which can further boost the efficacy of SDT. It was also demonstrated that the nanodroplets combined with SDT can enhance the effect of anti-PD-L1 immunotherapy in PDAC orthotopic and distant tumor models [77].
In addition to PDT/SDT, CDT is another ROS-mediated treatment method. However, unlike PDT/SDT, CDT causes damage to the tumor cells by converting ROS into highly toxic hydroxyl radicals (OH) through the Fenton reaction. Transition metal ions (i.e., Fe, Co, Ni, Cu, and Mn) are commonly used as reagents for CDT [136]. Chen et al. developed a tailored nanocomplex through the self-assembly of synthetic 4-(phosphonooxy) phenyl-2,4-dinitrobenzenesulfonate and Fe3+, followed by an HA modification to realize CDT for PDAC. By controlling the release of its components in a GSH-sensitive manner under the unique redox homeostasis of cancer cells and TAMs, the nanocomplex selectively triggers the Fenton reaction to induce oxidative damage in cancer cells, while repolarizing TAMs and deactivating stromal cells, thus attenuating stroma deposition. They further demonstrated that the CDT treatment provided a better tumor suppression than the conventional gemcitabine treatment, and did not show significant side effects in a PDAC mouse model; thus, CDT presents a promising strategy for PDAC [78].
In summary, PTT/PDT/SDT/CDT are promising strategies for cancer treatment because of their negligible toxicity and non-invasiveness. Additionally, these treatments can induce ICD, and thus can be used in combination with tumor immunotherapy. However, due to the insufficient penetration ability and special TME, traditional PTT/PDT/SDT/CDT has extremely limited effects in PDAC treatment. The application of nanoparticles facilitates use of these therapies for PDAC treatment. Nanoparticles can be designed for a deeper penetration or specific environmental responses, further exerting long-lasting antitumor effects. Furthermore, nanomedicine strategies could be designed to systematically co-deliver photosensitizer/sonosensitizer CDT agents and immunomodulators, thus boosting the immune-enhancing antitumor effects of these therapeutic strategies. These studies show that nanoparticle-based PDT/CDT/SDT/PTT has promising benefits in the PDAC immunotherapy and can be an important research direction for PDAC.

3.6. Nanoparticle-Based Imaging and Theranostic Agents for PDAC Immunotherapy

Nanoparticles also shows promise as imaging and theranostic agents [137]. For example, labelling of human mesenchymal stem cells with gold-poly-L-lysine nanocomplexes can achieve computed tomography (CT) in vivo tracking [138]. Iron oxide nanoparticles (IONs) radiolabeled with 68 Ga and 177 Lu could be used as potential agents for positron emission tomography (PET) for diagnosis and cancer therapy [139]. Chelator-free copper-64 incorporated with IONs showed good in vivo stability and could be used as a contrast agent for PET/MRI [140].
Several studies reported that modified multifunctional nanoparticles could be used as probes for imaging and theranostic agents in PDAC [141,142]. In this part, we focus on the value and potential applications of nanoparticles in imaging and theranostic roles in PDAC immunotherapy. The designed nanoparticles can increase efficiency of detection for PDAC, especially detection of early PDAC. For example, Wang et al. constructed Enolase 1 targeted superparamagnetic iron oxide nanoparticles that can increase MRI efficiency for detecting PDAC and facilitate early and accurate detection of PDAC [143]. Further, nanoparticles could be engineered to deliver imaging agents alone or in combination with anti-tumor agents, and thus used for imaging as well as image-guided and targeted cancer therapy. For example, Zhou et al. developed insulin-like growth factor 1 receptor receptor (IGF1R) multifunctional theranostic nanoparticles conjugated with recombinant human insulin-like growth factor 1 (IGF1) to magnetic IONs, carrying doxorubicin. These nanoparticles not only exhibited excellent anti-tumor effects in the orthotopic patient-derived xenograft (PDX) model, but could also be used as contrasts for MRI detection [144]. Image-guided treatment strategies can provide detailed information about each patient under treatment, which can then help monitor prognosis and adjust therapeutic interventions. Additionally, nanoparticles can be designed as probes to screen patients with PDAC suitable for immunotherapy. Liu et al. proposed nanoprobes that can sense tumor acidosis and hypoxic TME. This nanoprobe-based MRI could be used to monitor hypoxia and further predict the response to radiotherapy and immunotherapy in PDAC [145]. Additionally, Luo et al. developed metabolizable dextran–indocyanine green nanoprobes, which could obtain the dynamic image of deep-seated TAMs in PDAC. Since TAMs in the TME are significantly related to treatment outcomes and prognosis of PDAC, these nanoprobes present great potential for precision therapy, including immunotherapy in PDAC [146]. Furthermore, the immunotherapy process can exhibit specific imaging manifestations, such as immunotherapy hyperprogression [147]. Currently, there are no studies, to our knowledge, reporting on the role of nanoparticles in immunotherapy hyperprogression imaging, which is well worth studying, based on the sensitivity of nanoparticles displayed in the imaging.
In summary, although only a few studies on nanoparticles in imaging and therapeutic aspects of PDAC immunotherapy have been conducted, based on their potential role in these processes, further studies are necessary and valuable.

4. Conclusions and Perspectives

As a malignant tumor with an increasing mortality, there is an urgent need to find a more effective therapeutic strategy to improve prognosis for patients with PDAC. Despite the limited clinical efficacy of the current immunotherapy regimens for PDAC, a variety of clinical and preclinical evidence suggests that immunotherapy still holds great promise in the treatment of PDAC. Recently, nanomaterials have shown potential in overcoming current limitations in PDAC immunotherapy. The advantages of nanoparticles include controllable sizes and drug release, prolonged circulation, and co-delivery of multiple therapeutic agents in a site-specific manner. Thus, small-size nanoparticles can penetrate the dense stroma of PDAC to reach deeper parts of the tumor and activate the PDAC response to immunotherapy by regulating the TIME. Additionally, nanoparticles can be used as carriers for several types of immunomodulators, such as microRNAs and siRNAs. Furthermore, some new nanoparticle-based therapies, such as the nanoparticle-based PTT, PDT, CDT, and SDT, have shown promising efficacies in provoking an immune activation and inducing antitumor effects in PDAC.
However, while we recognize the hopes of immunotherapy in PDAC, we should also consider the challenges that nanomedicine may face in this. These challenges include unknown toxicity, selection of the right particle carrier, and translating results from preclinical studies to a clinical setting. Solving these problems will help nanomedicine improve the efficacy of immunotherapy and OS of patients with PDAC. This review provides new research directions for the clinical translation. We expect that nanomedicine will soon cooperate with immunotherapy to improve prognosis of patients with PDAC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pharmaceutics14102033/s1, Table S1: Published clinical trials of other immunotherapy in PDAC.

Author Contributions

Conceptualization, H.Z. and W.H.; Investigation, W.H. and B.Y. and H.Z.; Writing—Original Draft, H.Z., B.Y. and W.H.; Writing—Review and Editing, H.Z., B.Y. and W.H.; All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by grants from the Key research projects of Science & Technology of Sichuan Province (2022YFS0189) and the Fundamental Research Funds for the Central Universities (2022SCU12058).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACIadoptive cellular immunotherapy
ACTadoptive cell transfer
APCantigen-presenting cell
CAFcancer-associated fibroblast
CDTchemodynamic therapy
CSF-1colony-stimulating factor-1
CSF-1Rcolony-stimulating factor-1 receptor
CTLcytotoxic T cell lymphocyte
CTLA-4cytotoxic T-lymphocyte-associated protein 4
CXCL12chemokine C-X-C motif ligand 12
DAMPsdamage-associated molecular patterns
DCdendritic cell
dMMRmismatch repair-deficient
ECMextracellular matrix
5-FU6-5-fluorouracil
HAhyaluronic acid
HAhyaluronic acid
ICDImmunogenic cell death
ICIimmune checkpoint inhibitor
IDOindoleamine 2,3-dioxygenase
IFPinterstitial fluid pressure
IL-10Interleukin-10
IL-12Interleukin-12
immuno-NPimmunostimulatory nanoparticle
LMWHlow-molecular-weight heparin
MDSCmyeloid-derived suppressor cells
MPphosphate-modified α-mangostin
MSI-Hhigh microsatellite instability
MSNPmesoporous silica nanoparticle
Nabnano-albumin bound
ORRoverall response rate
OSoverall survival
PD-1programmed cell death protein-1
PDACpancreatic ductal adenocarcinoma
PD-L1programmed cell death ligand-1
PDTphotodynamic therapy
PEGpolyethyleneglycol
PEIpolyethylenimine
PEIpolyethylenimine
PFSprogression-free survival
PRpartial response
PSCpancreatic stellate cells
PTTphotothermal therapy
RIG-Iretinoic acid-inducible gene I
RLRretinoic acid-inducible gene I-like receptor
SBRTstereotactic body radiotherapy
SDstable disease
SDTsonodynamic therapy
SHHsonic hedgehog
TAMtumor-associated macrophage
TDLNtumor-draining lymph node
TILtumor-infiltrating lymphocyte
TIMEtumor immune microenvironment
TMEtumor microenvironment
Tregregulatory T cell
FAKfocal adhesion kinase
EPRenhanced permeability and retention
NIRnear-infrared
IPTTinterventional PTT
MRImagnetic resonance imaging
IONiron oxide nanoparticle
ICGindocyanine green
IMQimmunoadjuvant imiquimod
ROSreactive oxygen species
BRD4ibromodomains, and extraterminal protein 4 inhibitor
R-ExoS-reassembled exosome
CTcomputed tomography
PETpositron emission tomography
IGF1Rinsulin-like growth factor 1 receptor receptor
IGF1insulin-like growth factor 1
PDXpatient-derived xenograft
IONIron oxide nanoparticle
STAT3signal transducer and activator of transcription 3
TGF-βtransforming growth factor-beta
CXCR4C-X-C chemokine receptor 4
CCR7chemokine receptor 7

References

  1. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef]
  2. Park, W.; Chawla, A.; O’Reilly, E.M. Pancreatic Cancer: A Review. JAMA 2021, 326, 851–862. [Google Scholar] [CrossRef]
  3. Neoptolemos, J.P.; Kleeff, J.; Michl, P.; Costello, E.; Greenhalf, W.; Palmer, D.H. Therapeutic developments in pancreatic cancer: Current and future perspectives. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 333–348. [Google Scholar] [CrossRef] [PubMed]
  4. Chakrabarti, S.; Kamgar, M.; Mahipal, A. Systemic Therapy of Metastatic Pancreatic Adenocarcinoma: Current Status, Challenges, and Opportunities. Cancers 2022, 14, 2588. [Google Scholar] [CrossRef]
  5. Schizas, D.; Charalampakis, N.; Kole, C.; Economopoulou, P.; Koustas, E.; Gkotsis, E.; Ziogas, D.; Psyrri, A.; Karamouzis, M.V. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat. Rev. 2020, 86, 102016. [Google Scholar] [CrossRef]
  6. Götze, J.; Nitschke, C.; Uzunoglu, F.G.; Pantel, K.; Sinn, M.; Wikman, H. Tumor-Stroma Interaction in PDAC as a New Approach for Liquid Biopsy and its Potential Clinical Implications. Front. Cell Dev. Biol. 2022, 10, 918795. [Google Scholar] [CrossRef] [PubMed]
  7. Mace, T.A.; Ameen, Z.; Collins, A.; Wojcik, S.; Mair, M.; Young, G.S.; Fuchs, J.R.; Eubank, T.D.; Frankel, W.L.; Bekaii-Saab, T.; et al. Pancreatic cancer-associated stellate cells promote differentiation of myeloid-derived suppressor cells in a STAT3-dependent manner. Cancer Res. 2013, 73, 3007–3018. [Google Scholar] [CrossRef] [PubMed]
  8. Di Federico, A.; Mosca, M.; Pagani, R.; Carloni, R.; Frega, G.; De Giglio, A.; Rizzo, A.; Ricci, D.; Tavolari, S.; Di Marco, M.; et al. Immunotherapy in Pancreatic Cancer: Why Do We Keep Failing? A Focus on Tumor Immune Microenvironment, Predictive Biomarkers and Treatment Outcomes. Cancers 2022, 14, 2429. [Google Scholar] [CrossRef] [PubMed]
  9. Herting, C.J.; Karpovsky, I.; Lesinski, G.B. The tumor microenvironment in pancreatic ductal adenocarcinoma: Current perspectives and future directions. Cancer Metastasis Rev. 2021, 40, 675–689. [Google Scholar] [CrossRef] [PubMed]
  10. Li, S.; Xu, H.X.; Wu, C.T.; Wang, W.Q.; Jin, W.; Gao, H.L.; Li, H.; Zhang, S.R.; Xu, J.Z.; Qi, Z.H.; et al. Angiogenesis in pancreatic cancer: Current research status and clinical implications. Angiogenesis 2019, 22, 15–36. [Google Scholar] [CrossRef] [PubMed]
  11. Morrison, A.H.; Byrne, K.T.; Vonderheide, R.H. Immunotherapy and Prevention of Pancreatic Cancer. Trends Cancer 2018, 4, 418–428. [Google Scholar] [CrossRef] [PubMed]
  12. Ino, Y.; Yamazaki-Itoh, R.; Shimada, K.; Iwasaki, M.; Kosuge, T.; Kanai, Y.; Hiraoka, N. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br. J. Cancer 2013, 108, 914–923. [Google Scholar] [CrossRef]
  13. Bu, F.; Nie, H.; Zhu, X.; Wu, T.; Lin, K.; Zhao, J.; Huang, J. A signature of 18 immune-related gene pairs to predict the prognosis of pancreatic cancer patients. Immun. Inflamm. Dis. 2020, 8, 713–726. [Google Scholar] [CrossRef]
  14. Ota, S.; Miyashita, M.; Yamagishi, Y.; Ogasawara, M. Baseline immunity predicts prognosis of pancreatic cancer patients treated with WT1 and/or MUC1 peptide-loaded dendritic cell vaccination and a standard chemotherapy. Hum. Vaccines Immunother. 2021, 17, 5563–5572. [Google Scholar] [CrossRef] [PubMed]
  15. Sadozai, H.; Acharjee, A.; Eppenberger-Castori, S.; Gloor, B.; Gruber, T.; Schenk, M.; Karamitopoulou, E. Distinct Stromal and Immune Features Collectively Contribute to Long-Term Survival in Pancreatic Cancer. Front. Immunol. 2021, 12, 643529. [Google Scholar] [CrossRef] [PubMed]
  16. Balachandran, V.P.; Łuksza, M.; Zhao, J.N.; Makarov, V.; Moral, J.A.; Remark, R.; Herbst, B.; Askan, G.; Bhanot, U.; Senbabaoglu, Y.; et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017, 551, 512–516. [Google Scholar] [CrossRef]
  17. Zhu, X.; Cao, Y.; Liu, W.; Ju, X.; Zhao, X.; Jiang, L.; Ye, Y.; Jin, G.; Zhang, H. Stereotactic body radiotherapy plus pembrolizumab and trametinib versus stereotactic body radiotherapy plus gemcitabine for locally recurrent pancreatic cancer after surgical resection: An open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2021, 22, 1093–1102. [Google Scholar] [CrossRef]
  18. Byrne, K.T.; Betts, C.B.; Mick, R.; Sivagnanam, S.; Bajor, D.L.; Laheru, D.A.; Chiorean, E.G.; O’Hara, M.H.; Liudahl, S.M.; Newcomb, C.; et al. Neoadjuvant Selicrelumab, an Agonist CD40 Antibody, Induces Changes in the Tumor Microenvironment in Patients with Resectable Pancreatic Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 4574–4586. [Google Scholar] [CrossRef]
  19. Kamath, S.D.; Kalyan, A.; Kircher, S.; Nimeiri, H.; Fought, A.J.; Benson, A., 3rd; Mulcahy, M. Ipilimumab and Gemcitabine for Advanced Pancreatic Cancer: A Phase Ib Study. Oncology 2020, 25, e808–e815. [Google Scholar] [CrossRef]
  20. Krenz, B.; Gebhardt-Wolf, A.; Ade, C.P.; Gaballa, A.; Roehrig, F.; Vendelova, E.; Baluapuri, A.; Eilers, U.; Gallant, P.; D’Artista, L.; et al. MYC- and MIZ1-Dependent Vesicular Transport of Double-Strand RNA Controls Immune Evasion in Pancreatic Ductal Adenocarcinoma. Cancer Res. 2021, 81, 4242–4256. [Google Scholar] [CrossRef] [PubMed]
  21. Kim, M.J.; Chang, H.; Nam, G.; Ko, Y.; Kim, S.H.; Roberts, T.M.; Ryu, J.H. RNAi-Based Approaches for Pancreatic Cancer Therapy. Pharmaceutics 2021, 13, 1638. [Google Scholar] [CrossRef] [PubMed]
  22. Zhou, W.; Zhou, Y.; Chen, X.; Ning, T.; Chen, H.; Guo, Q.; Zhang, Y.; Liu, P.; Zhang, Y.; Li, C.; et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials 2021, 268, 120546. [Google Scholar] [CrossRef]
  23. Lakkakula, S.; Chalikonda, G.; Lakkakula, B. Nanoparticles in Pancreatic Cancer Imaging and Therapy. Crit. Rev. Oncog. 2019, 24, 139–148. [Google Scholar] [CrossRef]
  24. Yardley, D.A. Nab-Paclitaxel mechanisms of action and delivery. J. Control. Release Off. J. Control. Release Soc. 2013, 170, 365–372. [Google Scholar] [CrossRef] [PubMed]
  25. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [PubMed]
  26. Frampton, J.E. Liposomal Irinotecan: A Review in Metastatic Pancreatic Adenocarcinoma. Drugs 2020, 80, 1007–1018. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, L.; Kshirsagar, P.G.; Gautam, S.K.; Gulati, M.; Wafa, E.I.; Christiansen, J.C.; White, B.M.; Mallapragada, S.K.; Wannemuehler, M.J.; Kumar, S.; et al. Nanocarriers for pancreatic cancer imaging, treatments, and immunotherapies. Theranostics 2022, 12, 1030–1060. [Google Scholar] [CrossRef] [PubMed]
  28. Jia, M.; Zhang, D.; Zhang, C.; Li, C. Nanoparticle-based delivery systems modulate the tumor microenvironment in pancreatic cancer for enhanced therapy. J. Nanobiotechnol. 2021, 19, 384. [Google Scholar] [CrossRef] [PubMed]
  29. 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, 807–821. [Google Scholar] [CrossRef] [PubMed]
  30. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef] [PubMed]
  31. Hou, W.; Zhou, X.; Yi, C.; Zhu, H. Immune Check Point Inhibitors and Immune-Related Adverse Events in Small Cell Lung Cancer. Front. Oncol. 2021, 11, 604227. [Google Scholar] [CrossRef] [PubMed]
  32. Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef]
  33. Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
  34. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef]
  35. Ahmad-Nielsen, S.A.; Bruun Nielsen, M.F.; Mortensen, M.B.; Detlefsen, S. Frequency of mismatch repair deficiency in pancreatic ductal adenocarcinoma. Pathol. Res. Pract. 2020, 216, 152985. [Google Scholar] [CrossRef]
  36. O’Reilly, E.M.; Oh, D.Y.; Dhani, N.; Renouf, D.J.; Lee, M.A.; Sun, W.; Fisher, G.; Hezel, A.; Chang, S.C.; Vlahovic, G.; et al. Durvalumab with or without Tremelimumab for Patients with Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019, 5, 1431–1438. [Google Scholar] [CrossRef]
  37. Deshmukh, S.K.; Tyagi, N.; Khan, M.A.; Srivastava, S.K.; Al-Ghadhban, A.; Dugger, K.; Carter, J.E.; Singh, S.; Singh, A.P. Gemcitabine treatment promotes immunosuppressive microenvironment in pancreatic tumors by supporting the infiltration, growth, and polarization of macrophages. Sci. Rep. 2018, 8, 12000. [Google Scholar] [CrossRef]
  38. Aglietta, M.; Barone, C.; Sawyer, M.B.; Moore, M.J.; Miller, W.H., Jr.; Bagalà, C.; Colombi, F.; Cagnazzo, C.; Gioeni, L.; Wang, E.; et al. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2014, 25, 1750–1755. [Google Scholar] [CrossRef]
  39. Kalyan, A.; Kircher, S.M.; Mohindra, N.A.; Nimeiri, H.S.; Maurer, V.; Rademaker, A.; Benson, A.B.; Mulcahy, M.F. Ipilimumab and gemcitabine for advanced pancreas cancer: A phase Ib study. J. Clin. Oncol. 2016, 34 (Suppl. S5), e15747. [Google Scholar] [CrossRef]
  40. Weiss, G.J.; Blaydorn, L.; Beck, J.; Bornemann-Kolatzki, K.; Urnovitz, H.; Schütz, E.; Khemka, V. Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Investig. New Drugs 2018, 36, 96–102. [Google Scholar] [CrossRef]
  41. Wainberg, Z.A.; Hochster, H.S.; Kim, E.J.; George, B.; Kaylan, A.; Chiorean, E.G.; Waterhouse, D.M.; Guiterrez, M.; Parikh, A.; Jain, R.; et al. Open-label, Phase I Study of Nivolumab Combined with nab-Paclitaxel Plus Gemcitabine in Advanced Pancreatic Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4814–4822. [Google Scholar] [CrossRef] [PubMed]
  42. Fu, Q.; Chen, Y.; Huang, D.; Guo, C.; Zhang, Q.; Li, X.; Zhang, X.; Gao, S.; Que, R.; Shen, Y.; et al. Randomized phase III study of sintilimab in combination with modified folfrinox versus folfrinox alone in patients with metastatic and recurrent pancreatic cancer in China: The CISPD3 trial. J. Clin. Oncol. 2022, 40 (Suppl. S4), 560. [Google Scholar] [CrossRef]
  43. Renouf, D.J.; Loree, J.M.; Knox, J.J.; Kavan, P.; Jonker, D.J.; Welch, S.; Couture, F.; Lemay, F.; Tehfe, M.; Harb, M.; et al. Predictive value of germline ATM mutations in the CCTG PA.7 trial: Gemcitabine (GEM) and nab-paclitaxel (Nab-P) versus GEM, nab-P, durvalumab (D) and tremelimumab (T) as first-line therapy in metastatic pancreatic ductal adenocarcinoma (mPDAC). J. Clin. Oncol. 2021, 39 (Suppl. S15), 4135. [Google Scholar] [CrossRef]
  44. Luo, W.; Wang, Y.; Zhang, T. Win or loss? Combination therapy does improve the oncolytic virus therapy to pancreatic cancer. Cancer Cell Int. 2022, 22, 160. [Google Scholar] [CrossRef] [PubMed]
  45. Lum, L.G.; Thakur, A.; Choi, M.; Deol, A.; Kondadasula, V.; Schalk, D.; Fields, K.; Dufrense, M.; Philip, P.; Dyson, G.; et al. Clinical and immune responses to anti-CD3 x anti-EGFR bispecific antibody armed activated T cells (EGFR BATs) in pancreatic cancer patients. Oncoimmunology 2020, 9, 1773201. [Google Scholar] [CrossRef]
  46. Middleton, G.; Silcocks, P.; Cox, T.; Valle, J.; Wadsley, J.; Propper, D.; Coxon, F.; Ross, P.; Madhusudan, S.; Roques, T.; et al. Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): An open-label, randomised, phase 3 trial. Lancet Oncol. 2014, 15, 829–840. [Google Scholar] [CrossRef]
  47. Lucia, F.; Geier, M.; Schick, U.; Bourbonne, V. Narrative Review of Synergistics Effects of Combining Immunotherapy and Stereotactic Radiation Therapy. Biomedicines 2022, 10, 1414. [Google Scholar] [CrossRef]
  48. Stump, C.T.; Roehle, K.; Manjarrez Orduno, N.; Dougan, S.K. Radiation combines with immune checkpoint blockade to enhance T cell priming in a murine model of poorly immunogenic pancreatic cancer. Open Biol. 2021, 11, 210245. [Google Scholar] [CrossRef]
  49. Chen, I.M.; Johansen, J.S.; Theile, S.; Hjaltelin, J.X.; Novitski, S.I.; Brunak, S.; Hasselby, J.P.; Willemoe, G.L.; Lorentzen, T.; Madsen, K.; et al. Randomized Phase II Study of Nivolumab with or without Ipilimumab Combined with Stereotactic Body Radiotherapy for Refractory Metastatic Pancreatic Cancer (CheckPAC). J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 3180–3189. [Google Scholar] [CrossRef]
  50. Parikh, A.R.; Szabolcs, A.; Allen, J.N.; Clark, J.W.; Wo, J.Y.; Raabe, M.; Thel, H.; Hoyos, D.; Mehta, A.; Arshad, S.; et al. Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase II trial. Nat. Cancer 2021, 2, 1124–1135. [Google Scholar] [CrossRef]
  51. Xie, C.; Duffy, A.G.; Brar, G.; Fioravanti, S.; Mabry-Hrones, D.; Walker, M.; Bonilla, C.M.; Wood, B.J.; Citrin, D.E.; Gil Ramirez, E.M.; et al. Immune Checkpoint Blockade in Combination with Stereotactic Body Radiotherapy in Patients with Metastatic Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2318–2326. [Google Scholar] [CrossRef] [PubMed]
  52. Brouwer, T.P.; Vahrmeijer, A.L.; de Miranda, N. Immunotherapy for pancreatic cancer: Chasing the light at the end of the tunnel. Cell. Oncol. 2021, 44, 261–278. [Google Scholar] [CrossRef] [PubMed]
  53. Yin, C.; Alqahtani, A.; Noel, M.S. The Next Frontier in Pancreatic Cancer: Targeting the Tumor Immune Milieu and Molecular Pathways. Cancers 2022, 14, 2619. [Google Scholar] [CrossRef] [PubMed]
  54. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef]
  55. Muller, M.; Haghnejad, V.; Schaefer, M.; Gauchotte, G.; Caron, B.; Peyrin-Biroulet, L.; Bronowicki, J.P.; Neuzillet, C.; Lopez, A. The Immune Landscape of Human Pancreatic Ductal Carcinoma: Key Players, Clinical Implications, and Challenges. Cancers 2022, 14, 995. [Google Scholar] [CrossRef]
  56. Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef]
  57. Thomas, D.; Radhakrishnan, P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol. Cancer 2019, 18, 14. [Google Scholar] [CrossRef]
  58. Murphy, K.J.; Chambers, C.R.; Herrmann, D.; Timpson, P.; Pereira, B.A. Dynamic Stromal Alterations Influence Tumor-Stroma Crosstalk to Promote Pancreatic Cancer and Treatment Resistance. Cancers 2021, 13, 3481. [Google Scholar] [CrossRef]
  59. Rhim, A.D.; Oberstein, P.E.; Thomas, D.H.; Mirek, E.T.; Palermo, C.F.; Sastra, S.A.; Dekleva, E.N.; Saunders, T.; Becerra, C.P.; Tattersall, I.W.; et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 2014, 25, 735–747. [Google Scholar] [CrossRef]
  60. Tao, J.; Yang, G.; Zhou, W.; Qiu, J.; Chen, G.; Luo, W.; Zhao, F.; You, L.; Zheng, L.; Zhang, T.; et al. Targeting hypoxic tumor microenvironment in pancreatic cancer. J. Hematol. Oncol. 2021, 14, 14. [Google Scholar] [CrossRef]
  61. Garcia, C.J.G.; Huang, Y.; Fuentes, N.R.; Turner, M.C.; Monberg, M.E.; Lin, D.; Nguyen, N.D.; Fujimoto, T.N.; Zhao, J.; Lee, J.J.; et al. Stromal HIF2 Regulates Immune Suppression in the Pancreatic Cancer Microenvironment. Gastroenterology 2022, 162, 2018–2031. [Google Scholar] [CrossRef] [PubMed]
  62. Wu, J.; Wang, X.; Chen, L.; Wang, J.; Zhang, J.; Tang, J.; Ji, Y.; Song, J.; Wang, L.; Zhao, Y.; et al. Oxygen microcapsules improve immune checkpoint blockade by ameliorating hypoxia condition in pancreatic ductal adenocarcinoma. Bioact. Mater. 2022, 20, 259–270. [Google Scholar] [CrossRef] [PubMed]
  63. Hegde, A.; Jayaprakash, P.; Couillault, C.A.; Piha-Paul, S.; Karp, D.; Rodon, J.; Pant, S.; Fu, S.; Dumbrava, E.E.; Yap, T.A.; et al. A Phase I Dose-Escalation Study to Evaluate the Safety and Tolerability of Evofosfamide in Combination with Ipilimumab in Advanced Solid Malignancies. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 3050–3060. [Google Scholar] [CrossRef]
  64. Li, S.; Zhang, Q.; Hong, Y. Tumor Vessel Normalization: A Window to Enhancing Cancer Immunotherapy. Technol. Cancer Res. Treat. 2020, 19, 1533033820980116. [Google Scholar] [CrossRef]
  65. Nagathihalli, N.S.; Castellanos, J.A.; Shi, C.; Beesetty, Y.; Reyzer, M.L.; Caprioli, R.; Chen, X.; Walsh, A.J.; Skala, M.C.; Moses, H.L.; et al. Signal Transducer and Activator of Transcription 3, Mediated Remodeling of the Tumor Microenvironment Results in Enhanced Tumor Drug Delivery in a Mouse Model of Pancreatic Cancer. Gastroenterology 2015, 149, 1932–1943.e1939. [Google Scholar] [CrossRef] [PubMed]
  66. Gilles, M.E.; Maione, F.; Cossutta, M.; Carpentier, G.; Caruana, L.; Di Maria, S.; Houppe, C.; Destouches, D.; Shchors, K.; Prochasson, C.; et al. Nucleolin Targeting Impairs the Progression of Pancreatic Cancer and Promotes the Normalization of Tumor Vasculature. Cancer Res. 2016, 76, 7181–7193. [Google Scholar] [CrossRef]
  67. Giordano, G.; Pancione, M.; Olivieri, N.; Parcesepe, P.; Velocci, M.; Di Raimo, T.; Coppola, L.; Toffoli, G.; D’Andrea, M.R. Nano albumin bound-paclitaxel in pancreatic cancer: Current evidences and future directions. World J. Gastroenterol. 2017, 23, 5875–5886. [Google Scholar] [CrossRef]
  68. Bhaw-Luximon, A.; Jhurry, D. New avenues for improving pancreatic ductal adenocarcinoma (PDAC) treatment: Selective stroma depletion combined with nano drug delivery. Cancer Lett. 2015, 369, 266–273. [Google Scholar] [CrossRef]
  69. Meng, H.; Nel, A.E. Use of nano engineered approaches to overcome the stromal barrier in pancreatic cancer. Adv. Drug Deliv. Rev. 2018, 130, 50–57. [Google Scholar] [CrossRef] [PubMed]
  70. Yu, Q.; Tang, X.; Zhao, W.; Qiu, Y.; He, J.; Wan, D.; Li, J.; Wang, X.; He, X.; Liu, Y.; et al. Mild hyperthermia promotes immune checkpoint blockade-based immunotherapy against metastatic pancreatic cancer using size-adjustable nanoparticles. Acta Biomater. 2021, 133, 244–256. [Google Scholar] [CrossRef]
  71. Li, S.; Zhang, W.; Xing, R.; Yuan, C.; Xue, H.; Yan, X. Supramolecular Nanofibrils Formed by Coassembly of Clinically Approved Drugs for Tumor Photothermal Immunotherapy. Adv. Mater. 2021, 33, e2100595. [Google Scholar] [CrossRef]
  72. Sun, J.; Wan, Z.; Xu, J.; Luo, Z.; Ren, P.; Zhang, B.; Diao, D.; Huang, Y.; Li, S. Tumor size-dependent abscopal effect of polydopamine-coated all-in-one nanoparticles for immunochemo-photothermal therapy of early- and late-stage metastatic cancer. Biomaterials 2021, 269, 120629. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, M.; Li, Y.; Wang, M.; Liu, K.; Hoover, A.R.; Li, M.; Towner, R.A.; Mukherjee, P.; Zhou, F.; Qu, J.; et al. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater. 2022, 138, 453–462. [Google Scholar] [CrossRef]
  74. Sun, F.; Zhu, Q.; Li, T.; Saeed, M.; Xu, Z.; Zhong, F.; Song, R.; Huai, M.; Zheng, M.; Xie, C.; et al. Regulating Glucose Metabolism with Prodrug Nanoparticles for Promoting Photoimmunotherapy of Pancreatic Cancer. Adv. Sci. 2021, 8, 2002746. [Google Scholar] [CrossRef]
  75. Jang, Y.; Kim, H.; Yoon, S.; Lee, H.; Hwang, J.; Jung, J.; Chang, J.H.; Choi, J.; Kim, H. Exosome-based photoacoustic imaging guided photodynamic and immunotherapy for the treatment of pancreatic cancer. J. Control. Release Off. J. Control. Release Soc. 2021, 330, 293–304. [Google Scholar] [CrossRef] [PubMed]
  76. Nicholas, D.; Nesbitt, H.; Farrell, S.; Logan, K.; McMullin, E.; Gillan, T.; Kelly, P.; O’Rourke, D.; Porter, S.; Thomas, K.; et al. Exploiting a Rose Bengal-bearing, oxygen-producing nanoparticle for SDT and associated immune-mediated therapeutic effects in the treatment of pancreatic cancer. Eur. J. Pharm. Biopharm. Off. J. Arb. Fur Pharm. Verfahr. Ev 2021, 163, 49–59. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, J.; Feng, L.; Jin, P.; Shen, J.; Lu, J.; Song, Y.; Wang, G.; Chen, Q.; Huang, D.; Zhang, Y.; et al. Cavitation assisted endoplasmic reticulum targeted sonodynamic droplets to enhanced anti-PD-L1 immunotherapy in pancreatic cancer. J. Nanobiotechnol. 2022, 20, 283. [Google Scholar] [CrossRef]
  78. Chen, Y.; Huang, Y.; Zhou, S.; Sun, M.; Chen, L.; Wang, J.; Xu, M.; Liu, S.; Liang, K.; Zhang, Q.; et al. Tailored Chemodynamic Nanomedicine Improves Pancreatic Cancer Treatment via Controllable Damaging Neoplastic Cells and Reprogramming Tumor Microenvironment. Nano Lett. 2020, 20, 6780–6790. [Google Scholar] [CrossRef]
  79. Zhao, X.; Yang, K.; Zhao, R.; Ji, T.; Wang, X.; Yang, X.; Zhang, Y.; Cheng, K.; Liu, S.; Hao, J.; et al. Inducing enhanced immunogenic cell death with nanocarrier-based drug delivery systems for pancreatic cancer therapy. Biomaterials 2016, 102, 187–197. [Google Scholar] [CrossRef] [PubMed]
  80. Liu, X.; Jiang, J.; Chang, C.H.; Liao, Y.P.; Lodico, J.J.; Tang, I.; Zheng, E.; Qiu, W.; Lin, M.; Wang, X.; et al. Development of Facile and Versatile Platinum Drug Delivering Silicasome Nanocarriers for Efficient Pancreatic Cancer Chemo-Immunotherapy. Small 2021, 17, e2005993. [Google Scholar] [CrossRef]
  81. Shen, J.; Sun, C.; Wang, Z.; Chu, Z.; Liu, C.; Xu, X.; Xia, M.; Zhao, M.; Wang, C. Sequential receptor-mediated mixed-charge nanomedicine to target pancreatic cancer, inducing immunogenic cell death and reshaping the tumor microenvironment. Int. J. Pharm. 2021, 601, 120553. [Google Scholar] [CrossRef] [PubMed]
  82. Parayath, N.N.; Hong, B.V.; Mackenzie, G.G.; Amiji, M.M. Hyaluronic acid nanoparticle-encapsulated microRNA-125b repolarizes tumor-associated macrophages in pancreatic cancer. Nanomedicine 2021, 16, 2291–2303. [Google Scholar] [CrossRef]
  83. Li, M.; Li, M.; Yang, Y.; Liu, Y.; Xie, H.; Yu, Q.; Tian, L.; Tang, X.; Ren, K.; Li, J.; et al. Remodeling tumor immune microenvironment via targeted blockade of PI3K-γ and CSF-1/CSF-1R pathways in tumor associated macrophages for pancreatic cancer therapy. J. Control. Release Off. J. Control. Release Soc. 2020, 321, 23–35. [Google Scholar] [CrossRef]
  84. Cao, S.; Saw, P.E.; Shen, Q.; Li, R.; Liu, Y.; Xu, X. Reduction-responsive RNAi nanoplatform to reprogram tumor lipid metabolism and repolarize macrophage for combination pancreatic cancer therapy. Biomaterials 2022, 280, 121264. [Google Scholar] [CrossRef]
  85. Lu, Z.; Long, Y.; Wang, Y.; Wang, X.; Xia, C.; Li, M.; Zhang, Z.; He, Q. Phenylboronic acid modified nanoparticles simultaneously target pancreatic cancer and its metastasis and alleviate immunosuppression. Eur. J. Pharm. Biopharm. Off. J. Arb. Fur Pharm. Verfahr. Ev 2021, 165, 164–173. [Google Scholar] [CrossRef] [PubMed]
  86. Lorkowski, M.E.; Atukorale, P.U.; Bielecki, P.A.; Tong, K.H.; Covarrubias, G.; Zhang, Y.; Loutrianakis, G.; Moon, T.J.; Santulli, A.R.; Becicka, W.M.; et al. Immunostimulatory nanoparticle incorporating two immune agonists for the treatment of pancreatic tumors. J. Control. Release Off. J. Control. Release Soc. 2021, 330, 1095–1105. [Google Scholar] [CrossRef] [PubMed]
  87. Miao, L.; Li, J.; Liu, Q.; Feng, R.; Das, M.; Lin, C.M.; Goodwin, T.J.; Dorosheva, O.; Liu, R.; Huang, L. Transient and Local Expression of Chemokine and Immune Checkpoint Traps to Treat Pancreatic Cancer. ACS Nano 2017, 11, 8690–8706. [Google Scholar] [CrossRef] [PubMed]
  88. Shen, L.; Li, J.; Liu, Q.; Song, W.; Zhang, X.; Tiruthani, K.; Hu, H.; Das, M.; Goodwin, T.J.; Liu, R.; et al. Local Blockade of Interleukin 10 and C-X-C Motif Chemokine Ligand 12 with Nano-Delivery Promotes Antitumor Response in Murine Cancers. ACS Nano 2018, 12, 9830–9841. [Google Scholar] [CrossRef]
  89. Das, M.; Shen, L.; Liu, Q.; Goodwin, T.J.; Huang, L. Nanoparticle Delivery of RIG-I Agonist Enables Effective and Safe Adjuvant Therapy in Pancreatic Cancer. Molecular therapy: J. Am. Soc. Gene Ther. 2019, 27, 507–517. [Google Scholar] [CrossRef] [PubMed]
  90. Han, B.J.; Murphy, J.D.; Qin, S.; Ye, J.; Uccello, T.P.; Garrett-Larsen, J.; Belt, B.A.; Prieto, P.A.; Egilmez, N.K.; Lord, E.M.; et al. Microspheres Encapsulating Immunotherapy Agents Target the Tumor-Draining Lymph Node in Pancreatic Ductal Adenocarcinoma. Immunol. Investig. 2020, 49, 808–823. [Google Scholar] [CrossRef]
  91. Jung, J.Y.; Ryu, H.J.; Lee, S.H.; Kim, D.Y.; Kim, M.J.; Lee, E.J.; Ryu, Y.M.; Kim, S.Y.; Kim, K.P.; Choi, E.Y.; et al. siRNA Nanoparticle Targeting PD-L1 Activates Tumor Immunity and Abrogates Pancreatic Cancer Growth in Humanized Preclinical Model. Cells 2021, 10, 2734. [Google Scholar] [CrossRef] [PubMed]
  92. Dong, H.; Wen, Z.F.; Chen, L.; Zhou, N.; Liu, H.; Dong, S.; Hu, H.M.; Mou, Y. Polyethyleneimine modification of aluminum hydroxide nanoparticle enhances antigen transportation and cross-presentation of dendritic cells. Int. J. Nanomed. 2018, 13, 3353–3365. [Google Scholar] [CrossRef]
  93. Lu, J.; Liu, X.; Liao, Y.P.; Salazar, F.; Sun, B.; Jiang, W.; Chang, C.H.; Jiang, J.; Wang, X.; Wu, A.M.; et al. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat. Commun. 2017, 8, 1811. [Google Scholar] [CrossRef]
  94. Huang, H.; Jiang, C.T.; Shen, S.; Liu, A.; Gan, Y.J.; Tong, Q.S.; Chen, S.B.; Gao, Z.X.; Du, J.Z.; Cao, J.; et al. Nanoenabled Reversal of IDO1-Mediated Immunosuppression Synergizes with Immunogenic Chemotherapy for Improved Cancer Therapy. Nano Lett. 2019, 19, 5356–5365. [Google Scholar] [CrossRef]
  95. Sun, J.; Wan, Z.; Chen, Y.; Xu, J.; Luo, Z.; Parise, R.A.; Diao, D.; Ren, P.; Beumer, J.H.; Lu, B.; et al. Triple drugs co-delivered by a small gemcitabine-based carrier for pancreatic cancer immunochemotherapy. Acta Biomater. 2020, 106, 289–300. [Google Scholar] [CrossRef] [PubMed]
  96. Tong, Q.S.; Miao, W.M.; Huang, H.; Luo, J.Q.; Liu, R.; Huang, Y.C.; Zhao, D.K.; Shen, S.; Du, J.Z.; Wang, J. A Tumor-Penetrating Nanomedicine Improves the Chemoimmunotherapy of Pancreatic Cancer. Small 2021, 17, e2101208. [Google Scholar] [CrossRef]
  97. Chen, Q.; Wang, Q.; Wang, Y.; Chu, Y.; Luo, Y.; You, H.; Su, B.; Li, C.; Guo, Q.; Sun, T.; et al. Penetrating Micelle for Reversing Immunosuppression and Drug Resistance in Pancreatic Cancer Treatment. Small 2022, 18, e2107712. [Google Scholar] [CrossRef] [PubMed]
  98. Hu, Y.; Chen, X.; Xu, Y.; Han, X.; Wang, M.; Gong, T.; Zhang, Z.R.; Kao, W.J.; Fu, Y. Hierarchical assembly of hyaluronan coated albumin nanoparticles for pancreatic cancer chemoimmunotherapy. Nanoscale 2019, 11, 16476–16487. [Google Scholar] [CrossRef]
  99. Liu, X.; Jiang, J.; Liao, Y.P.; Tang, I.; Zheng, E.; Qiu, W.; Lin, M.; Wang, X.; Ji, Y.; Mei, K.C.; et al. Combination Chemo-Immunotherapy for Pancreatic Cancer Using the Immunogenic Effects of an Irinotecan Silicasome Nanocarrier Plus Anti-PD-1. Adv. Sci. 2021, 8, 2002147. [Google Scholar] [CrossRef]
  100. Wang, M.; Hu, Q.; Huang, J.; Zhao, X.; Shao, S.; Zhang, F.; Yao, Z.; Ping, Y.; Liang, T. Engineered a dual-targeting biomimetic nanomedicine for pancreatic cancer chemoimmunotherapy. J. Nanobiotechnol. 2022, 20, 85. [Google Scholar] [CrossRef] [PubMed]
  101. Huang, Y.; Chen, Y.; Zhou, S.; Chen, L.; Wang, J.; Pei, Y.; Xu, M.; Feng, J.; Jiang, T.; Liang, K.; et al. Dual-mechanism based CTLs infiltration enhancement initiated by Nano-sapper potentiates immunotherapy against immune-excluded tumors. Nat. Commun. 2020, 11, 622. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, Y.; Gao, Z.; Du, X.; Chen, S.; Zhang, W.; Wang, J.; Li, H.; He, X.; Cao, J.; Wang, J. Co-inhibition of the TGF-β pathway and the PD-L1 checkpoint by pH-responsive clustered nanoparticles for pancreatic cancer microenvironment regulation and anti-tumor immunotherapy. Biomater. Sci. 2020, 8, 5121–5132. [Google Scholar] [CrossRef] [PubMed]
  103. Zhao, J.; Xiao, Z.; Li, T.; Chen, H.; Yuan, Y.; Wang, Y.A.; Hsiao, C.H.; Chow, D.S.; Overwijk, W.W.; Li, C. Stromal Modulation Reverses Primary Resistance to Immune Checkpoint Blockade in Pancreatic Cancer. ACS Nano 2018, 12, 9881–9893. [Google Scholar] [CrossRef] [PubMed]
  104. Blair, A.B.; Kim, V.M.; Muth, S.T.; Saung, M.T.; Lokker, N.; Blouw, B.; Armstrong, T.D.; Jaffee, E.M.; Tsujikawa, T.; Coussens, L.M.; et al. Dissecting the Stromal Signaling and Regulation of Myeloid Cells and Memory Effector T Cells in Pancreatic Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 5351–5363. [Google Scholar] [CrossRef]
  105. Blair, A.B.; Wang, J.; Davelaar, J.; Baker, A.; Li, K.; Niu, N.; Wang, J.; Shao, Y.; Funes, V.; Li, P.; et al. Dual stromal targeting sensitizes pancreatic adenocarcinoma for anti-PD-1 therapy. Gastroenterology 2022. [Google Scholar] [CrossRef]
  106. Chen, X.; Zhou, W.; Liang, C.; Shi, S.; Yu, X.; Chen, Q.; Sun, T.; Lu, Y.; Zhang, Y.; Guo, Q.; et al. Codelivery Nanosystem Targeting the Deep Microenvironment of Pancreatic Cancer. Nano Lett. 2019, 19, 3527–3534. [Google Scholar] [CrossRef]
  107. Xie, Y.; Hang, Y.; Wang, Y.; Sleightholm, R.; Prajapati, D.R.; Bader, J.; Yu, A.; Tang, W.; Jaramillo, L.; Li, J.; et al. Stromal Modulation and Treatment of Metastatic Pancreatic Cancer with Local Intraperitoneal Triple miRNA/siRNA Nanotherapy. ACS Nano 2020, 14, 255–271. [Google Scholar] [CrossRef]
  108. Ahmed, A.; Tait, S.W.G. Targeting immunogenic cell death in cancer. Mol. Oncol. 2020, 14, 2994–3006. [Google Scholar] [CrossRef]
  109. Duan, X.; Chan, C.; Lin, W. Nanoparticle-Mediated Immunogenic Cell Death Enables and Potentiates Cancer Immunotherapy. Angew. Chem. 2019, 58, 670–680. [Google Scholar]
  110. Jiang, M.; Zeng, J.; Zhao, L.; Zhang, M.; Ma, J.; Guan, X.; Zhang, W. Chemotherapeutic drug-induced immunogenic cell death for nanomedicine-based cancer chemo-immunotherapy. Nanoscale 2021, 13, 17218–17235. [Google Scholar] [CrossRef]
  111. von Itzstein, M.S.; Burke, M.C.; Brekken, R.A.; Aguilera, T.A.; Zeh, H.J.; Beg, M.S. Targeting TAM to Tame Pancreatic Cancer. Target. Oncol. 2020, 15, 579–588. [Google Scholar] [CrossRef] [PubMed]
  112. Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; DeNardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014, 74, 5057–5069. [Google Scholar] [CrossRef]
  113. Pergamo, M.; Miller, G. Myeloid-derived suppressor cells and their role in pancreatic cancer. Cancer Gene Ther. 2017, 24, 100–105. [Google Scholar] [CrossRef] [PubMed]
  114. Li, S.; Wang, Q.; Shen, Y.; Hassan, M.; Shen, J.; Jiang, W.; Su, Y.; Chen, J.; Bai, L.; Zhou, W.; et al. Pseudoneutrophil Cytokine Sponges Disrupt Myeloid Expansion and Tumor Trafficking to Improve Cancer Immunotherapy. Nano Lett. 2020, 20, 242–251. [Google Scholar] [CrossRef] [PubMed]
  115. Long, Y.; Lu, Z.; Xu, S.; Li, M.; Wang, X.; Zhang, Z.; He, Q. Self-Delivery Micellar Nanoparticles Prevent Premetastatic Niche Formation by Interfering with the Early Recruitment and Vascular Destruction of Granulocytic Myeloid-Derived Suppressor Cells. Nano Lett. 2020, 20, 2219–2229. [Google Scholar] [CrossRef]
  116. Wu, W.; Wang, X.; Zhang, W.; Tian, L.; Booth, J.L.; Duggan, E.S.; More, S.; Liu, L.; Dozmorov, M.; Metcalf, J.P. RIG-I Signaling via MAVS Is Dispensable for Survival in Lethal Influenza Infection In Vivo. Mediat. Inflamm. 2018, 2018, 6808934. [Google Scholar] [CrossRef]
  117. Wu, Y.; Chen, W.; Xu, Z.P.; Gu, W. PD-L1 Distribution and Perspective for Cancer Immunotherapy-Blockade, Knockdown, or Inhibition. Front. Immunol. 2019, 10, 2022. [Google Scholar] [CrossRef]
  118. Zhou, J.; Wang, G.; Chen, Y.; Wang, H.; Hua, Y.; Cai, Z. Immunogenic cell death in cancer therapy: Present and emerging inducers. J. Cell. Mol. Med. 2019, 23, 4854–4865. [Google Scholar] [CrossRef]
  119. Zhao, T.; Ren, H.; Jia, L.; Chen, J.; Xin, W.; Yan, F.; Li, J.; Wang, X.; Gao, S.; Qian, D.; et al. Inhibition of HIF-1α by PX-478 enhances the anti-tumor effect of gemcitabine by inducing immunogenic cell death in pancreatic ductal adenocarcinoma. Oncotarget 2015, 6, 2250–2262. [Google Scholar] [CrossRef]
  120. Zhan, H.X.; Zhou, B.; Cheng, Y.G.; Xu, J.W.; Wang, L.; Zhang, G.Y.; Hu, S.Y. Crosstalk between stromal cells and cancer cells in pancreatic cancer: New insights into stromal biology. Cancer Lett. 2017, 392, 83–93. [Google Scholar] [CrossRef]
  121. Hosein, A.N.; Brekken, R.A.; Maitra, A. Pancreatic cancer stroma: An update on therapeutic targeting strategies. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 487–505. [Google Scholar] [CrossRef] [PubMed]
  122. Olive, K.P.; Jacobetz, M.A.; Davidson, C.J.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.A.; Caldwell, M.E.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461. [Google Scholar] [CrossRef] [PubMed]
  123. Onishi, H.; Katano, M. Hedgehog signaling pathway as a new therapeutic target in pancreatic cancer. World J. Gastroenterol. 2014, 20, 2335–2342. [Google Scholar] [CrossRef] [PubMed]
  124. Ebelt, N.D.; Zuniga, E.; Passi, K.B.; Sobocinski, L.J.; Manuel, E.R. Hyaluronidase-Expressing Salmonella Effectively Targets Tumor-Associated Hyaluronic Acid in Pancreatic Ductal Adenocarcinoma. Mol. Cancer Ther. 2020, 19, 706–716. [Google Scholar] [CrossRef]
  125. Wong, K.M.; Horton, K.J.; Coveler, A.L.; Hingorani, S.R.; Harris, W.P. Targeting the Tumor Stroma: The Biology and Clinical Development of Pegylated Recombinant Human Hyaluronidase (PEGPH20). Curr. Oncol. Rep. 2017, 19, 47. [Google Scholar] [CrossRef]
  126. Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; Von Hoff, D.D.; Hingorani, S.R. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012, 21, 418–429. [Google Scholar] [CrossRef]
  127. Ramanathan, R.K.; McDonough, S.L.; Philip, P.A.; Hingorani, S.R.; Lacy, J.; Kortmansky, J.S.; Thumar, J.; Chiorean, E.G.; Shields, A.F.; Behl, D.; et al. Phase IB/II Randomized Study of FOLFIRINOX Plus Pegylated Recombinant Human Hyaluronidase Versus FOLFIRINOX Alone in Patients With Metastatic Pancreatic Adenocarcinoma: SWOG S1313. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2019, 37, 1062–1069. [Google Scholar] [CrossRef]
  128. Van Cutsem, E.; Tempero, M.A.; Sigal, D.; Oh, D.Y.; Fazio, N.; Macarulla, T.; Hitre, E.; Hammel, P.; Hendifar, A.E.; Bates, S.E.; et al. Randomized Phase III Trial of Pegvorhyaluronidase Alfa with Nab-Paclitaxel Plus Gemcitabine for Patients with Hyaluronan-High Metastatic Pancreatic Adenocarcinoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 3185–3194. [Google Scholar] [CrossRef]
  129. Hakim, N.; Patel, R.; Devoe, C.; Saif, M.W. Why HALO 301 Failed and Implications for Treatment of Pancreatic Cancer. Pancreas 2019, 3, e1–e4. [Google Scholar] [CrossRef]
  130. Huang, X.; Lu, Y.; Guo, M.; Du, S.; Han, N. Recent strategies for nano-based PTT combined with immunotherapy: From a biomaterial point of view. Theranostics 2021, 11, 7546–7569. [Google Scholar] [CrossRef]
  131. Banerjee, S.M.; El-Sheikh, S.; Malhotra, A.; Mosse, C.A.; Parker, S.; Williams, N.R.; MacRobert, A.J.; Hamoudi, R.; Bown, S.G.; Keshtgar, M.R. Photodynamic Therapy in Primary Breast Cancer. J. Clin. Med. 2020, 10, 483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Wojewoda, K.; Gillstedt, M.; Tovi, J.; Salah, L.; Wennberg Larkö, A.M.; Sjöholm, A.; Sandberg, C. Optimizing treatment of acne with photodynamic therapy (PDT) to achieve long-term remission and reduce side effects. A prospective randomized controlled trial. J. Photochem. Photobiol. B 2021, 223, 112299. [Google Scholar] [CrossRef] [PubMed]
  133. DeWitt, J.M.; Sandrasegaran, K.; O’Neil, B.; House, M.G.; Zyromski, N.J.; Sehdev, A.; Perkins, S.M.; Flynn, J.; McCranor, L.; Shahda, S. Phase 1 study of EUS-guided photodynamic therapy for locally advanced pancreatic cancer. Gastrointest. Endosc. 2019, 89, 390–398. [Google Scholar] [CrossRef] [PubMed]
  134. Huggett, M.T.; Jermyn, M.; Gillams, A.; Illing, R.; Mosse, S.; Novelli, M.; Kent, E.; Bown, S.G.; Hasan, T.; Pogue, B.W.; et al. Phase I/II study of verteporfin photodynamic therapy in locally advanced pancreatic cancer. Br. J. Cancer 2014, 110, 1698–1704. [Google Scholar] [CrossRef]
  135. Miyoshi, N.; Kundu, S.K.; Tuziuti, T.; Yasui, K.; Shimada, I.; Ito, Y. Combination of Sonodynamic and Photodynamic Therapy against Cancer Would Be Effective through Using a Regulated Size of Nanoparticles. Nanosci. Nanoeng. 2016, 4, 1–11. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, R.; Liu, T.; Li, W.; Ma, Z.; Pei, P.; Zhang, W.; Yang, K.; Tao, Y. Tumor microenvironment-responsive BSA nanocarriers for combined chemo/chemodynamic cancer therapy. J. Nanobiotechnol. 2022, 20, 223. [Google Scholar] [CrossRef] [PubMed]
  137. Nabil, G.; Bhise, K.; Sau, S.; Atef, M.; El-Banna, H.A.; Iyer, A.K. Nano-engineered delivery systems for cancer imaging and therapy: Recent advances, future direction and patent evaluation. Drug Discov. Today 2019, 24, 462–491. [Google Scholar] [CrossRef] [PubMed]
  138. Kim, T.; Lee, N.; Arifin, D.R.; Shats, I.; Janowski, M.; Walczak, P.; Hyeon, T.; Bulte, J.W.M. In Vivo Micro-CT Imaging of Human Mesenchymal Stem Cells Labeled with Gold-Poly-L-Lysine Nanocomplexes. Adv. Funct. Mater. 2017, 27, 1604213. [Google Scholar] [CrossRef] [PubMed]
  139. Salvanou, E.A.; Kolokithas-Ntoukas, A.; Liolios, C.; Xanthopoulos, S.; Paravatou-Petsotas, M.; Tsoukalas, C.; Avgoustakis, K.; Bouziotis, P. Preliminary Evaluation of Iron Oxide Nanoparticles Radiolabeled with 68Ga and 177Lu as Potential Theranostic Agents. Nanomaterials 2022, 12, 2490. [Google Scholar] [CrossRef] [PubMed]
  140. Jang, H.M.; Jung, M.H.; Lee, J.S.; Lee, J.S.; Lim, I.C.; Im, H.; Kim, S.W.; Kang, S.A.; Cho, W.J.; Park, J.K. Chelator-Free Copper-64-Incorporated Iron Oxide Nanoparticles for PET/MR Imaging: Improved Radiocopper Stability and Cell Viability. Nanomaterials 2022, 12, 2791. [Google Scholar] [CrossRef]
  141. Hu, X.; Xia, F.; Lee, J.; Li, F.; Lu, X.; Zhuo, X.; Nie, G.; Ling, D. Tailor-Made Nanomaterials for Diagnosis and Therapy of Pancreatic Ductal Adenocarcinoma. Adv. Sci. 2021, 8, 2002545. [Google Scholar] [CrossRef] [PubMed]
  142. Huang, X.; Fan, C.; Zhu, H.; Le, W.; Cui, S.; Chen, X.; Li, W.; Zhang, F.; Huang, Y.; Sh, D.; et al. Glypican-1-antibody-conjugated Gd-Au nanoclusters for FI/MRI dual-modal targeted detection of pancreatic cancer. Int. J. Nanomed. 2018, 13, 2585–2599. [Google Scholar] [CrossRef] [PubMed]
  143. Wang, L.; Yin, H.; Bi, R.; Gao, G.; Li, K.; Liu, H.L. ENO1-targeted superparamagnetic iron oxide nanoparticles for detecting pancreatic cancer by magnetic resonance imaging. J. Cell. Mol. Med. 2020, 24, 5751–5757. [Google Scholar] [CrossRef] [PubMed]
  144. Zhou, H.; Qian, W.; Uckun, F.M.; Wang, L.; Wang, Y.A.; Chen, H.; Kooby, D.; Yu, Q.; Lipowska, M.; Staley, C.A.; et al. IGF1 Receptor Targeted Theranostic Nanoparticles for Targeted and Image-Guided Therapy of Pancreatic Cancer. ACS Nano 2015, 9, 7976–7991. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, J.; Cabral, H.; Song, B.; Aoki, I.; Chen, Z.; Nishiyama, N.; Huang, Y.; Kataoka, K.; Mi, P. Nanoprobe-Based Magnetic Resonance Imaging of Hypoxia Predicts Responses to Radiotherapy, Immunotherapy, and Sensitizing Treatments in Pancreatic Tumors. ACS Nano 2021, 16, 13526–13538. [Google Scholar] [CrossRef]
  146. Luo, X.; Hu, D.; Gao, D.; Wang, Y.; Chen, X.; Liu, X.; Zheng, H.; Sun, M.; Sheng, Z. Metabolizable Near-Infrared-II Nanoprobes for Dynamic Imaging of Deep-Seated Tumor-Associated Macrophages in Pancreatic Cancer. ACS Nano 2021, 15, 10010–10024. [Google Scholar] [CrossRef] [PubMed]
  147. Frelaut, M.; Le Tourneau, C.; Borcoman, E. Hyperprogression under Immunotherapy. Int. J. Mol. Sci. 2019, 20, 2674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The tumor immune microenvironment (TIME) of PDAC.
Figure 1. The tumor immune microenvironment (TIME) of PDAC.
Pharmaceutics 14 02033 g001
Table 2. Nanoparticle-based therapeutic strategies for enhanced PDAC immunotherapy.
Table 2. Nanoparticle-based therapeutic strategies for enhanced PDAC immunotherapy.
StrategyAgent/DrugNanoparticleAdministrationImmune EffectPublication YearRef.
Nano-based ICDoxaliplatin amphiphilic diblock copolymer nanoparticlesivDAMPs↑;
DCs↑;
CD3+CD8+ cytotoxic T lymphocytes↑
2016[79]
oxaliplatinsilicasome nanocarrierivCD8+/FoxP3+ T cell ratios↑;2021[80]
ingenol-3-mebutate (I3A)2-(3-((S)-5-amino-1-car_x0002_boxypentyl)-ureido) pentanedioate (ACUPA− ) and triphenylphosphonium (TPP+) modified nanomicellesivDCs↑;
CD8+ T cells↑;
CD4+ T cells↑
2021[81]
Nano-based immune modulationM2 peptides + miR-125bhyaluronic acid-poly(ethylene imine) nanoparticlesipM1-to-M2 macrophage ratio↑;2021[82]
PI3K-γ inhibitor NVP-BEZ 235 + CSF-1R-siRNA nanomicelle (M2 TAMs targeting
nanomicelles)
ivM2-TAMs↓;
MDSCs↓;
M1-TAMs↑;
CD8+ and CD4+ T cell infiltration↑;
2020[83]
monoacylglycerol lipase siRNA + endocannabinoid receptor-2 (key receptor regulating macrophage phenotype) siRNA reduction-responsive poly (disulfide amide) (PDSA)-based nanoplatformivrepolarization of TAMs into tumor-inhibiting M1-like phenotype2021[84]
paclitaxel-loaded 3-aminophenylboronic acid_x005f_x0002_modified low molecular weight heparin–D-α-tocopheryl succinatemicellar nanoparticleivinfiltration of CD4+ T cells and CD8+ T cells↑;
MDSCs ↓
2021[85]
cyclic diguanylate monophosphate (cdGMP), an agonist of the stimulator of interferon genes (STING) pathway + monophosphoryl lipid A (MPLA), a Toll-like receptor 4 (TLR4) pathway agonist immuno-nanoparticlesivCD45+ immune cells↑;
DC cells↑;
macrophages↑
2021[86]
IL-10 trap + CXCL12 trapliposome-protamine-DNAipM2 macrophages↓;
MDSCs↓;
PD-L1+ cells↓;
Immuno suppressive plasma cells (ISPCs)↓;
activated DCs↑;
NK cells↑;
CD8+ T cells in tumor↑
2018[87]
CXCL12 trap + PD-L1 trapliposome-protamine-DNAivMDSCs↓;
Treg cells↓;
accumulated macrophages↓;
M1/M2 ↑;
T cell infiltration↑
2017[88]
Bcl2 double-stranded RNA (dsRNA) + Retinoic acid-inducible gene I (RIG-I)-like receptors lipid calcium phosphate nanoparticlesivTh1 cytokines↑;
CD8+ T cells/Treg cells↑;
M1/M2 ↑;
immunosuppressive B regulatory cells↓
2019[89]
IL-12+SBRTmicrospheresintratumoral injectionupregulation of Th1 and antitumor factors IL-12, IFN-γ, CXCL10, and granzyme B 2020[90]
PD-L1poly(lactic-co-glycolic acid;PLGA)-based siRNA nanoparticle ivIFN-gamma positive CD8+ T cells↑;
Granzyme B+ cell↑;
2021[91]
oxygenmicrocapsulesintratumoral injection by
ultrasound-guided percutaneous injection
oxygen microcapsules + anti-PD-1 antibody:
the infiltration of TAMs and polarize pro-tumor M2↓;
macrophages into anti-tumor M1 macrophages;
the proportion of Th1 cells↑;
CTLs↑
2022[62]
DCs vaccinealuminum hydroxide nanoparticle with polyethyleneimine (PEI) modification (LV@HPA/PEI).subcutaneously vaccinated CD3+ CD8+ IFNγ+ T cells↑;2018[92]
Nano-based chemo-immunotherapyIDO inhibitor, indoximod (IND)+ oxaliplatin (OX)mesoporous silica nanoparticles(MSNP)ivCD8+/Tregs↑;
DC maturation↑;
cytotoxic T lymphocytes↑;
Foxp3+ T cells(Tregs)↓;
2017[93]
IDO1 siRNA+oxaliplatincationic lipid-assisted nanoparticlesivDC maturation↑;
tumor-infiltrating T lymphocytes↓;
Treg↓;
central memory T cells (TCM)↑;
effector memory T cells (TEM)↑;
2019[94]
GEM + NLG919 (IDO1 inhibitor) + paclitaxel micellesivCD4+ IFNγ+ T cells↑;
CD8+ IFNγ+ T cells↑;
Treg cells↓
2020[95]
gemcitabinepH-sensitive polymerip and ivdown-regulated the infiltration of macrophages in the tumor tissue;
up-regulated the PD-L1 expression on the surface of cancer cells;
MDSC↓;
CD3+T cells↑;
CD8+ T cells↑;
CD4+ T cells↑;
TAMs↓;
Tregs↓;
2021[96]
GEM + signal transducer and activator of transcription 3 (STAT3) inhibitor (HJC0152)micelleivreversing M2 to M1-TAMs, M1/M2↑;
downregulating MDSCs/Tregs, and upregulating Teff
2022[97]
celastrol (CLT) + 1-methyltryptophan (MT, IDO inhibitor) hyaluronic acid coated cationic albumin nanoparticleivCD4+ T cells in the spleen↑;
CD8+ T cellsin the spleen↑;
2019[98]
irinotecan+Anti-PD-1 antibodysilicasomeiv(irinotecan) + ip(anti-PD-1)irinotecan by the silicasome:
CD8+ T cells/Treg cells↑;
PD-L1 expression↑;
2021[99]
galectin-9 siRNA + oxaliplatin exosomeivM2-TAMs↓;
Tregs↓;
M1-TAMs↑;
cytotoxic T cells and helper T cells↑;
CD8+ CTLs↑;
mature DC ↑;
2020[22]
gemcitabine-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles + M2-like macrophages peptides (M2pep) +PD-L1 antibodybiomimetic dual-targeting nanomedicine ivelimination of PD-L1-positive macrophages and the downregulation of PD-L1 expression; reprogrammed macrophages, downregulated
PD-L1 expression, and sustained T cell populations,
2022[100]
Nano-based stoma modulationα-mangostin + LIGHT (tumor necrosis factor superfamily 14)calcium phosphate liposomeivCD45+CD3+CD8+ T cells↑;
CD45+CD3+CD4+ T cells ↑;
CD45+B220+ B cells↑;
Tregs and F4/80+ macrophages↓;
CD8+ T/CD4+ T↑;
CD4+T/Treg ↑;
induces the intratumoral tertiary lymphoid structures
2020[101]
TGF-β receptor inhibitors (LY2157299) + siRNA targeting PD-L1 (siPD-L1) acidic tumor extracellular pH (pHe) responsive clustered nanoparticleivtumor infiltrating CD8+ T cells↑;
IFN-γ↑;
2020[102]
sonic hedgehog inhibitor (cyclopamine) + cytotoxic chemotherapy drug (paclitaxel)polymeric micellesivinfiltration CD8+T cells↑;2018[103]
PEGPH20+GVAXPEGylated recombinant human PH20 hyaluronidaseivinfiltrating CD3+CD8+ T cells↑;
infiltrating CD3+CD4+ T cells↑
2019[104]
PEGPH20+FAKinhibitor+anti-PD-1 antibodyPEGylated recombinant human PH20 hyaluronidaseiv + oral gavage + ip Increases T cell
infiltration and alters T cell phenotype towards effector memory T cells.
2022[105]
paclitaxel + phosphorylated gemcitabinecodelivery micelles ivcytotoxic T cells↑;
T helper cells↑;
Tregs cells↓;
tumor infiltration by the CD4+ and CD8+ T cells↑;
2019[106]
CXCR4 antagonist + anti-miR-210 + siKRASG12D cholesterol-modified polymericip CD8+ T cells infiltration↑;
M2 TAMs↓;
2020[107]
Nano-based PTT/PDT/CDT/SDTmild hyperthermia + immune checkpoint blockade (BMS-202) size-adjustable thermo-and fibrotic matrix-sensitive liposomes iv + laser irradiationDC maturation↑;
infiltration of CD4+ T cells↑;
infiltration of CD8+ T cells↑;
Tregs↓;
chemokines (IL-6, IFN-γ) ↑
2021[70]
immunomodulatory thymopentin + near-infrared indocyanine greenself-assembly nanoparticlein situ injection + laser irradiationCD3+CD4+ T cells↑;
CD3+CD8+ T cells↑;
CD4+IL-4+ T cells ↑;
CD8+INF-γ+ T cells↑;
2021[71]
GEM+NLG919 (IDO inhibitor)polydopamine (dp)-coated nanoparticlesiv + laser irradiationintratumoral infiltration of CD8+ T↑;
(IFN-γ) in CTLs↑;
GZB positive NK cells↑
2021[72]
indocyanine green (ICG,photothermal agent) + imiquimod (IMQ,toll-like-receptor-7 agonist)iron oxide nanoplatformiv + laser irradiationpolarization of macrophages to the M1 phenotype;
tumor infiltration of T cells↑;
Tregs↓;
CD8+ T cells/Treg cells↑;
CD4+ T cells/Treg cells↑
2022[73]
bromodomain-containing protein 4 inhibitor (BRD4i) JQ1+ cyclodextrin-grafted hyaluronic acid (HA-CD) + pyropheophorbide a (PPa)supramolecular prodrug nanoplatformiv + laser irradiationDC maturation↑;
intratumoral infiltration of CD8+ T ↑;
intratumoral infiltration of Tregs ↓
memory T lymphocytes (TEM) infiltration↑
2021[74]
chlorin e6 photosensitizertumor-derived exosomesiv + laser irradiationrelease of cytokines from immune cells2021[75]
Rose Bengal+ CaO2 nanoparticle pH-sensitive polymethacrylate-coated CaO2 nanoparticleiv + ultrasoundCD8+T cells↑;
Treg↓
2021[76]
cavitation-assisted endoplasmic reticulum targeted sonodynamic droplets+PD-L1 antibodynanodropletsiv + ultrasoundDC maturation↑ 2022[77]
4-(phosphonooxy)phenyl-2,4-dinitrobenzenesulfonate + Fe3 + hyaluronic acid decorationchemodynamic nanocomplexivinduced the polarization of the M2 phenotype back to the M1 phenotype2020[78]
iv: intravenous administration; ip: intraperitoneal administration; ↑: increase in number; ↓: decrease in number.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hou, W.; Yang, B.; Zhu, H. Nanoparticle-Based Therapeutic Strategies for Enhanced Pancreatic Ductal Adenocarcinoma Immunotherapy. Pharmaceutics 2022, 14, 2033. https://doi.org/10.3390/pharmaceutics14102033

AMA Style

Hou W, Yang B, Zhu H. Nanoparticle-Based Therapeutic Strategies for Enhanced Pancreatic Ductal Adenocarcinoma Immunotherapy. Pharmaceutics. 2022; 14(10):2033. https://doi.org/10.3390/pharmaceutics14102033

Chicago/Turabian Style

Hou, Wanting, Biao Yang, and Hong Zhu. 2022. "Nanoparticle-Based Therapeutic Strategies for Enhanced Pancreatic Ductal Adenocarcinoma Immunotherapy" Pharmaceutics 14, no. 10: 2033. https://doi.org/10.3390/pharmaceutics14102033

APA Style

Hou, W., Yang, B., & Zhu, H. (2022). Nanoparticle-Based Therapeutic Strategies for Enhanced Pancreatic Ductal Adenocarcinoma Immunotherapy. Pharmaceutics, 14(10), 2033. https://doi.org/10.3390/pharmaceutics14102033

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop