Next Article in Journal
Design of Liquid Formulation Based on F127-Loaded Natural Dimeric Flavonoids as a New Perspective Treatment for Leishmaniasis
Previous Article in Journal
Development of a Versatile Lipid Core for Nanostructured Lipid Carriers (NLCs) Using Design of Experiments (DoE) and Raman Mapping
Previous Article in Special Issue
Correction: Kalvala et al. Cannabidiol-Loaded Extracellular Vesicles from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Paclitaxel-Induced Peripheral Neuropathy. Pharmaceutics 2023, 15, 554
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress

1
Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
2
Department of Orthopedics, Hunan Loudi Central Hospital, Loudi 417000, China
3
Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China
4
Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2024, 16(2), 251; https://doi.org/10.3390/pharmaceutics16020251
Submission received: 13 November 2023 / Revised: 20 January 2024 / Accepted: 31 January 2024 / Published: 8 February 2024
(This article belongs to the Special Issue Functionalized Nanoparticles in Cancer Therapeutics)

Abstract

:
Immunosuppressive elements within the tumor microenvironment are the primary drivers of tumorigenesis and malignant advancement. The presence, as well as the crosstalk between myeloid-derived suppressor cells (MDSCs), osteosarcoma-associated macrophages (OS-Ms), regulatory T cells (Tregs), and endothelial cells (ECs) with osteosarcoma cells cause the poor prognosis of OS. In addition, the consequent immunosuppressive factors favor the loss of treatment potential. Nanoparticles offer a means to dynamically and locally manipulate immuno-nanoparticles, which present a promising strategy for transforming OS-TME. Additionally, chimeric antigen receptor (CAR) technology is effective in combating OS. This review summarizes the essential mechanisms of immunosuppressive cells in the OS-TME and the current immune-associated strategies. The last part highlights the limitations of existing therapies and offers insights into future research directions.

1. Introduction

Osteosarcoma, the most prevalent malignant bone-related cancer in adolescents, poses a significant treatment challenge [1]. The survival rate for this condition over a five-year period ranges from 60% to 70%, and it is more frequently observed in males and individuals of African American descent [2]. Despite extensive research efforts, 5-year survival rates for osteosarcoma patients have remained around 20% in recent decades [3]. What is worse, the prognosis for patients with metastatic or relapsed osteosarcoma has remained bleak and stagnant over the past 30 years [4]. The current management strategy for primary OS is surgery plus neoadjuvant chemotherapy (preoperative treatments), and the survival rate has been discovered as 35–40% [5,6]. Rotation-plasty, a well-established method for knee tumor reconstruction, can yield functional and psychological outcomes equal or superior to endoprosthetic reconstruction [7,8]. However, it presents cosmetic challenges [7,9]. Axial skeleton sarcoma surgery is complex due to high local recurrence risk and frequent reconstruction complications [7]. About 30% of primary metastatic osteosarcoma patients and over 40% achieving complete remission can be long-term survivors [10]. Lung metastases in advanced OS patients decrease the 5-year survival rate to 5–10%; in this case, the adjuvant combination of chemotherapy and immunotherapy significantly enhances survival rates. Despite this progress, the immunotherapy of OS still requires in-depth investigation.
The tumor microenvironment (TME) is a complex system composed of not only cancer cells, but also various cell types, including immune cells such as myeloid-derived suppressor cells (MDSCs), osteosarcoma-macrophage (OS-M), and stromal cells like the cancer-associated fibroblast [11]. Among these, MDSCs are generated in the bone marrow, and in the context of cancer (tumor-bearing hosts), they further migrate to peripheral lymphoid organs and the tumor site [12] and exert immune suppressive activity [13], inhibiting the functions of cytotoxic T cells and natural killer cells [14]. Moreover, the tumor microenvironment in osteosarcoma (OS-TME) houses essential macrophage populations, which can be categorized into two main phenotypes: the classically activated macrophages, often referred to as Type-1 (OS-M1), and the alternative Type-2 macrophages (OS-M2) [15]. Within the OS-TME, the prevailing majority of macrophages primarily display the pro-tumor Type-2 phenotype, lacking anti-tumor functionalities [16]. Osteosarcoma-associated neutrophils also participate in the OS-TME [17]. Initially, neutrophils are responsible for immune attack; however, the OS educates the neutrophils to become tumor-promoting neutrophils [18]. Regulatory T lymphocytes (Tregs) in the immunosuppressive or cold OS-TME also matter, favoring OS progression and metastasis [19]. To effectively treat OS, it is essential to alter the OS-TME.
Traditional intervention approaches, such as surgical resection and chemotherapy, are primary strategies against OS. Cancer immunotherapy has surfaced as an important avenue for the improvement in OS-TME, benefiting the inhibition of metastasis and recurrence of OS [20]. For the therapy associated with the OS-M, we divided current immunotherapy for regulating the OS-M into two types: conventional immunotherapeutic and novel engineered cell therapy [21,22]. The conventional approaches encompass the inhibition of recruitment of tumor-promoting OS-M, the impairment of OS-M, and the recovery of OS-M phagocytosis function [23]. Nanotechnology is a significant platform for the therapeutics of modulating the OS-TME. Nanoparticles offer multiple advantages, including the ability to be intricately engineered for the robust and precise activation of T cells before their adoptive transfer [24]. Additionally, Nps can be used to provide the added functionality of immunotherapy [25,26]. Thus, nanoparticles present a promising solution to address the challenges associated with T cell therapy [27,28]. To enhance the accumulation of therapeutics at tumor sites, it is crucial for nanocarriers (NCs) to exhibit prolonged circulation, a characteristic achievable through surface modification with hydrophilic polymers like polyethylene glycol (PEGylation) [29]. Additionally, nanoparticles can also encapsulate agents with a size range of 50–100 nm and can effectively enter parenchymal hepatic cells, stimulating T cell activity [30]. Nanocarriers with a size less than 50 nm can penetrate the cellular barriers, promoting nanoparticle distribution into the bone lesion [31].
Another type of immunotherapy focus is on enhancing the natural ability of adaptive immune cells, particularly cytotoxic T lymphocytes (CTLs) [32]. Adoptive T cell therapy stands as a promising frontier in the realm of cancer treatment [33,34]. Clinical trials have provided compelling evidence of its potential in the context of both adult and pediatric populations, demonstrating the effectiveness of advanced immunotherapy in combatting bone cancer [23,35]. The impact of this therapy has been further underscored by the approval of three notable T cell-based therapeutics—Kymriah, Yescarta, and Breyanzi—by the US FDA [36,37,38]. However, though there has been considerable success in hematologic malignancy treatment, including B cell leukemia and lymphoma treatment, adoptive T cell therapy faces inherent challenges that limit its efficacy in the treatment of many solid tumors including OS [39]. The underlying reason includes the difficulty of T cell infiltration towards the solid tumor because of extensive surrounding stromal cells around OS [40]. To overcome this barrier, one of the approaches is to eliminate the stromal cells with other therapeutics, and nanoparticles can serve as efficient carriers for these therapeutics. Moreover, for future CAR T therapies, it is important to incorporate “on or off switches” that ensure CAR T efficacy in the tumor lesion [41]. Additionally, the serious side effects of CAR T such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) also hinder the success of CAR T [42,43]. Therefore, the combinations of anti-cytokine therapy or synergistic therapeutics to reduce the dose of CAR T will benefit CAR T cell application. In addition, it is reported that a third-generation GD2-CAR exerted effective recognition for the GD2+ sarcoma cell lines in vitro [44]. However, these GD2-CAR T cells were unsuccessful in an in vivo xenograft tumor model [44]. Similarly, it is significant to monitor the HER2 CAR T cell potency using a xenograft or validated model [45]. All finding indicated that the combinational strategies is significant. Another approach enhancing cancer specificity and clinical response is represented by a bispecific CAR T cell molecule [46,47,48].
In this review, we delve into the role of immunosuppressive cells within the OS-TME at first. Then we emphasize the pressing need for continued research and innovative strategies to address these challenges and expand the potential of T cell therapy in osteosarcoma. Additionally, we acknowledge the limitations that may hinder the viability of T cell therapy as a treatment option for this formidable bone malignancy. This perspective underscores recent advancements in preclinical research that leverage nanoparticles to augment the effectiveness of T cell therapy.

2. Immunosuppressive Osteosarcoma Microenvironment

Osteosarcoma is known for its high heterogenicity and low tumor immunogenicity. The extensive immune cell infiltration in the OS-TME leads to the formation of a niche for OS proliferation, metastasis, and resistance [17,49]. This immunosuppressive OS-TME is correlated with the presence of MDSCs (Myeloid-Derived Suppressor Cells), OS-Ms (osteosarcoma-associated macrophages), ECs (endothelial cells), and Tregs (regulatory T lymphocytes) [50]. Among them, MDSCs are a heterogeneous population of immune cells that play a crucial role in suppressing the anti-tumor immune response [51]. OS-M refers to tumor promoting macrophages, which, in general, contribute to tumor progression via promoting angiogenesis, tissue remodeling, and immunosuppression. ECs (endothelial cells) are responsible for the supply of nutrients [52,53]. Regulatory T lymphocytes (Tregs) is beneficial for the immune surveillance [19].

2.1. MDSC

As per the brief discussion on MDSCs in the introduction, MDSC is significant component in the OS-TME. Even in the initial stage, MDSC contributes to the pathogenesis of OS through several mechanisms [14]. Firstly, MDSCs hinder T cell migration and reduce T cell viability, making T cell access to the OS more difficult [54]. Furthermore, MDSCs alter T cell fitness by the production of immune-inhibitory molecules like nitric oxide (NO), reactive oxygen species (ROS), and reactive nitrogen species (RNS). Additionally, MDSC reduces T cell-mediated immune responses via the consumption of L-arginine [55]. Lastly, MDSCs consume vital metabolites necessary for T lymphocyte fitness, further compromising the immune response [56]. Additionally, MDSCs can further migrate to peripheral lymphoid organs, resulting in antigen-specific T cell tolerance, and contributing to the metastasis of OS [12]. Firstly, MDSCs play a role in promoting tumor angiogenesis through the secretion of factors like vascular endothelial growth factor (VEGF) and matrix metalloproteinase 9 (MMP9) [57]. Both of them support the growth of micro-vessels within the tumor and aid the tumor’s expansion [58]. MDSCs also secrete elevated levels of transforming growth factor-beta (TGF-β) and hepatocyte growth factor (HGF) to benefit the growth of OS in other distant organs [59]. The existing TGF-β and HGF induce epithelial–mesenchymal transition (EMT), a process that enhances the tumor’s ability to invade and metastasize [60]. In the metastatic niche, MDSCs secrete a molecule called versican to contribute to the establishment of metastatic tumor growth [61].

2.2. OS-M

Besides MDSCs, in the OS-TME, OS-M functions as a mutineer [62]. In the initial harsh OS-TME characterized by factors such as hypoxia, low pH, elevated glutathione (GSH) levels, and dysregulated kinase systems [63,64], OS cancer cells educate macrophages to adopt tumor-proliferation-supportive roles. Then, as a feedback, the infiltration of OS-M further aggravates the PD-L1 expression in OS, negatively impacting the cytotoxicity of T cells [65,66]. Furthermore, the hypoxic environment in OS-TME promotes tumor angiogenesis [67,68]. In addition, tumor cells also release signals like IL12 and IL4, along with hypoxia-inducible factors HIF-1α and HIF-2α, to maintain OS-M education and support the dysfunction of DCs [69]. OS-M elevates the levels of vascular endothelial growth factor as well as matrix metalloprotease 9 [70]. This facilitates angiogenesis and the formation of a pre-metastatic niche, demonstrating a strong association with osteosarcoma metastasis [71].

2.3. Endothelial Cell

Endothelial cells (ECs) play a role in promoting the acquisition of tumor cell properties, including cell growth, invasion, metastasis, and chemoresistance [72,73]. EC proliferation is associated with nutrient supplies for the OS-TME [74]. It is identified that cyclin-dependent kinase 2 and 5 (Cdk2, Cdk5) serve as key mediators of neo-angiogenesis [75,76]. Additionally, a specific signal named Yin Yang 1 (YY1) protein from osteosarcoma (SaOS) cells plays a crucial role in driving the proliferation of human aortic endothelial cells (HAECs) [76]. In addition to resident endothelial cells, there are circulating ECs. Elevated levels of circulating endothelial cells (CECs) have been found in the peripheral blood of OS patients compared to control groups [77,78]. On the contrary, circulating endothelial progenitor cells (CEPs) are cells derived from the bone, specifically contributing to tumor-associated vasculogenic effects [79]. Additionally, ECs function as both modulators and effectors in the context of OS, contributing to the acceleration of OS exacerbation through the release of von Willebrand factor (VWF) [80].

2.4. Treg

Treg cells represent a dynamic subset of CD4+ T lymphocytes that regulate both normal and aberrant immune system responses [81,82]. Tregs in the TME play critical roles in enabling tumor cells to evade immune surveillance [83]. Some important molecules associated with Tregs will be discerned in this part. For instance, CD39, an ectonucleotidase elevated on Treg cell surfaces, facilitates immunosuppression [84,85]. CD39 converts adenosine triphosphate to adenosine; the subsequent biological binding of adenosine to the A2A receptors (A2AR) and/or A2B receptors (A2BR) has a negative impact on the functions of natural killer and dendritic cells in the TME [85,86,87]. What is worse, adenosine potentiates the expansion of tumor promoting cells, including MDSCs and OS-M2 [87]. Additionally, Tregs secrete perforin and granzymes affecting effector T cells [88,89]. The neuropilin-1 (Nrp1) semaphorin-4a (Sema4a) axis is a newly found factor associated with Tregs in the TME [90]. In addition, CTLA-4 on Tregs causes the direct suppression of the APC function of DCs and hampers the abilities of effector T cells [91]. Thus, the manipulation of Treg function through therapeutic interventions has become a promising strategy.

2.5. OS-Ns

OS-Ns refers to osteosarcoma-related neutrophils [92,93]. OS-Ns has phenotypic heterogeneity and functional versatility. In osteosarcoma, research on TANs is still in its early stages. The lifespan of OS-Ns is longer than that of circulating neutrophils [94,95]. Liu et al. used a meta-analysis to examine the possible correlation between matrix metalloproteinases -9 (MMP-9) mediated by OS-N and a poor prognosis for patients [94,95]. The higher the level of matrix MMP-9 expression, the higher the poor-prognosis risk of patients with OS [94]. Of note, this study faced challenges from another researcher, and thus more studies are essential to validate the prognostic value of MMP9 in OS [94,96]. Neutrophil extracellular traps are web-like chromatin structures formed by the granule proteins and chromatin generated by OS-Ns, which contribute to metastasis, and the underlying mechanism is associated with the DNA receptor coiled-coil domain which includes protein 25 [97]. Another significant chromatin that forms OS-N extracellular traps is peptidylarginine deiminase 4 which overexpressed on OS [98]. Furthermore, the infiltration of neutrophils promotes the translation of hypoxia-related genes, resulting in an increasingly hypoxic microenvironment [99]. A hypoxic TME is not beneficial for the efficacy of anti-cancer therapy [100,101]. It is also deserve to mention that the predominant OS-N in the OS lesion significantly increases the immune escape evasion of OS cells [99].

3. Interactions of Tumor Promoting Cells in the OS-TME

Osteoimmunology is a new term used to describe the immune TME in bone-related disease. The abovementioned immune cells facilitate the evasion of immune attack via immunoediting or other escape mechanisms [102,103]. The OS cell itself is able to downregulate human immunity or achieve immune escape via PDL1/L2 [104], B7-H3 [105], HHLA2 [106], or MHC class II [52,107]. In addition, OS cells release abundant VEGF that interacts with VEGFRs in the endothelial cells (ECs), benefiting angiogenesis and facilitating OS nutrient supply [108].
OS cells release TGF-beta to increase the ratio of Treg in the OS-TME [17]. MDSCs release IL-10 and TNF-alpha, which decrease the activity of cytotoxic T lymphocytes (CTLs) [109]. In the OS-TME, after neo-adjuvant chemotherapy, elevated cytotoxic lymphocyte TILs are associated with a decrease in MDSCs [110]. OS-Ms generally exert pro-tumoral function and show a high correlation with OS aggressiveness and poor prognosis [110,111]. In the OS-TME, OS-M also benefits the formation of neo-vessels via the interaction of the VEGF/VEGF receptor [52]. CTL is responsible for the killing of OS; however, the development of immune surveillance pathways, including the PD-L1/L2-PD1 andCTLA-4 [112], T cell immunoglobulin and mucin-domain-containing-3 (TIM-3) as well as lymphocyte activation gene-3 (LAG-3) [113], ensures the immune escape of OS [62]. Recent preclinical studies have evidenced the TME-promoting role for Tregs in OS [114]. Yoshida. et al. reported that the decrease in Tregs is paralleled by the increase in TILs in the OS-TME [115]. Tregs negatively impact the cytotoxic activity of T cells via TIM-3 as well as LAG-3 [113]. Immunoglobulin and tyrosine-based inhibitory motif (ITIM) domain (TIGIT) on T cells have surfaced as immune targets [116]. All cells actively interact with each other and form an immunosuppressive OS-TME (Figure 1).

4. Conventional Nanoparticle-Based Therapy and Engineered Immunotherapy for OS-M

OS-dominated tumor-promoting macrophages possess characteristics suppressing immunity and promoting tumor development [117,118]. Thus, it is of importance to treat OS-M. Given the massive investigations associated with macrophages, we have introducted nanoparticles for OS-M in one section and will discuss the applications of nanoparticles for other immune cells in OS-TME later. Traditional approaches for the OS-M include three approaches. Firstly, reducing the source of OS-M using nanoparticles containing inhibitors of recruitment pathways such as CSF1 [119], CCL2 [120], CCL5 [121], and CXCL12 [122] pathways. By inhibiting these pathways, monocyte recruitment into osteosarcoma lesions can be decreased, consequently reducing the differentiation of resident OS-Ms. Secondly, nanoparticles carry therapeutics that interfere with the function of tumor-promoting OS-Ms. For instance, in one study, when treating OS with graphene oxide with PEG modification, pro-tumoral OS-M2 was suppressed, consequently resulting in the regression of bone cancer [123]. Thirdly, certain nanoparticles, either alone or conjugated with antibodies, have demonstrated the ability to restore macrophage phagocytosis. For instance, siRPa-CD47 blockage can be achieved using nanoparticle-based approaches [124]. Other nanoparticles potentially impacting MCHII and PD1/PDL1 pathways can also recover phagocytic abilities [20,125,126]. Furthermore, the combination of CD47 mAb and ferumoxytol-loaded nanostructures can enhance the anti-tumor OS/M1 ratio [127]. CTLA-4 (aka CD152) is regarded as overexpressed in the OS-TME [128]. The synergistic application of CTLA-4 inhibitors with nanoparticles provides insights for OS treatment [129]. In addition, PD-1/PD-L1 is reported to negatively correlate to the survival rate, and this pathway has emerged as a target in osteosarcoma [130]. The blockade of PD-1 using pembrolizumab leads to the inhibition of osteosarcoma [131,132]. Because macrophage is a type of immune cell, so the therapy targeting macrophage can be regarded as conventional immunotherapy.
Additionally, CAR-M technology presents a novel therapeutic approach for modifying tumor-promoting macrophages into anti-tumor macrophages, thereby enabling macrophages to recognize combat cancer [133]. Because although conventional therapies are promising in modulating OS-M, the effects cannot be maintained for a long time due toa defense mechanism developed by OS [134]. Thus we need immune cell that has memory. CAR-based therapy has been regarded as “living drug” [135]. Chimeric antigen receptor macrophage (CAR-M) cells has gained prominence in the solid tumor field [136]. However, it need to point out, currently, there is no promising progress for CAR-M in OS treatments. Still, we would like to introduce CAR-M and hopefully it will provide insights for future researchers. In CAR-M therapy, isolated patient immune cells (monocytes) are engineered to express chimeric antigen receptors and then are infused back to the patient [137]. Consequently, the CAR modification can help macrophages to discern cancer cells, making modified macrophages a promising tool in the fight against osteosarcoma [138,139]. The initial efforts to introduce CARs into macrophages were pioneered by Biglari and colleagues in 2006, in which a chimeric CD64 molecule was developed, comprising a single-chain variable fragment (scFv) designed to target the human carcinoembryonic antigen (CEA), along with the transmembrane and cytoplasmic domains of human CD64, resulting a significant reduction in the tumor growth rate in a xenotransplantation model [140]. Additionally, the CAR-M technology presents a novel therapeutic approach for enhancing the macrophages’ ability to phagocytose cancer cells and present antigens, thereby enabling macrophages against cancer [125]. Engineered CAR-Ms have resulted in increase of anti-tumor cytokines, such as IL6, and chemokines, such as CXCL18, in the OS-TME [141,142]. The production of these beneficial cytokines fosters a shift from a cold OS-TME towards a hot OS-TME [143]. Nothing is perfect, it also should pay attention to the levels of IL6, massive IL6 can cause cytokine release syndrome [144]. Alternatively, other investigators used computational modeling tools to anticipate the performance of a newly designed CAR molecule capable of facilitating bispecific antibodies (Figure 2).

5. Advanced Immunotherapy—Chimeric Antigen Receptor (CAR)-T Cell Therapy

5.1. Chimeric Antigen Receptor (CAR) T Cell Immunotherapy

In addition to the conventional NP-associated therapies, the successful implementation of chimeric antigen receptor (CAR) T cell immunotherapy in the treatment of hematological malignancies proves to be a possible strategy worth exploring in the context of OS [145]. CAR-modified T cells utilizing the immune system’s potent cytotoxic mechanisms against tumor cancer cells have shown promising progress [146]. Nevertheless, the development of successful CAR T cell therapies for solid tumors encounters challenges. [147,148] Thus, combinational therapy has been considered as a more promising approach. It is reported that OS prominently expresses the GD2 antigen, which has been identified as a viable immunotherapeutic target [149]. In addition, researchers have evaluated the potential of CAR-modified T cells targeting GD2 to induce cytotoxicity against OS tumor cells [150,151]. It is demonstrated that GD2 CAR-modified T cells exhibited high efficacy in inducing cell death in OS tumor cells [151]. However, the OS cells gradually developed the ability to escape the attack of GD2 CAR-modified T cells via PD-L1 expression in OS cells [152]. The PDL1–PD1 interaction gives the wrong signal to CAR T cells, stopping the activity of CAR T cells and finally inducing CAR T cell apoptosis [153]. A cyclic peptide, CGRRAGGSC (using the single-letter amino acid code), displayed on an IL-11 mimic phage exhibited specific binding to immobilized IL-11Rα [154,155].
Importantly, IL-11Rα has been recognized as a target for OS molecular focus in CAR T cell studies [152]. In a study conducted by Moonat, Hatel et al., engineered CAR T cells, using IL-11Rα as the tumor-specific immunoreceptor, exhibited enhanced cytotoxicity against multiple OS cell lines [156]. Moreover, there have been investigations into the use of γδ T cells in the context of osteosarcoma [157,158]. Interestingly, zoledronic acid enhances the susceptibility of rhabdomyosarcoma cells to destruction by human γδ T cells [159]. Additionally, HER2-specific CAR T therapy has been explored in mouse models of osteosarcoma. phA2 CAR T cells effectively target and eliminate EphA2-positive osteosarcoma and Ewing’s sarcoma cells in vitro and induce complete tumor regression in subcutaneous xenograft models [160]. In vivo, direct injection into established tumors extends survival, while systemic delivery requires a higher cell dose. In murine osteosarcoma, a strategy involves combining CTLA-4 blockade either with tumor lysate-pulsed dendritic cells (DCs) or PD-L1 blockade therapeutics [160]. Building on this, studies have confirmed that the co-administration of lenalidomide with EGFR vIII-CAR T cells enhances F-actin polymerization, CAR T cell infiltration, and cytotoxicity at the tumor site in a murine glioblastoma model, ultimately extending the survival of treated mice [136]. CD28-based (NCT00902044) and CD28-CD3z-OX40 CAR T cell therapy (NCT01953900) have been used in sarcoma patients in clinical trials. Moreover, a trial investigating the combination of CAR T cells with low-dose chemotherapeutic agents is still ongoing (NCT04433221). More information associate with CART please refer to Table 1. Because there is no commercial product specific for OS, thus we summarized the progression of CART in liquid cancer.
The successful infiltration of modified T cells is significant and nanoparticles can be useful in tracking CAR T. For instance, iron oxide nanoparticles offer a clinically applicable method for tagging CAR T cells, facilitating the noninvasive tracking of these labeled cells through magnetic particle imaging (MPI), photoacoustic imaging (PAT), and MRI. This enables the identification of their distribution within the body [161]. In this study, T lymphocytes labeled with the Fe2O3 nanoparticles were found in osteosarcoma [161]. This study explores the monitoring of CAR T cells with ferumoxytol, opening up the possibility of detecting CAR T cells in solid tumors [161].
Table 1. The examples of successful CAR T therapy in liquid cancer and solid cancer.
Table 1. The examples of successful CAR T therapy in liquid cancer and solid cancer.
TargetsDiseaseOutcomesProduct/Pre-Clinical Trail/Clinical Trails
CD19Acute lymphoma leukemia Success in pre-clinical study [36]
CD19Acute lymphoma leukemiaSuccess in pre-clinical study[37]
CD19Relapsed/Refractory B cell precursor ALLSuccessTisagenlecleucel/Kymariah
CD19Relapsed/Refractory large B cell lymphoma SuccessAxicabtagene ciloleucel/Kymariah
CD19Relapsed/Refractory follicular lymphoma (FL)Success (May 2022, FDA-approved)Axicabtagene ciloleucel/Kymariah
BCMARelapsed/Refractory multiple myelomaSuccess (February 2022, FDA-approved)Ciltacabtagene autoleucel/Carvykti
BCMARelapsed/Refractory multiple myelomaSuccess (March 2021, FDA-approved)Idecabtagenevicleucel/Abecma
CD19Relapsed/Refractory large B cell lymphomaSuccess (February 2021, FDA-approved)Lisocabtagene autoleucel/Breyanzi
CD19Relapsed/Refractory B cell lymphoma Success (October 2017, FDA-approved)Axicabtagene ciloleucel/Yescarta [162]
CD19Relapsed/Refractory follicular lymphoma (FL) Success (March 2021, FDA-approved)Axicabtagene ciloleucel/Yescarta
IL13RGlioblastoma Success in preclinical trials[163]
Epidermal growth factor receptor variant III (EGFRvIII)Recurrent GlioblastomaSuccess in human patient [164]

5.2. Inhibition of the Interaction between Immunosuppressive Cells to Improve CAR T Function

MDSC and OS-M have significant negative implications for the immune response in osteosarcoma [110]. Hence, modifying the conditions within the tumor microenvironment represents another potential approach to enhance the effectiveness of CAR T cell therapy [165]. In a clinical trial involving third-generation CD19 CAR T cell therapy, low levels of monocytic MDSCs (M-MDSCs) were observed. M-MDSCs were associated with a positive treatment response in patients with lymphoma and leukemia [166]. This suggests that a lower presence of M-MDSCs in the patient’s immune system may be correlated with better outcomes when using CAR T cell therapy to treat these types of cancers [167]. The presence of MDSCs, which can suppress the immune response, is a significant factor to consider when evaluating the success of immunotherapies like CAR T cell therapy [165]. Strategies to mitigate the effects of MDSCs and enhance the immune response has attracted investigators’ attention. The removal of MDSCs using an anti-Gr-1 antibody demonstrated the enhanced effectiveness of CAR T cell therapy in mouse models [168,169]. Moreover, CAR T cells designed to target the tumor vasculature via VEGFR-2 were successful in decreasing MDSCs within the TME [170]. Notably, MDSCs also expressed VEGFR-2, the same target as the CAR T cells, indicating a potential mechanism for their reduction [170]. When CAR T cells were combined with substances like polyinosinic-polycytidylic acid (poly I:C), all-trans retinoic acid (ATRA) [166], and gemtuzumab ozogamicin (GO) [171], enhanced anti-tumor effectiveness was observed. The suppressive activity of MDSCs was associated with an increase in therapeutic efficacy [172,173]. TAMs secrete immunosuppressive cytokines such as transforming growth factor-beta (TGF-β) and interleukin-10 (IL-10) [174]. These cytokines support the differentiation and expansion of Tregs while inhibiting the activity of effector T cells [174]. TAM-derived TGF-β, in particular, has been implicated in promoting Treg development and function [59]. Additionally, Tregs can reshape the profiles of immune cells within the tumor, suppressing IFN-γ secretion by CD8+ T cells and promoting the development of OS-M through sterol regulatory element-binding protein 1 (SREBP1)-mediated fatty acid synthesis [175,176]. The inhibition of these cytokines transforms the tumor microenvironment and benefits the prevention of cancer cell immune escape, thus improving CAR T function [149].

5.3. Limitations of Engineered Cell Therapy

Another challenge is the infiltration of CAR immune cells. To improve their infiltration into human tumors, we can try the following method: develop approaches to modulate the tumor microenvironment in a way that facilitates CAR T/M infiltration, which involves altering the levels of immunosuppressive factors or increasing the presence of tumor-specific chemo-attractants.
It is important to consider how chemokine signaling pathways can be integrated into the treatment strategies for osteosarcoma, especially in combination with immunotherapies. An important pathway for the CAR targeting these pathways may be the modulation of the tumor microenvironment, reduction in immunosuppression, and improvement in the effectiveness of immunotherapeutic approaches in osteosarcoma.
In addition, designing CAR T/M cells with improved homing mechanisms to recognize and migrate to specific tumor sites more efficiently may represent another strategy. As we know, the CAR scFv recognizes and binds to the specific tumor-associated antigen (or other targeted antigen) on the surface of cancer cells or infected cells. And, the intracellular domains of the CAR contain signaling elements, typically derived from co-stimulatory molecules (e.g., CD28 and 4-1BB) and the CD3ζ chain, for which signaling domains activate downstream signaling cascades upon antigen binding so we can further develop new co-stimulatory molecules. Moreover, we can fine-tune the design of CARs used in CAR-M therapy to ensure they are well-suited for navigating the complex human tumor microenvironment.
Additionally, to improve the effects of CAR T/M, combinatorial therapies are good strategy. For instance, CAR-M therapy can be combined with other treatments, such as checkpoint inhibitors, to help overcome immunosuppression in the tumor microenvironment and promote CAR-M infiltration. To further improve infiltration, a drug delivery strategy can also be considered. Research can focus on exploring local delivery methods to directly introduce CAR-M cells into or near tumor sites, potentially bypassing some of the obstacles posed by the complex tumor microenvironment.

6. Other Nanoparticles Affecting OS-N and Treg

Due to the pro-tumor impact of MDSCs in the OS-TME- [74], the blocking, reprogramming, or elimination of MDSCs has been surfaced as an avenue for transforming the OS-TME. For instance, to decrease the ratio of MDSCs in the TME, a nanoparticle named HA/ZIF-8@Gem/D-1-MT NP was designed to treat OS [22]. The sufficient encapsulation of the chemotherapeutic agent gemcitabine in the nanoparticle exerts cytotoxic activity, and, in another aspect, the incorporated IDO inhibitor 1-methyl-DL-tryptophan (1-MT) can suppress IDO [22]. MDSC is an inflammatory cell in the OS-TME. Nano-based efforts reduce tumor-related inflammation and can modulate the immune response in the OS-TME. Indocyanine green-encapsulated calcium phosphosilicate nanoparticles (ICG-CPSNPs) were utilized to impact the OS-TME. Via the mitigation of inflammation-MDSCs, the use of ICG-CPSNPs generates PDT effects inhibiting OS [177].
The reduced population of T regulatory cells (Tregs) within the TME can enhance the effectiveness of a new nanoparticle hyperthermia therapy [178]. However, keywords such as “nanoparticle for treg cell in osteosarcoma” and “inhibition of function of regulatory T cells in osteosarcoma using nanoparticles” provide nearly zero results, suggesting the absence of nanoparticles using Treg as a direct target in the OS-TME. The HA/ZIF-8@Gem/D-1-MT NPs discussed above not only suppressed MDSC but also reduced the regulatory T cells [22]. Hongbin Fan and his colleagues unraveled the diversity and immunosuppressive role of regulatory T cells in osteosarcoma via the analysis of single-cell RNA transcriptome data [179].

7. Other Nanoparticle Effects

Nanoparticles offer significant potential for delivering a range of anti-osteosarcoma agents, and ongoing research in this area promises novel insights and strategies for combatting osteosarcoma through immunotherapy [180]. Firstly, as we discussed nanoparticles (Nps) can be used for the temporal and spatial regulation or modulation of Tregs, TAMs, and MDSCs in the OS-TME. Nps offers a strategy to transform “cold” OS-TME into “hot” OS-TME, enhancing the immune response within the tumor microenvironment [181]. Secondly, the nanoparticle-based therapy regenerates the dying OS and causes immunogenic cell death (ICD) effects. Moreover, nanoparticles have favorable optical and magnetic attributes, making them suitable for combination with other therapeutics, including photodynamic therapy (PDT) [182]. Thirdly, nanomaterials improve the efficacy of CAR T or CAR-M in OS treatment [125].

7.1. NP-Induced ICD

Photodynamic therapy (PDT) can cause cancer cell apoptosis in osteosarcoma (OS) via the production of reactive oxygen species (ROS) [5,6]. Nanocarrier encapsulated photosensitizers (PSs) increase PDT-induced tumor cell death. More importantly, the balance between apoptosis and autophagy is of significance in the rescuing pathway against cell death, and a lower ratio of autophagy leads to backfiring effects, i.e., triggering the anti-tumorigenic effects in the PDT-treated TME [9]. However, inhibiting autophagy increases calreticulin (CRT) induced by ROS, suggesting elevated immunogenic cell death (ICD) [10,11]. Given the controversial findings, additional in-depth PDT studies on OS are still required. In a study, researchers designed a bovine serum albumin-zinc phthalocyanine (BSA-ZnPc; BZ) to assess the above findings. In this study, the OS tumor was resected at first, followed by BSA-ZnPc treatments. With combinations, the autophagy protein beclin, ATG5, and LC3II increased; however, the contrary protein p62 decreased, suggesting an increased level of autophagy. Furthermore, when cells were treated together with autophagy inhibitor, 3-MA, PD-L1 expression in cancer cells increased, indicating the correlation of decreased PDL-1 and decreased autophagy levels [183]. Moreover, gold nanoparticles encapsulated within tumor cell membranes offer the potential for precise drug delivery to specific sites, controlled drug release, and effective phototherapy [184]. As an example, researchers developed a mitochondria-targeted nano micelle, called OPDEA-PDCA, with the ability to induce mitochondrial oxidative stress and trigger pyroptosis in osteosarcoma cells [185]. In this study, mitochondria were specifically targeted. Additionally, the authors incorporated modified dichloroacetate (DCA) to inhibit pyruvate dehydrogenase kinase 1 (PDHK1), thereby inducing mitochondrial oxidative stress, causing pyroptosis, and further facilitating ICD [185]. Several studies have demonstrated that when osteosarcoma cells undergo cell death, dying OS cells become immunogenic and subsequently boost the immune response in the human body [186,187]. The combinational treatments initiated immunogenic cell death (ICD), which enhanced the macrophage’s innate immune response [188]. In animal models of metastatic osteosarcoma, the combined blockades of CTLA-4 and PD-L1 completely eliminated the bone carcinoma. Moreover, the addition of tumor lysate-pulsed dendritic cells (DCs) exerted a strong immune response in murine osteosarcomas [189].

7.2. NP-Induced Cold OS-TME to Hot OS-TME Transformation

The concept of cold OS-TME indicates the immunosuppressive OS-TME, where the immune reaction is dominant. To treat the OS, we need to heat the OS-TME. As mentioned, the MDSC inhibits the immune system’s response. Thus, the depletion of immune-inhibitory cells, such as MDSCs, benefits the hot OS-TME construction [190]. Additionally, combinations of inhibitory cytokines like IL-4 and TGF-β can contribute to a comprehensive approach to enhance the antitumor immune response [136]. A good example is gemcitabine (Gem) plus docetaxel or sirolimus. The underlying anti-cancer mechanism is associated with the impairment in DNA replication in cancer cells. The combinations exert excellent effects towards OS treatment; however, current therapeutic outcomes remain an unsolved challenge. To solve this dilemma, an innovative nanoparticle (HA/ZIF-8@Gem/D-1-MT NPs) has been designed to efficiently encapsulate the chemotherapeutic drug gemcitabine and an IDO inhibitor, 1-methyl-DL-tryptophan (1-MT) [22], leading to positive therapeutic effects and a synergistic anti-tumor immune response in OS-TME [22]. Zoledronic acid, a third-generation bisphosphonate, triggers apoptosis in tumor cells and inhibits angiogenesis and metastasis in vivo. This effect is achieved by arresting the S phase and activating DNA damage in cancer [191]. Indoleamine 2,3-dioxygenase (IDO) is an enzyme responsible for converting tryptophan into inhibitory metabolites that hinder T cell proliferation, creating an environment for tumor cells to evade the immune system [192]. In osteosarcoma, the combination of IDO inhibitors and platinum (IV) prodrugs acts synergistically to reverse low immune responses [193]. This combined approach enhances the efficacy of cancer chemotherapy and immunotherapy for osteosarcoma [193]. Moreover, a metal–organic framework enhances chemo-immunotherapy for osteosarcoma by inhibiting MDSCs and indoleamine 2,3-dioxygenase [22].
The activation of stimulator of interferon genes (STINGs) in the TME suggests the production of anti-tumor cytokines and interferons; thus, the level of STING is an indicator of the cold TME and hot TME [194,195]. Activating the STING pathway with NPs could enhance the anti-OS effect [196]. Additionally, radiotherapy can activate the cGAS-STING pathway by inducing DNA breaks [197]. The combination of hafnium oxide NP with radiotherapy can augment the efficacy of radiotherapy in patients with soft tissue sarcoma [197]. It is reported that Pt(IV)–C12 and IDOi (NLG919) not only hindered tryptophan metabolism in cancer cells but also caused DNA damage and triggered the STING pathway [197]. The impact of NP-Pt-IDOi on inhibiting cancer cells was assessed using various osteosarcoma cell lines in laboratory settings [186] docetaxel (DTX) and alendronate (ALD) were incorporated into chitosan-conjugated PLGA NPs to against OS [198,199]. These NPs, prepared by the nanoprecipitation technique, are found within the tumor targeting range (~200 nm) and display an effective positive charge (20 mV) capable of increasing cellular uptake efficiency [198,199].

7.3. NP-Improved TME for Combating Immune-Based Therapy Resistance in OS

Why Is There Resistance to ICD Effects in OS-TME? First, ABCB1 lowers the intracellular accumulation of ICD reagents such as doxorubicin, preventing DOX from reaching a concentration sufficient to induce ICD [187]. Additionally, ABCB1 increases the cell response towards ER stress and therefore impairs the magnitude of ICD [200,201] Finally, the resistant genes in OS cells hinder the functions of anti-cancer factors-calreticulin developed with the ICD effects triggered by immunotherapeutic ingredients [202]. In addition to ABCB1, OS cells release soluble factors, including bone-derived molecules such as transforming growth factor-beta (TGF-βs), being notable for inhibiting T cell function [50,203]. MDSCs and derived indoleamine 2,3-dioxygenase (IDO) also contribute to the OS-TIME [22,204,205]. TAM-induced vascular endothelial growth factor (VEGF) is an emerging factor in the OS-TIME [206]. Thus, to recover the immunity of cells, a protective microenvironment for immunotherapy for OS is important. Nanoparticles -Induced OS-TME is effective in transforming the TME. Np incorporating both albendazole (ABZ) and doxorubicin were designed, and the ABZ component of these nanoparticles demonstrated effective suppression of the expression of HIF-1α and its downstream protein VEGF [207]. In this study, the combination of ABZ with DOX drastically increases intracellular reactive oxygen species (ROS) and thus elevates tumor cell apoptosis. More significantly, ABZ effectively suppresses the expression of its downstream vascular endothelial growth factor (VEGF) via downregulating the levels of hypoxia-inducible factor-1α (HIF-1α) [207]. It is worth to mention that MDSCs contribute to bone resorption by activating osteoclasts in an HIF-1α-dependent manner [14]. HIF-1α further promotes the release of soluble factors, including basic fibroblast growth factor (bFGF) and matrix metalloproteinase-9 (MMP-9), favoring OS cell invasion [200]. Additionally, nanoparticles can be engineered to modulate the immune response, potentially overcoming immunosuppressive factors present in the OS-TME [132,165,208]. Nanoparticles can enhance the function of T cell therapeutics and promote a more robust anti-tumor immune response [209]. Because of the lower level of Her2 in the OS, the immunotherapy for Her2 alone is not effective in OS treatment, and graphene oxide nanoparticles conjugated with anti-Her2 Trastuzumab dramatically reduce OS growth [210]. The coating of OS cell membrane can increase the uptake of therapeutics in OS cells. Nanoparticles coated with a K7M2 membrane (derived from a murine (mouse) model of osteosarcoma) carrying ginsenoside Rh2 and alendronate increase the antitumor activity of TILs and decrease immunosuppressive Treg cells [211]. Curcumin-loaded nanoparticles stimulate autophagy, increase DC infiltration, and show synergistic effects with anti-PD-1/PD-L1pathway inhibitors [212]. Telomerase-specific oncolytic adenovirus OBP-502 in combination with anti-PD-1 increases efficacy by inducing autophagy, restoring ICD, and upregulating PD-L1 [213]. To sum up, in the NP-induced OS-TME, tumor-promoting factors such as ROS, VEGF, HIF-A, Bfgf, and MMP-9 can be decreased.

7.4. Engineering Cell Approaches

The cutting-edge field of engineering cell therapies represents a novel and promising avenue for tackling osteosarcoma-associated macrophages. This approach involves manipulating or modifying immune cells, such as macrophages, to enhance their anti-tumor activity. Genetic engineering, immunotherapy, and cell-based therapies fall under this category. The figure details the intricate cellular and molecular mechanisms underlying engineering cell approaches.

8. Conclusions and Future Directions

Osteosarcoma majorly develops in adolescence, causing a dramatic decrease in living quality. The reason for its poor prognosis and therapeutic challenges is due to immune escape. In addition, OS cells can invade other organs and develop resistance towards therapeutics. The above activity is closely associated with the TME; thus, a comprehensive understanding of the complicated OS-TME is significant. In the first part of this review, we discussed the roles of crucial immune cells in the OS-TME including OS-Ms, OS-Ns, MDSCs, and Treg. Given the importance of the TME, remodeling the OS-TME has surfaced as a key strategy to treat OS. Therefore, we highlighted the use of nanotechnology to cover the regimens to modulate the OS-TME. Indeed, compared with the past, investigations into cancer cell communication with immune cells have transformed from a less-traveled road to a flow of traffic. Because of in-depth study, strategies focused on the interactions of cells have made successful progress. However, there are obstacles in this wide road.
Firstly, the heterogeneity of cells within the tumor microenvironment poses a significant challenge to the effectiveness of therapeutic approaches. This diversity is particularly evident in MDSCs and OS-Ms [214], even though both polymorphonuclear MDSCs (PMN-MDSC) and monocytic MDSCs (M-MDSCs) promote OS progression [215]. The heterogeneity of cells requires close attention when choosing therapeutics. For instance, the STAT3 less-expressed M-MDSCs can differentiate to anti-tumor DCs, and this complete elimination may cause decreased DCs in the OS-TME, leading to non-beneficial effects of OS treatment [109]. TAM has a distinct tumor-promoting sub-phenotype. It is reported that M2b is beneficial for anti-tumor activity, demonstrating the difficulty in choosing strategies. As a result, sophisticated platforms like “organ-on-chip” models may be necessary for more comprehensive assays and to gain a deeper understanding of the role MDSCs play in the context of cancer. Secondly, there is a gap between laboratory models and humans post-challenge in real-Sworld conditions. MDSCs in humans exhibit distinct characteristics when compared to their murine counterparts [214]. Specifically, human MDSCs are typically characterized by a lack or low expression of HLA-DR, a major histocompatibility complex class II receptor, and a high expression of CD33, a myeloid cell marker [216]. Enhancing understanding of this difference is indeed a critical issue that needs to be addressed to achieve expected therapeutic efficacy.

Author Contributions

Conceptualization, S.C. and H.Z.; writing—original draft preparation, S.C., H.W., X.K. and H.Z.; writing—review and editing, S.C., H.W., X.K. and H.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jafari, F.; Javdansirat, S.; Sanaie, S.; Naseri, A.; Shamekh, A.; Rostamzadeh, D.; Dolati, S. Osteosarcoma: A comprehensive review of management and treatment strategies. Ann. Diagn. Pathol. 2020, 49, 151654. [Google Scholar] [CrossRef]
  2. Lawrence, R.C.; Helmick, C.G.; Arnett, F.C.; Deyo, R.A.; Felson, D.T.; Giannini, E.H.; Heyse, S.P.; Hirsch, R.; Hochberg, M.C.; Hunder, G.G.; et al. Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum. Off. J. Am. Coll. Rheumatol. 1998, 41, 778–799. [Google Scholar] [CrossRef]
  3. Zhao, X.; Wu, Q.; Gong, X.; Liu, J.; Ma, Y. Osteosarcoma: A review of current and future therapeutic approaches. Biomed. Eng. Online 2021, 20, 1–14. [Google Scholar] [CrossRef]
  4. Lin, Z.; Wu, Z.; Luo, W. Chimeric antigen receptor T-cell therapy: The light of day for osteosarcoma. Cancers 2021, 13, 4469. [Google Scholar] [CrossRef]
  5. Brierley, J.D.; Gospodarowicz, M.K.; Wittekind, C. TNM Classification of Malignant Tumours; John Wiley & Sons: Hoboken, NJ, USA, 2017. [Google Scholar]
  6. Stiller, C.; Bielack, S.; Jundt, G.; Steliarova-Foucher, E. Bone tumours in European children and adolescents, 1978–1997. Report from the Automated Childhood Cancer Information System project. Eur. J. Cancer 2006, 42, 2124–2135. [Google Scholar] [CrossRef]
  7. Tomita, K.; Kawahara, N.; Murakami, H.; Demura, S. Total en bloc spondylectomy for spinal tumors: Improvement of the technique and its associated basic background. J. Orthop. Sci. 2006, 11, 3–12. [Google Scholar] [CrossRef]
  8. Hillmann, A.; Hoffmann, C.; Gosheger, G.; Krakau, H.; Winkelmann, W. Malignant tumor of the distal part of the femur or the proximal part of the tibia: Endoprosthetic replacement or rotationplasty. Functional outcome and quality-of-life measurements. JBJS 1999, 81, 462–468. [Google Scholar] [CrossRef]
  9. Hoffmann, C.; Gosheger, G.; Gebert, C.; Jürgens, H.; Winkelmann, W. Functional results and quality of life after treatment of pelvic sarcomas involving the acetabulum. JBJS 2006, 88, 575–582. [Google Scholar]
  10. Saeter, G.; Høie, J.; Stenwig, A.E.; Johansson, A.K.; Hannisdal, E.; Solheim, Ø.P. Systemic relapse of patients with osteogenic sarcoma. Prognostic factors for long term survival. Cancer 1995, 75, 1084–1093. [Google Scholar] [CrossRef]
  11. Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The tumor microenvironment: A milieu hindering and obstructing antitumor immune responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
  12. Umansky, V.; Blattner, C.; Gebhardt, C.; Utikal, J. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines 2016, 4, 36. [Google Scholar] [CrossRef]
  13. De Vlaeminck, Y.; González-Rascón, A.; Goyvaerts, C.; Breckpot, K. Cancer-associated myeloid regulatory cells. Front. Immunol. 2016, 7, 113. [Google Scholar] [CrossRef] [PubMed]
  14. Ling, Z.; Yang, C.; Tan, J.; Dou, C.; Chen, Y. Beyond immunosuppressive effects: Dual roles of myeloid-derived suppressor cells in bone-related diseases. Cell. Mol. Life Sci. 2021, 78, 7161–7183. [Google Scholar] [CrossRef]
  15. Zając, A.E.; Czarnecka, A.M.; Rutkowski, P. The Role of Macrophages in Sarcoma Tumor Microenvironment and Treatment. Cancers 2023, 15, 5294. [Google Scholar] [CrossRef]
  16. Wu, K.; Lin, K.; Li, X.; Yuan, X.; Xu, P.; Ni, P.; Xu, D. Redefining tumor-associated macrophage subpopulations and functions in the tumor microenvironment. Front. Immunol. 2020, 11, 1731. [Google Scholar] [CrossRef]
  17. Nirala, B.K.; Yamamichi, T.; Petrescu, D.I.; Shafin, T.N.; Yustein, J.T. Decoding the Impact of Tumor Microenvironment in Osteosarcoma Progression and Metastasis. Cancers 2023, 15, 5108. [Google Scholar] [CrossRef]
  18. Wu, L.; Saxena, S.; Awaji, M.; Singh, R.K. Tumor-associated neutrophils in cancer: Going pro. Cancers 2019, 11, 564. [Google Scholar] [CrossRef]
  19. Ha, T.-Y. The role of regulatory T cells in cancer. Immune Netw. 2009, 9, 209–235. [Google Scholar] [CrossRef]
  20. Yu, L.; Zhang, J.; Li, Y. Effects of microenvironment in osteosarcoma on chemoresistance and the promise of immunotherapy as an osteosarcoma therapeutic modality. Front. Immunol. 2022, 13, 871076. [Google Scholar] [CrossRef]
  21. Payoe, K.S. Identification of Novel Therapeutic Targets in Osteosarcoma for the Development of Nanoparticle Based Drug Delivery Systems. Ph.D. Thesis, Brunel University London, London, UK, 2023. [Google Scholar]
  22. Fan, Q.; Zuo, J.; Tian, H.; Huang, C.; Nice, E.C.; Shi, Z.; Kong, Q. Nanoengineering a metal–organic framework for osteosarcoma chemo-immunotherapy by modulating indoleamine-2, 3-dioxygenase and myeloid-derived suppressor cells. J. Exp. Clin. Cancer Res. 2022, 41, 162. [Google Scholar] [CrossRef]
  23. Lu, Y.; Zhang, J.; Chen, Y.; Kang, Y.; Liao, Z.; He, Y.; Zhang, C. Novel immunotherapies for osteosarcoma. Front. Oncol. 2022, 12, 830546. [Google Scholar] [CrossRef] [PubMed]
  24. Li, Y.; Rezvani, K.; Rafei, H. Next-generation chimeric antigen receptors for T-and natural killer-cell therapies against cancer. Immunol. Rev. 2023, 320, 217–235. [Google Scholar] [CrossRef] [PubMed]
  25. Zeng, Z.; Pu, K. Improving cancer immunotherapy by cell membrane-camouflaged nanoparticles. Adv. Funct. Mater. 2020, 30, 2004397. [Google Scholar] [CrossRef]
  26. Muluh, T.A.; Chen, Z.; Li, Y.; Xiong, K.; Jin, J.; Fu, S.; Wu, J. Enhancing cancer immunotherapy treatment goals by using nanoparticle delivery system. Int. J. Nanomed. 2021, 16, 2389–2404. [Google Scholar] [CrossRef]
  27. Tang, L.; Zheng, Y.; Melo, M.B.; Mabardi, L.; Castaño, A.P.; Xie, Y.-Q.; Li, N.; Kudchodkar, S.B.; Wong, H.C.; Jeng, E.K. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 2018, 36, 707–716. [Google Scholar] [CrossRef]
  28. Zheng, C.; Zhang, J.; Chan, H.F.; Hu, H.; Lv, S.; Na, N.; Tao, Y.; Li, M. Engineering nano-therapeutics to boost adoptive cell therapy for cancer treatment. Small Methods 2021, 5, 2001191. [Google Scholar] [CrossRef]
  29. Attia, M.F.; Anton, N.; Wallyn, J.; Omran, Z.; Vandamme, T.F. An overview of active and passive targeting strategies to improve the nanocarriers efficiency to tumour sites. J. Pharm. Pharmacol. 2019, 71, 1185–1198. [Google Scholar] [CrossRef]
  30. Balakrishnan, P.B.; Sweeney, E.E. Nanoparticles for enhanced adoptive T cell therapies and future perspectives for CNS tumors. Front. Immunol. 2021, 12, 600659. [Google Scholar] [CrossRef]
  31. Barua, S.; Mitragotri, S. Challenges associated with penetration of nanoparticles across cell and tissue barriers: A review of current status and future prospects. Nano Today 2014, 9, 223–243. [Google Scholar] [CrossRef]
  32. Durgeau, A.; Virk, Y.; Corgnac, S.; Mami-Chouaib, F. Recent advances in targeting CD8 T-cell immunity for more effective cancer immunotherapy. Front. Immunol. 2018, 9, 14. [Google Scholar] [CrossRef] [PubMed]
  33. Cerrano, M.; Ruella, M.; Perales, M.-A.; Vitale, C.; Faraci, D.G.; Giaccone, L.; Coscia, M.; Maloy, M.; Sanchez-Escamilla, M.; Elsabah, H. The advent of CAR T-cell therapy for lymphoproliferative neoplasms: Integrating research into clinical practice. Front. Immunol. 2020, 11, 888. [Google Scholar] [CrossRef]
  34. McKee, M.D.; Fichera, A.; Nishimura, M.I. T cell immunotherapy. Front. Biosci.-Landmark 2007, 12, 919–932. [Google Scholar] [CrossRef]
  35. Chen, C.; Xie, L.; Ren, T.; Huang, Y.; Xu, J.; Guo, W. Immunotherapy for osteosarcoma: Fundamental mechanism, rationale, and recent breakthroughs. Cancer Lett. 2021, 500, 1–10. [Google Scholar] [CrossRef]
  36. Ittershagen, S.; Ericson, S.; Eldjerou, L.; Shojaee, A.; Bleickardt, E.; Patel, M.; Taran, T.; Anak, O.; Hall, C.; Leung, M. Industry’s giant leap into cellular therapy: Catalyzing chimeric antigen receptor T cell (CAR-T) immunotherapy. Curr. Hematol. Malign. Rep. 2019, 14, 47–55. [Google Scholar] [CrossRef]
  37. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  38. Wang, X.; Rivière, I. Manufacturing of CAR-T Cells: The Assembly Line. In Gene and Cellular Immunotherapy for Cancer; Springer: Berlin/Heidelberg, Germany, 2022; pp. 121–139. [Google Scholar]
  39. Mirzaei, H.R.; Rodriguez, A.; Shepphird, J.; Brown, C.E.; Badie, B. Chimeric antigen receptors T cell therapy in solid tumor: Challenges and clinical applications. Front. Immunol. 2017, 8, 1850. [Google Scholar] [CrossRef] [PubMed]
  40. Paijens, S.T.; Vledder, A.; de Bruyn, M.; Nijman, H.W. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell. Mol. Immunol. 2021, 18, 842–859. [Google Scholar] [CrossRef] [PubMed]
  41. Melosky, B. Cutaneous Reactions to Tyrosine Kinase Inhibitors. In Skin Diseases in the Immunocompromised; Springer: Berlin/Heidelberg, Germany, 2014; pp. 107–120. [Google Scholar]
  42. Sheth, V.S.; Gauthier, J. Taming the beast: CRS and ICANS after CAR T-cell therapy for ALL. Bone Marrow Transplant. 2021, 56, 552–566. [Google Scholar] [CrossRef] [PubMed]
  43. Wallet, F.; Sesques, P.; Devic, P.; Levrard, M.; Ader, F.; Friggeri, A.; Bachy, E. CAR-T cell: Toxicities issues: Mechanisms and clinical management. Bull. Du Cancer 2021, 108, S117–S127. [Google Scholar] [CrossRef] [PubMed]
  44. Long, A.H.; Highfill, S.L.; Cui, Y.; Smith, J.P.; Walker, A.J.; Ramakrishna, S.; El-Etriby, R.; Galli, S.; Tsokos, M.G.; Orentas, R.J. Reduction of MDSCs with all-trans retinoic acid improves CAR therapy efficacy for sarcomas. Cancer Immunol. Res. 2016, 4, 869–880. [Google Scholar] [CrossRef] [PubMed]
  45. Ahmed, N.; Salsman, V.S.; Yvon, E.; Louis, C.U.; Perlaky, L.; Wels, W.S.; Dishop, M.K.; Kleinerman, E.E.; Pule, M.; Rooney, C.M. Immunotherapy for osteosarcoma: Genetic modification of T cells overcomes low levels of tumor antigen expression. Mol. Ther. 2009, 17, 1779–1787. [Google Scholar] [CrossRef]
  46. Strohl, W.R.; Naso, M. Bispecific T-cell redirection versus chimeric antigen receptor (CAR)-T cells as approaches to kill cancer cells. Antibodies 2019, 8, 41. [Google Scholar] [CrossRef]
  47. Slaney, C.Y.; Wang, P.; Darcy, P.K.; Kershaw, M.H. CARs versus BiTEs: A comparison between T cell–redirection strategies for cancer treatment. Cancer Discov. 2018, 8, 924–934. [Google Scholar] [CrossRef] [PubMed]
  48. Jena, B.; Dotti, G.; Cooper, L.J. Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood J. Am. Soc. Hematol. 2010, 116, 1035–1044. [Google Scholar] [CrossRef]
  49. Somaiah, N.; Conley, A.P.; Parra, E.R.; Lin, H.; Amini, B.; Soto, L.S.; Salazar, R.; Barreto, C.; Chen, H.; Gite, S. Durvalumab plus tremelimumab in advanced or metastatic soft tissue and bone sarcomas: A single-centre phase 2 trial. Lancet Oncol. 2022, 23, 1156–1166. [Google Scholar] [CrossRef]
  50. Xie, X.; Feng, Y.; Zhang, H.; Su, Q.; Song, T.; Yang, G.; Li, N.; Wei, X.; Li, T.; Qin, X.; et al. Remodeling tumor immunosuppressive microenvironment via a novel bioactive nanovaccines potentiates the efficacy of cancer immunotherapy. Bioact. Mater. 2022, 16, 107–119. [Google Scholar] [CrossRef] [PubMed]
  51. Wen, L.; Tian-Cong, W.; Dong-Mei, H.; Yue, H.; Ting, F.; Wen-Jie, G.; Qiang, X.J. Carnosic acid enhances the anti-lung cancer effect of cisplatin by inhibiting myeloid-derived suppressor cells. Chin. J. Nat. Med. 2018, 16, 907–915. [Google Scholar]
  52. Corre, I.; Verrecchia, F.; Crenn, V.; Redini, F.; Trichet, V. The osteosarcoma microenvironment: A complex but targetable ecosystem. Cells 2020, 9, 976. [Google Scholar] [CrossRef] [PubMed]
  53. Verhoeven, J.; Baelen, J.; Agrawal, M.; Agostinis, P. Endothelial cell autophagy in homeostasis and cancer. FEBS Lett. 2021, 595, 1497–1511. [Google Scholar] [CrossRef]
  54. Pramanik, A.; Bhattacharyya, S. Myeloid derived suppressor cells and innate immune system interaction in tumor microenvironment. Life Sci. 2022, 305, 120755. [Google Scholar] [CrossRef]
  55. Pinton, L. The Crosstalk between Activated T Cells and Myeloid Derived Suppressor Cells: Characterization of Molecular Mechanisms Involved in Immune Suppression. 2014. Available online: https://www.research.unipd.it/handle/11577/3423699 (accessed on 1 April 2014).
  56. Le Bourgeois, T.; Strauss, L.; Aksoylar, H.-I.; Daneshmandi, S.; Seth, P.; Patsoukis, N.; Boussiotis, V.A. Targeting T cell metabolism for improvement of cancer immunotherapy. Front. Oncol. 2018, 8, 237. [Google Scholar] [CrossRef]
  57. Rivera, L.B.; Bergers, G. Myeloid cell-driven angiogenesis and immune regulation in tumors. Trends Immunol. 2015, 36, 240. [Google Scholar] [CrossRef]
  58. Rak, J.W.; St Croix, B.D.; Kerbel, R.S. Consequences of angiogenesis for tumor progression, metastasis and cancer therapy. Anti-Cancer Drugs 1995, 6, 3–18. [Google Scholar] [CrossRef]
  59. Xue, V.W.; Chung, J.Y.-F.; Córdoba, C.A.G.; Cheung, A.H.-K.; Kang, W.; Lam, E.W.-F.; Leung, K.-T.; To, K.-F.; Lan, H.-Y.; Tang, P.M.-K. Transforming growth factor-β: A multifunctional regulator of cancer immunity. Cancers 2020, 12, 3099. [Google Scholar] [CrossRef]
  60. Tsubakihara, Y.; Moustakas, A. Epithelial-mesenchymal transition and metastasis under the control of transforming growth factor β. Int. J. Mol. Sci. 2018, 19, 3672. [Google Scholar] [CrossRef] [PubMed]
  61. Ya, G.; Ren, W.; Qin, R.; He, J.; Zhao, S. Role of myeloid-derived suppressor cells in the formation of pre-metastatic niche. Front. Oncol. 2022, 12, 975261. [Google Scholar] [CrossRef]
  62. Meftahpour, V.; Aghebati-Maleki, A.; Fotouhi, A.; Safarzadeh, E.; Aghebati-Maleki, L. Prognostic significance and therapeutic potentials of immune checkpoints in osteosarcoma. EXCLI J. 2022, 21, 250. [Google Scholar]
  63. Imtiyaz, H.Z.; Simon, M.C. Hypoxia-inducible factors as essential regulators of inflammation. In Diverse Effects of Hypoxia on Tumor Progression; Springer: Berlin/Heidelberg, Germany, 2010; pp. 105–120. [Google Scholar]
  64. Balkwill, F. Cancer and the chemokine network. Nat. Rev. Cancer 2004, 4, 540–550. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, F.; Feng, Y.C.; Zhao, G.; Zhang, R.; Cheng, Z.Z.; Kong, W.N.; Wu, H.L.; Xu, B.; Lv, X.; Ma, X.M. Tumor-Associated CD163(+) M2 Macrophage Infiltration is Highly Associated with PD-L1 Expression in Cervical Cancer. Cancer Manag. Res. 2020, 12, 5831–5843. [Google Scholar] [CrossRef] [PubMed]
  66. Harada, K.; Dong, X.; Estrella, J.S.; Correa, A.M.; Xu, Y.; Hofstetter, W.L.; Sudo, K.; Onodera, H.; Suzuki, K.; Suzuki, A.; et al. Tumor-associated macrophage infiltration is highly associated with PD-L1 expression in gastric adenocarcinoma. Gastric Cancer 2018, 21, 31–40. [Google Scholar] [CrossRef]
  67. Pierrevelcin, M.; Fuchs, Q.; Lhermitte, B.; Messé, M.; Guérin, E.; Weingertner, N.; Martin, S.; Lelong-Rebel, I.; Nazon, C.; Dontenwill, M. Focus on hypoxia-related pathways in pediatric osteosarcomas and their druggability. Cells 2020, 9, 1998. [Google Scholar] [CrossRef]
  68. He, Z.; Zhang, S. Tumor-associated macrophages and their functional transformation in the hypoxic tumor microenvironment. Front. Immunol. 2021, 12, 741305. [Google Scholar] [CrossRef]
  69. Chen, P.-J.; Chang, C.-H.; Kuo, Y.-L.; Lin, Y.-C. Designing and evaluating a wearable sEMG device for the elderly. In 14th International Conference on ICT, Society, and Human Beings, ICT 2021, 18th International Conference on Web Based Communities and Social Media, WBC 2021 and 13th International Conference on e-Health, EH 2021-Held at the 15th Multi-Conference on Computer Science and Information Systems, MCCSIS 2021; IADIS: Budapest, Hungary, 2021. [Google Scholar]
  70. Zhou, J.; Liu, T.; Wang, W. Prognostic significance of matrix metalloproteinase 9 expression in osteosarcoma: A meta-analysis of 16 studies. Medicine 2018, 97, e13051. [Google Scholar] [CrossRef]
  71. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  72. Sobierajska, K.; Ciszewski, W.M.; Sacewicz-Hofman, I.; Niewiarowska, J. Endothelial cells in the tumor microenvironment. In Tumor Microenvironment: Non-Hematopoietic Cells; Springer: Berlin/Heidelberg, Germany, 2020; pp. 71–86. [Google Scholar]
  73. Minami, K.; Ueda, N.; Ishimoto, K.; Tsujiuchi, T. LPA5-mediated signaling induced by endothelial cells and anticancer drug regulates cellular functions of osteosarcoma cells. Exp. Cell Res. 2020, 388, 111813. [Google Scholar] [CrossRef] [PubMed]
  74. Rodrigues, J.; Sarmento, B.; Pereira, C.L. Osteosarcoma tumor microenvironment: The key for the successful development of biologically relevant 3D in vitro models. In Vitro Models 2022, 1, 5–27. [Google Scholar] [CrossRef]
  75. Pozo, K.; Bibb, J.A. The emerging role of Cdk5 in cancer. Trends Cancer 2016, 2, 606–618. [Google Scholar] [CrossRef] [PubMed]
  76. de Nigris, F.; Mancini, F.P.; Schiano, C.; Infante, T.; Zullo, A.; Minucci, P.B.; Al-Omran, M.; Giordano, A.; Napoli, C.J. Osteosarcoma cells induce endothelial cell proliferation during neo-angiogenesis. J. Cell. Physiol. 2013, 228, 846–852. [Google Scholar] [CrossRef] [PubMed]
  77. Ribatti, D. Morphofunctional Aspects of Tumor Microcirculation; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012. [Google Scholar]
  78. Albulescu, R.; Tanase, C. Tumor Angiogenesis: A Target for Renal Cell Carcinoma Therapy. Current Perspectives and Novel Strategies. Recent Pat. Biomark. 2012, 2, 99–106. [Google Scholar]
  79. Furuya, M.; Yonemitsu, Y. Cancer neovascularization and proinflammatory microenvironments. Curr. Cancer Drug Targets 2008, 8, 253–265. [Google Scholar] [CrossRef] [PubMed]
  80. Tawil, N. Studies on Glioblastoma Associated Thrombosis: Impact of Tumor Cell Heterogeneity and Procoagulant Extracellular Vesicles. Ph.D. Thesis, McGill University, Montréal, QC, Canada, 2021. [Google Scholar]
  81. Phetsouphanh, C.; Xu, Y.; Zaunders, J. CD4 T cells mediate both positive and negative regulation of the immune response to HIV infection: Complex role of T follicular helper cells and regulatory T cells in pathogenesis. Front. Immunol. 2015, 5, 681. [Google Scholar] [CrossRef]
  82. Fehérvari, Z.; Sakaguchi, S. CD4+ Tregs and immune control. J. Clin. Investig. 2004, 114, 1209–1217. [Google Scholar] [CrossRef]
  83. Lucca, L.E.; Dominguez-Villar, M. Modulation of regulatory T cell function and stability by co-inhibitory receptors. Nat. Rev. Immunol. 2020, 20, 680–693. [Google Scholar] [CrossRef] [PubMed]
  84. Whiteside, T.L. FOXP3+ Treg as a therapeutic target for promoting anti-tumor immunity. Expert Opin. Ther. Targets 2018, 22, 353–363. [Google Scholar] [CrossRef] [PubMed]
  85. Borsellino, G.; Kleinewietfeld, M.; Di Mitri, D.; Sternjak, A.; Diamantini, A.; Giometto, R.; Höpner, S.; Centonze, D.; Bernardi, G.; Dell’Acqua, M.L. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: Hydrolysis of extracellular ATP and immune suppression. Blood J. Am. Soc. Hematol. 2007, 110, 1225–1232. [Google Scholar] [CrossRef] [PubMed]
  86. Young, A.; Mittal, D.; Stagg, J.; Smyth, M.J. Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov. 2014, 4, 879–888. [Google Scholar] [CrossRef] [PubMed]
  87. Sek, K.; Mølck, C.; Stewart, G.D.; Kats, L.; Darcy, P.K.; Beavis, P.A. Targeting adenosine receptor signaling in cancer immunotherapy. Int. J. Mol. Sci. 2018, 19, 3837. [Google Scholar] [CrossRef] [PubMed]
  88. Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef] [PubMed]
  89. Raffin, C.; Vo, L.T.; Bluestone, J.A. Treg cell-based therapies: Challenges and perspectives. Nat. Rev. Immunol. 2020, 20, 158–172. [Google Scholar] [CrossRef] [PubMed]
  90. Delgoffe, G.M.; Woo, S.-R.; Turnis, M.E.; Gravano, D.M.; Guy, C.; Overacre, A.E.; Bettini, M.L.; Vogel, P.; Finkelstein, D.; Bonnevier, J. Stability and function of regulatory T cells is maintained by a neuropilin-1–semaphorin-4a axis. Nature 2013, 501, 252–256. [Google Scholar] [CrossRef]
  91. Sansom, D.M.; Walker, L.S. The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol. Rev. 2006, 212, 131–148. [Google Scholar] [CrossRef]
  92. Yang, S.; Wu, C.; Wang, L.; Shan, D.; Chen, B. Pretreatment inflammatory indexes as prognostic predictors for survival in osteosarcoma patients. Int. J. Clin. Exp. Pathol. 2020, 13, 515. [Google Scholar]
  93. Yapar, A.; Tokgöz, M.A.; Yapar, D.; Atalay, İ.B.; Ulucaköy, C.; Güngör, B. Diagnostic and prognostic role of neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, and lymphocyte/monocyte ratio in patients with osteosarcoma. Jt. Dis. Relat. Surg. 2021, 32, 489. [Google Scholar] [CrossRef]
  94. Li, H.; Zhang, K.; Liu, L.-H.; Ouyang, Y.; Bu, J.; Guo, H.-B.; Xiao, T. A systematic review of matrix metalloproteinase 9 as a biomarker of survival in patients with osteosarcoma. Tumor Biol. 2014, 35, 5487–5491. [Google Scholar] [CrossRef]
  95. Liu, Y.; Wang, Y.; Teng, Z.; Chen, J.; Li, Y.; Chen, Z.; Li, Z.; Zhang, Z. Matrix metalloproteinase 9 expression and survival of patients with osteosarcoma: A meta-analysis. Eur. J. Cancer Care 2017, 26, e12364. [Google Scholar] [CrossRef]
  96. Zhang, Q.; Li, J.; Liu, F.; Li, Z. Comments on Li H et al. “A systematic review of matrix metalloproteinase 9 as a biomarker of survival in patients with osteosarcoma”. Tumour Biol. 2015, 36, 5–6. [Google Scholar] [CrossRef]
  97. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  98. Leshner, M.; Wang, S.; Lewis, C.; Zheng, H.; Chen, X.A.; Santy, L.; Wang, Y. PAD4 mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front. Immunol. 2012, 3, 307. [Google Scholar] [CrossRef] [PubMed]
  99. Fu, Y.; Bao, Q.; Liu, Z.; He, G.; Wen, J.; Liu, Q.; Xu, Y.; Jin, Z.; Zhang, W. Development and validation of a hypoxia-associated prognostic signature related to osteosarcoma metastasis and immune infiltration. Front. Cell Dev. Biol. 2021, 9, 633607. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, Y.; Wang, Z.; Jia, F.; Xu, Q.; Shu, Z.; Deng, J.; Li, A.; Yu, M.; Yu, Z. CXCR4-guided liposomes regulating hypoxic and immunosuppressive microenvironment for sorafenib-resistant tumor treatment. Bioact. Mater. 2022, 17, 147–161. [Google Scholar] [CrossRef] [PubMed]
  101. Li, S.; Yue, H.; Wang, S.; Li, X.; Wang, X.; Guo, P.; Ma, G.; Wei, W. Advances of bacteria-based delivery systems for modulating tumor microenvironment. Adv. Drug Deliv. Rev. 2022, 188, 114444. [Google Scholar] [CrossRef]
  102. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef]
  103. Teng, M.W.; Galon, J.; Fridman, W.-H.; Smyth, M.J. From mice to humans: Developments in cancer immunoediting. J. Clin. Investig. 2015, 125, 3338–3346. [Google Scholar] [CrossRef]
  104. Tanaka, K.; Albin, M.J.; Yuan, X.; Yamaura, K.; Habicht, A.; Murayama, T.; Grimm, M.; Waaga, A.M.; Ueno, T.; Padera, R.F.; et al. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. J. Immunol. 2007, 179, 5204–5210. [Google Scholar] [CrossRef] [PubMed]
  105. Castellanos, J.R.; Purvis, I.J.; Labak, C.M.; Guda, M.R.; Tsung, A.J.; Velpula, K.K.; Asuthkar, S. B7-H3 role in the immune landscape of cancer. Am. J. Clin. Exp. Immunol. 2017, 6, 66. [Google Scholar] [PubMed]
  106. Mortezaee, K. HHLA2 immune-regulatory roles in cancer. Biomed. Pharmacother. 2023, 162, 114639. [Google Scholar] [CrossRef] [PubMed]
  107. Xu, W.; Hiếu, T.; Malarkannan, S.; Wang, L. The structure, expression, and multifaceted role of immune-checkpoint protein VISTA as a critical regulator of anti-tumor immunity, autoimmunity, and inflammation. Cell. Mol. Immunol. 2018, 15, 438–446. [Google Scholar] [CrossRef] [PubMed]
  108. Gardner, V.; Madu, C.O.; Lu, Y. Anti-VEGF therapy in cancer: A double-edged sword. In Physiologic and Pathologic Angiogenesis-Signaling Mechanisms and Targeted Therapy; InTech: Rijeka, Croatia, 2017; pp. 385–410. Available online: https://books.google.co.jp/books?hl=zh-TW&lr=&id=3PiODwAAQBAJ&oi=fnd&pg=PA385&dq=Anti-VEGF+therapy+in+cancer:+A+double-edged+sword.+In+Physiologic+and+Pathologic+Angiogenesis-Signaling+Mechanisms+and+Targeted+Therapy%3B+2017&ots=XEqyI2AyHw&sig=_VD70iZXLFpr8Stt2DnwO9MUwIU&redir_esc=y#v=onepage&q=Anti-VEGF%20therapy%20in%20cancer%3A%20A%20double-edged%20sword.%20In%20Physiologic%20and%20Pathologic%20Angiogenesis-Signaling%20Mechanisms%20and%20Targeted%20Therapy%3B%202017&f=false (accessed on 1 May 2017).
  109. Li, K.; Shi, H.; Zhang, B.; Ou, X.; Ma, Q.; Chen, Y.; Shu, P.; Li, D.; Wang, Y. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct. Target. Ther. 2021, 6, 362. [Google Scholar] [CrossRef] [PubMed]
  110. Cascini, C.; Chiodoni, C. The immune landscape of osteosarcoma: Implications for prognosis and treatment response. Cells 2021, 10, 1668. [Google Scholar] [CrossRef] [PubMed]
  111. Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61. [Google Scholar] [CrossRef]
  112. Wang, S.-D.; Li, H.-Y.; Li, B.-H.; Xie, T.; Zhu, T.; Sun, L.-L.; Ren, H.-Y.; Ye, Z.-M. The role of CTLA-4 and PD-1 in anti-tumor immune response and their potential efficacy against osteosarcoma. Int. Immunopharmacol. 2016, 38, 81–89. [Google Scholar] [CrossRef]
  113. Pu, F.; Chen, F.; Zhang, Z.; Qing, X.; Lin, H.; Zhao, L.; Xia, P.; Shao, Z. TIM-3 expression and its association with overall survival in primary osteosarcoma. Oncol. Lett. 2019, 18, 5294–5300. [Google Scholar] [CrossRef]
  114. Zhu, T.; Han, J.; Yang, L.; Cai, Z.; Sun, W.; Hua, Y.; Xu, J. Immune microenvironment in osteosarcoma: Components, therapeutic strategies and clinical applications. Front. Immunol. 2022, 13, 907550. [Google Scholar] [CrossRef] [PubMed]
  115. Yoshida, K.; Okamoto, M.; Sasaki, J.; Kuroda, C.; Ishida, H.; Ueda, K.; Ideta, H.; Kamanaka, T.; Sobajima, A.; Takizawa, T.; et al. Anti-PD-1 antibody decreases tumour-infiltrating regulatory T cells. BMC Cancer 2020, 20, 25. [Google Scholar] [CrossRef]
  116. Kurita, M.; Yoshihara, Y.; Ishiuji, Y.; Chihara, M.; Ishiji, T.; Asahina, A.; Yanaba, K. Expression of T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain on CD4+ T cells in patients with atopic dermatitis. J. Dermatol. 2019, 46, 37–42. [Google Scholar] [CrossRef] [PubMed]
  117. Mantovani, A.; Bottazzi, B.; Colotta, F.; Sozzani, S.; Ruco, L. The origin and function of tumor-associated macrophages. Immunol. Today 1992, 265, 265–270. [Google Scholar] [CrossRef]
  118. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  119. Hume, D.A.; MacDonald, K.P.A. Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 2012, 119, 1810–1820. [Google Scholar] [CrossRef]
  120. Qin, R.; Ren, W.; Ya, G.; Wang, B.; He, J.; Ren, S.; Jiang, L.; Zhao, S.J.C. Role of chemokines in the crosstalk between tumor and tumor-associated macrophages. Clin. Exp. Med. 2023, 23, 1359–1373. [Google Scholar] [CrossRef] [PubMed]
  121. Zhou, K.; Cheng, T.; Zhan, J.; Peng, X.; Zhang, Y.; Wen, J.; Chen, X.; Ying, M. Targeting tumor-associated macrophages in the tumor microenvironment. Oncol. Lett. 2020, 20, 234. [Google Scholar] [CrossRef]
  122. Mota, J.M.; Leite, C.A.; Souza, L.E.; Melo, P.H.; Nascimento, D.C.; de-Deus-Wagatsuma, V.M.; Temporal, J.; Figueiredo, F.; Noushmehr, H.; Alves-Filho, J.C. Post-sepsis state induces tumor-associated macrophage accumulation through CXCR4/CXCL12 and favors tumor progression in mice. Cancer Immunol. Res. 2016, 4, 312–322. [Google Scholar] [CrossRef]
  123. Deng, X.; Liang, H.; Yang, W.; Shao, Z. Polarization and function of tumor-associated macrophages mediate graphene oxide-induced photothermal cancer therapy. J. Photochem. Photobiol. B Biol. 2020, 208, 111913. [Google Scholar] [CrossRef] [PubMed]
  124. Sylvestre, M. Engineering pH-Responsive Biomaterials for Cancer Immunotherapy; University of Washington: Seattle, WA, USA, 2021. [Google Scholar]
  125. Kumari, N.; Choi, S.H. Tumor-associated macrophages in cancer: Recent advancements in cancer nanoimmunotherapies. J. Exp. Clin. Cancer Res. 2022, 41, 68. [Google Scholar] [CrossRef] [PubMed]
  126. He, Y.; de Araújo Júnior, R.F.; Cruz, L.J.; Eich, C. Functionalized nanoparticles targeting tumor-associated macrophages as cancer therapy. Pharmaceutics 2021, 13, 1670. [Google Scholar] [CrossRef] [PubMed]
  127. Mohanty, S.; Yerneni, K.; Theruvath, J.L.; Graef, C.M.; Nejadnik, H.; Lenkov, O.; Pisani, L.; Rosenberg, J.; Mitra, S.; Cordero, A.S.; et al. Nanoparticle enhanced MRI can monitor macrophage response to CD47 mAb immunotherapy in osteosarcoma. Cell Death Dis. 2019, 10, 36. [Google Scholar] [CrossRef] [PubMed]
  128. Azimnasab-Sorkhabi, P.; Soltani-Asl, M.; Kfoury Junior, J.R. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) as an undetermined tool in tumor cells. Hum. Cell 2023, 36, 1225–1232. [Google Scholar] [CrossRef] [PubMed]
  129. Pagnotti, G.M.; Trivedi, T.; Mohammad, K.S. Translational Strategies to Target Metastatic Bone Disease. Cells 2022, 11, 1309. [Google Scholar] [CrossRef] [PubMed]
  130. Hashimoto, K.; Nishimura, S.; Akagi, M. Characterization of PD-1/PD-L1 immune checkpoint expression in osteosarcoma. Diagnostics 2020, 10, 528. [Google Scholar] [CrossRef] [PubMed]
  131. Nuytemans, L.; Sys, G.; Creytens, D.; Lapeire, L. NGS-analysis to the rescue: Dual checkpoint inhibition in metastatic osteosarcoma–a case report and review of the literature. Acta Clin. Belg. 2021, 76, 162–167. [Google Scholar] [CrossRef]
  132. Xu, C.; Wang, M.; Guo, W.; Sun, W.; Liu, Y. Curcumin in osteosarcoma therapy: Combining with immunotherapy, chemotherapeutics, bone tissue engineering materials and potential synergism with photodynamic therapy. Front. Oncol. 2021, 11, 672490. [Google Scholar] [CrossRef]
  133. Kloosterman, D.J.; Akkari, L. Macrophages at the interface of the co-evolving cancer ecosystem. Cell 2023, 186, 1627–1651. [Google Scholar] [CrossRef] [PubMed]
  134. Ying, H.; Li, Z.-Q.; Li, M.-P.; Liu, W.-C. Metabolism and senescence in the immune microenvironment of osteosarcoma: Focus on new therapeutic strategies. Front. Endocrinol. 2023, 14, 1217669. [Google Scholar] [CrossRef] [PubMed]
  135. Baker, D.J.; Arany, Z.; Baur, J.A.; Epstein, J.A.; June, C.H. CAR T therapy beyond cancer: The evolution of a living drug. Nature 2023, 619, 707–715. [Google Scholar] [CrossRef] [PubMed]
  136. Al-Haideri, M.; Tondok, S.B.; Safa, S.H.; Maleki, A.H.; Rostami, S.; Jalil, A.T.; Al-Gazally, M.E.; Alsaikhan, F.; Rizaev, J.A.; Mohammad, T.A.M. CAR-T cell combination therapy: The next revolution in cancer treatment. Cancer Cell Int. 2022, 22, 365. [Google Scholar] [CrossRef]
  137. Sánchez-Paulete, A.R.; Mateus-Tique, J.; Mollaoglu, G.; Nielsen, S.R.; Marks, A.; Lakshmi, A.; Khan, J.A.; Wilk, C.M.; Pia, L.; Baccarini, A.; et al. Targeting macrophages with CAR T cells delays solid tumor progression and enhances antitumor immunity. Cancer Immunol. Res. 2022, 10, 1354–1369. [Google Scholar] [CrossRef]
  138. Köksal, H.; Müller, E.; Inderberg, E.M.; Bruland, Ø.; Wälchli, S. Treating osteosarcoma with CAR T cells. Scand. J. Immunol. 2019, 89, e12741. [Google Scholar] [CrossRef]
  139. Moreno, C.; Haynie, C.; Cheever, A.; Weber, K.S. Alternative CAR therapies: Recent approaches in engineering chimeric antigen receptor immune cells to combat cancer. Biomedicines 2022, 10, 1493. [Google Scholar] [CrossRef]
  140. Biglari, A.; Southgate, T.D.; Fairbairn, L.J.; Gilham, D.E. Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo. Gene Ther. 2006, 13, 602–610. [Google Scholar] [CrossRef]
  141. Brady, R.V.; Thamm, D.H. Tumor-associated macrophages: Prognostic and therapeutic targets for cancer in humans and dogs. Front. Immunol. 2023, 14, 1176807. [Google Scholar] [CrossRef] [PubMed]
  142. Chen, S.; Lai, S.W.; Brown, C.E.; Feng, M. Harnessing and enhancing macrophage phagocytosis for cancer therapy. Front. Immunol. 2021, 12, 635173. [Google Scholar] [CrossRef] [PubMed]
  143. de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef] [PubMed]
  144. Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. Immunother. Cancer 2018, 6, 56. [Google Scholar] [CrossRef] [PubMed]
  145. Yan, W.; Liu, Z.; Liu, J.; Xia, Y.; Hu, K.; Yu, J. Application of chimeric antigen receptor T cells in the treatment of hematological malignancies. BioMed Res. Int. 2020, 2020, 4241864. [Google Scholar] [CrossRef] [PubMed]
  146. Quiroz-Munoz, M.; Izadmehr, S.; Arumugam, D.; Wong, B.; Kirschenbaum, A.; Levine, A.C. Mechanisms of osteoblastic bone metastasis in prostate cancer: Role of prostatic acid phosphatase. J. Endocr. Soc. 2019, 3, 655–664. [Google Scholar] [CrossRef] [PubMed]
  147. Dagar, G.; Gupta, A.; Masoodi, T.; Nisar, S.; Merhi, M.; Hashem, S.; Chauhan, R.; Dagar, M.; Mirza, S.; Bagga, P.J.; et al. Harnessing the potential of CAR-T cell therapy: Progress, challenges, and future directions in hematological and solid tumor treatments. J. Transl. Med. 2023, 21, 449. [Google Scholar] [CrossRef]
  148. Xia, A.-L.; Wang, X.-C.; Lu, Y.-J.; Lu, X.-J.; Sun, B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: Challenges and opportunities. Oncotarget 2017, 8, 90521. [Google Scholar] [CrossRef]
  149. Chan, G.; Chan, C. Anti-GD2 Directed Immunotherapy for High-Risk and Metastatic Neuroblastoma. Biomolecules 2022, 12, 358. [Google Scholar] [CrossRef]
  150. Chulanetra, M.; Morchang, A.; Sayour, E.; Eldjerou, L.; Milner, R.; Lagmay, J.; Cascio, M.; Stover, B.; Slayton, W.; Chaicumpa, W.; et al. GD2 chimeric antigen receptor modified T cells in synergy with sub-toxic level of doxorubicin targeting osteosarcomas. Am. J. Cancer Res. 2020, 10, 674. [Google Scholar]
  151. Corti, C.; Venetis, K.; Sajjadi, E.; Zattoni, L.; Curigliano, G.; Fusco, N. CAR-T cell therapy for triple-negative breast cancer and other solid tumors: Preclinical and clinical progress. Expert Opin. Investig. Drugs 2022, 31, 593–605. [Google Scholar] [CrossRef]
  152. Zhu, J.; Wan, R.; Huang, W. CAR T targets and microenvironmental barriers of osteosarcoma. Cytotherapy 2022, 24, 567–576. [Google Scholar] [CrossRef]
  153. Gargett, T.; Yu, W.; Dotti, G.; Yvon, E.S.; Christo, S.N.; Hayball, J.D.; Lewis, I.D.; Brenner, M.K.; Brown, M.P. GD2-specific CAR T cells undergo potent activation and deletion following antigen encounter but can be protected from activation-induced cell death by PD-1 blockade. Mol. Ther. 2016, 24, 1135–1149. [Google Scholar] [CrossRef]
  154. Lewis, V.O. IL-11Rα: A novel target for the treatment of osteosarcoma. In Current Advances in Osteosarcoma; Springer: Berlin/Heidelberg, Germany, 2014; pp. 285–289. [Google Scholar]
  155. Zurita, A.J.; Troncoso, P.; Cardó-Vila, M.; Logothetis, C.J.; Pasqualini, R.; Arap, W. Combinatorial screenings in patients: The interleukin-11 receptor α as a candidate target in the progression of human prostate cancer. Cancer Res. 2004, 64, 435–439. [Google Scholar] [CrossRef]
  156. Moonat, H.; Huang, G.; Dhupkar, P.; Schadler, K.; Gordon, N.; Kleinerman, E. Combination of Interleukin-11R [alpha] chimeric antigen receptor T-cells and programmed death-1 blockade as an approach to targeting osteosarcoma cells In vitro. Cancer Transl. Med. 2017, 3. [Google Scholar] [CrossRef]
  157. Fisher, J.P.; Heuijerjans, J.; Yan, M.; Gustafsson, K.; Anderson, J. γδ T cells for cancer immunotherapy: A systematic review of clinical trials. Oncoimmunology 2014, 3, e27572. [Google Scholar] [CrossRef]
  158. Gomes, A.Q.; Martins, D.S.; Silva-Santos, B. Targeting γδ T lymphocytes for cancer immunotherapy: From novel mechanistic insight to clinical application. Cancer Res. 2010, 70, 10024–10027. [Google Scholar] [CrossRef] [PubMed]
  159. Li, Z. Potential of human γδ T cells for immunotherapy of osteosarcoma. Mol. Biol. Rep. 2013, 40, 427–437. [Google Scholar] [CrossRef] [PubMed]
  160. Giordano, G.; Merlini, A.; Ferrero, G.; Mesiano, G.; Fiorino, E.; Brusco, S.; Centomo, M.L.; Leuci, V.; D’Ambrosio, L.; Aglietta, M. Epha2 expression in bone sarcomas: Bioinformatic analyses and preclinical characterization in patient-derived models of osteosarcoma, ewing’s sarcoma and chondrosarcoma. Cells 2021, 10, 2893. [Google Scholar] [CrossRef] [PubMed]
  161. Kiru, L.; Zlitni, A.; Tousley, A.M.; Dalton, G.N.; Wu, W.; Lafortune, F.; Liu, A.; Cunanan, K.M.; Nejadnik, H.; Sulchek, T.; et al. In vivo imaging of nanoparticle-labeled CAR T cells. Proc. Natl. Acad. Sci. USA 2022, 119, e2102363119. [Google Scholar] [CrossRef] [PubMed]
  162. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef] [PubMed]
  163. Bagley, S.J.; Desai, A.S.; Linette, G.P.; June, C.H.; O’Rourke, D.M. CAR T-cell therapy for glioblastoma: Recent clinical advances and future challenges. Neuro-Oncol. 2018, 20, 1429–1438. [Google Scholar] [CrossRef] [PubMed]
  164. O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef] [PubMed]
  165. Tian, H.; Cao, J.; Li, B.; Nice, E.C.; Mao, H.; Zhang, Y.; Huang, C. Managing the immune microenvironment of osteosarcoma: The outlook for osteosarcoma treatment. Bone Res. 2023, 11, 11. [Google Scholar] [CrossRef] [PubMed]
  166. Rodriguez-Garcia, A.; Palazon, A.; Noguera-Ortega, E.; Powell, D.J., Jr.; Guedan, S. CAR-T cells hit the tumor microenvironment: Strategies to overcome tumor escape. Front. Immunol. 2020, 11, 1109. [Google Scholar] [CrossRef]
  167. Parihar, R.; Rivas, C.; Huynh, M.; Omer, B.; Lapteva, N.; Metelitsa, L.S.; Gottschalk, S.M.; Rooney, C.M. NK cells expressing a chimeric activating receptor eliminate MDSCs and rescue impaired CAR-T cell activity against solid tumors. Cancer Immunol. Res. 2019, 7, 363–375. [Google Scholar] [CrossRef] [PubMed]
  168. Katz, S.; Point, G.R.; Cunetta, M.; Thorn, M.; Guha, P.; Espat, N.J.; Boutros, C.; Hanna, N.; Junghans, R.P. Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther. 2016, 23, 142–148. [Google Scholar] [CrossRef]
  169. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef]
  170. Chinnasamy, D.; Yu, Z.; Kerkar, S.P.; Zhang, L.; Morgan, R.A.; Restifo, N.P.; Rosenberg, S.A. Local delivery of lnterleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin. Cancer Res. 2012, 18, 1672–1683. [Google Scholar] [CrossRef]
  171. Andrea, A.E.; Chiron, A.; Mallah, S.; Bessoles, S.; Sarrabayrouse, G.; Hacein-Bey-Abina, S. Advances in CAR-T cell genetic engineering strategies to overcome hurdles in solid tumors treatment. Front. Immunol. 2022, 13, 830292. [Google Scholar] [CrossRef]
  172. Guha, P.; Gardell, J.; Darpolor, J.; Cunetta, M.; Lima, M.; Miller, G.; Espat, N.J.; Junghans, R.P.; Katz, S.C. STAT3 inhibition induces Bax-dependent apoptosis in liver tumor myeloid-derived suppressor cells. Oncogene 2019, 38, 533–548. [Google Scholar] [CrossRef] [PubMed]
  173. Fultang, L.; Panetti, S.; Ng, M.; Collins, P.; Graef, S.; Rizkalla, N.; Booth, S.; Lenton, R.; Noyvert, B.; Shannon-Lowe, C. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers. EBioMedicine 2019, 47, 235–246. [Google Scholar] [CrossRef] [PubMed]
  174. Zong, Z.; Zou, J.; Mao, R.; Ma, C.; Li, N.; Wang, J.; Wang, X.; Zhou, H.; Zhang, L.; Shi, Y. M1 Macrophages Induce PD-L1 Expression in Hepatocellular Carcinoma Cells Through IL-1beta Signaling. Front. Immunol. 2019, 10, 1643. [Google Scholar] [CrossRef]
  175. Adema, G.J.; Hartgers, F.; Verstraten, R.; de Vries, E.; Marland, G.; Menon, S.; Foster, J.; Xu, Y.; Nooyen, P.; McClanahan, T. A dendritic-cell-derived C–C chemokine that preferentially attracts naive T cells. Nature 1997, 387, 713–717. [Google Scholar] [CrossRef] [PubMed]
  176. Schutyser, E.; Struyf, S.; Proost, P.; Opdenakker, G.; Laureys, G.; Verhasselt, B.; Peperstraete, L.; Van de Putte, I.; Saccani, A.; Allavena, P. Identification of biologically active chemokine isoforms from ascitic fluid and elevated levels of CCL18/pulmonary and activation-regulated chemokine in ovarian carcinoma. J. Biol. Chem. 2002, 277, 24584–24593. [Google Scholar] [CrossRef] [PubMed]
  177. Barth, B.M.; Altinoğlu, E.I.; Shanmugavelandy, S.S.; Kaiser, J.M.; Crespo-Gonzalez, D.; DiVittore, N.A.; McGovern, C.; Goff, T.M.; Keasey, N.R.; Adair, J.H.; et al. Targeted indocyanine-green-loaded calcium phosphosilicate nanoparticles for in vivo photodynamic therapy of leukemia. ACS Nano 2011, 5, 5325–5337. [Google Scholar] [CrossRef] [PubMed]
  178. Chen, H.; Luan, X.; Paholak, H.J.; Burnett, J.P.; Stevers, N.O.; Sansanaphongpricha, K.; He, M.; Chang, A.E.; Li, Q.; Sun, D. Depleting tumor-associated Tregs via nanoparticle-mediated hyperthermia to enhance anti-CTLA-4 immunotherapy. Nanomedicine 2020, 15, 77–92. [Google Scholar] [CrossRef] [PubMed]
  179. Cheng, D.; Zhang, Z.; Mi, Z.; Tao, W.; Liu, D.; Fu, J.; Fan, H. Deciphering the heterogeneity and immunosuppressive function of regulatory T cells in osteosarcoma using single-cell RNA transcriptome. Comput. Biol. Med. 2023, 165, 107417. [Google Scholar] [CrossRef] [PubMed]
  180. Barani, M.; Mukhtar, M.; Rahdar, A.; Sargazi, S.; Pandey, S.; Kang, M. Recent advances in nanotechnology-based diagnosis and treatments of human osteosarcoma. Biosensors 2021, 11, 55. [Google Scholar] [CrossRef] [PubMed]
  181. Ge, J.; Yang, N.; Yang, Y.; Yu, H.; Yang, X.; Wang, Y.; Wang, T.; Cheng, S.; Wang, Y.; Han, Z. The combination of eddy thermal effect of biodegradable magnesium with immune checkpoint blockade shows enhanced efficacy against osteosarcoma. Bioact. Mater. 2023, 25, 73–85. [Google Scholar] [CrossRef] [PubMed]
  182. Roy, I. Therapeutic applications of magnetic nanoparticles: Recent advances. Mater. Adv. 2022, 3, 7425–7444. [Google Scholar]
  183. Yu, W.; Wang, Y.; Zhu, J.; Jin, L.; Liu, B.; Xia, K.; Wang, J.; Gao, J.; Liang, C.; Tao, H. Autophagy inhibitor enhance ZnPc/BSA nanoparticle induced photodynamic therapy by suppressing PD-L1 expression in osteosarcoma immunotherapy. Biomaterials 2019, 192, 128–139. [Google Scholar] [CrossRef]
  184. Kong, F.-Y.; Zhang, J.-W.; Li, R.-F.; Wang, Z.-X.; Wang, W.-J.; Wang, W. Unique roles of gold nanoparticles in drug delivery, targeting and imaging applications. Molecules 2017, 22, 1445. [Google Scholar] [CrossRef]
  185. Jin, J.; Yuan, P.; Yu, W.; Lin, J.; Xu, A.; Xu, X.; Lou, J.; Yu, T.; Qian, C.; Liu, B. Mitochondria-targeting polymer micelle of dichloroacetate induced pyroptosis to enhance osteosarcoma immunotherapy. ACS Nano 2022, 16, 10327–10340. [Google Scholar] [CrossRef] [PubMed]
  186. Zhang, Y.; Yuan, T.; Li, Z.; Luo, C.; Wu, Y.; Zhang, J.; Zhang, X.; Fan, W. Hyaluronate-based self-stabilized nanoparticles for immunosuppression reversion and immunochemotherapy in osteosarcoma treatment. ACS Biomater. Sci. Eng. 2021, 7, 1515–1525. [Google Scholar] [CrossRef] [PubMed]
  187. Buondonno, I.; Gazzano, E.; Jean, S.R.; Audrito, V.; Kopecka, J.; Fanelli, M.; Salaroglio, I.C.; Costamagna, C.; Roato, I.; Mungo, E.; et al. Mitochondria-targeted doxorubicin: A new therapeutic strategy against doxorubicin-resistant osteosarcoma. Mol. Cancer Ther. 2016, 15, 2640–2652. [Google Scholar] [CrossRef] [PubMed]
  188. Ouyang, J.; Xie, A.; Zhou, J.; Liu, R.; Wang, L.; Liu, H.; Kong, N.; Tao, W. Minimally invasive nanomedicine: Nanotechnology in photo-/ultrasound-/radiation-/magnetism-mediated therapy and imaging. Chem. Soc. Rev. 2022, 51, 4996–5041. [Google Scholar] [CrossRef] [PubMed]
  189. Gu, Y.-Z.; Zhao, X.; Song, X.-R. Ex vivo pulsed dendritic cell vaccination against cancer. Acta Pharmacol. Sin. 2020, 41, 959–969. [Google Scholar] [CrossRef] [PubMed]
  190. Li, T.; Liu, T.; Zhu, W.; Xie, S.; Zhao, Z.; Feng, B.; Guo, H.; Yang, R. Targeting MDSC for immune-checkpoint blockade in cancer immunotherapy: Current progress and new prospects. Clin. Med. Insights Oncol. 2021, 15, 11795549211035540. [Google Scholar] [CrossRef] [PubMed]
  191. Ory, B.; Blanchard, F.; Battaglia, S.; Gouin, F.; Redini, F.; Heymann, D. Zoledronic acid activates the DNA S-phase checkpoint and induces osteosarcoma cell death characterized by apoptosis-inducing factor and endonuclease-G translocation independently of p53 and retinoblastoma status. Mol. Pharmacol. 2007, 71, 333–343. [Google Scholar] [CrossRef] [PubMed]
  192. Katz, J.B.; Muller, A.J.; Prendergast, G.C. Indoleamine 2, 3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunol. Rev. 2008, 222, 206–221. [Google Scholar] [CrossRef]
  193. Xiang, D.; Han, X.; Li, J.; Zhang, J.; Xiao, H.; Li, T.; Zhao, X.; Xiong, H.; Xu, M.; Bi, W. Combination of IDO inhibitors and platinum (IV) prodrugs reverses low immune responses to enhance cancer chemotherapy and immunotherapy for osteosarcoma. Mater. Today Bio 2023, 20, 100675. [Google Scholar] [CrossRef]
  194. Luo, K.; Li, N.; Ye, W.; Gao, H.; Luo, X.; Cheng, B. Activation of stimulation of interferon genes (STING) signal and cancer immunotherapy. Molecules 2022, 27, 4638. [Google Scholar] [CrossRef] [PubMed]
  195. Wang-Bishop, L.; Wehbe, M.; Shae, D.; James, J.; Hacker, B.C.; Garland, K.; Chistov, P.P.; Rafat, M.; Balko, J.M.; Wilson, J.T. Potent STING activation stimulates immunogenic cell death to enhance antitumor immunity in neuroblastoma. J. Immunother. Cancer 2020, 8, e000282. [Google Scholar] [CrossRef]
  196. Gan, Y.; Li, X.; Han, S.; Liang, Q.; Ma, X.; Rong, P.; Wang, W.; Li, W. The cGAS/STING pathway: A novel target for cancer therapy. Front. Immunol. 2022, 12, 795401. [Google Scholar] [CrossRef] [PubMed]
  197. Xiang, D.; Han, X.; Li, J.; Zhang, J.; Xiao, H.; Li, T.; Zhao, X.; Xiong, H.; Xu, M.; Bi, W. Combination of IDO Inhibitors and Platinum (IV) Prodrugs Via Nanoparticles Reverses Low Immune Responses in Osteosarcoma Through Activation of STING Pathway to Enhance Cancer Chemotherapy and Immunotherapy. Available online: https://papers.ssrn.com/sol3/papers.cfm?abstract_id=4412802 (accessed on 1 June 2023).
  198. Li, M.; Xu, Y.; Sun, J.; Wang, M.; Yang, D.; Guo, X.; Song, H.; Cao, S.; Yan, Y. Fabrication of charge-conversion nanoparticles for cancer imaging by flash nanoprecipitation. ACS Appl. Mater. Interfaces 2018, 10, 10752–10760. [Google Scholar] [CrossRef] [PubMed]
  199. Liu, Y.-F.; Liu, R.; Li, X.-Y.; Song, Z.; Zhao, X.-H. Development of docetaxel and alendronate-loaded chitosan-conjugated polylactide-co-glycolide nanoparticles: In vitro characterization in osteosarcoma cells. Trop. J. Pharm. Res. 2016, 15, 1353–1360. [Google Scholar] [CrossRef]
  200. Hattinger, C.M.; Salaroglio, I.C.; Fantoni, L.; Godel, M.; Casotti, C.; Kopecka, J.; Scotlandi, K.; Ibrahim, T.; Riganti, C.; Serra, M. Strategies to Overcome Resistance to Immune-Based Therapies in Osteosarcoma. Int. J. Mol. Sci. 2023, 24, 799. [Google Scholar] [CrossRef]
  201. Buondonno, I.; Gazzano, E.; Tavanti, E.; Chegaev, K.; Kopecka, J.; Fanelli, M.; Rolando, B.; Fruttero, R.; Gasco, A.; Hattinger, C.J.C.; et al. Endoplasmic reticulum-targeting doxorubicin: A new tool effective against doxorubicin-resistant osteosarcoma. Cell. Mol. Life Sci. 2019, 76, 609–625. [Google Scholar] [CrossRef]
  202. Kopecka, J.; Campia, I.; Brusa, D.; Doublier, S.; Matera, L.; Ghigo, D.; Bosia, A.; Riganti, C.J. Nitric oxide and P-glycoprotein modulate the phagocytosis of colon cancer cells. J. Cell. Mol. Med. 2011, 15, 1492–1504. [Google Scholar] [CrossRef]
  203. Verrecchia, F.; Rédini, F. Transforming growth factor-β signaling plays a pivotal role in the interplay between osteosarcoma cells and their microenvironment. Front. Oncol. 2018, 8, 133. [Google Scholar] [CrossRef]
  204. Passos Barbosa, M.M. Investigating the Role of Indoleamine 2, 3-Dioxygenase 1 and Transforming Growth Factor Beta 1 in Rebound Immune Suppression Following Chemotherapy in Osteosarcoma. Ph.D. Thesis, University of Illinois at Urbana-Champaign, Champaign, IL, USA, 2023. [Google Scholar]
  205. Farooq, A.; Zulfiqar, B.; Asghar, K. Indoleamine 2, 3-Dioxygenase: A Novel Immunotherapeutic Target for Osteosarcoma. J. Cancer Allied Spec. 2023, 9, 501. [Google Scholar] [CrossRef]
  206. Kuo, C.-L.; Chou, H.-Y.; Lien, H.-W.; Yeh, C.-A.; Wang, J.-R.; Chen, C.-H.; Fan, C.-C.; Hsu, C.-P.; Kao, T.-Y.; Ko, T.-M.; et al. A Fc-VEGF chimeric fusion enhances PD-L1 immunotherapy via inducing immune reprogramming and infiltration in the immunosuppressive tumor microenvironment. Cancer Immunol. Immunother. 2023, 72, 351–369. [Google Scholar] [CrossRef] [PubMed]
  207. Zhao, T.-T.; Zhou, T.-J.; Zhang, C.; Liu, Y.-X.; Wang, W.-J.; Li, C.; Xing, L.; Jiang, H.-L. Hypoxia Inhibitor Combined with Chemotherapeutic Agents for Antitumor and Antimetastatic Efficacy against Osteosarcoma. Mol. Pharm. 2023, 20, 2612–2623. [Google Scholar] [CrossRef] [PubMed]
  208. Cersosimo, F.; Lonardi, S.; Bernardini, G.; Telfer, B.; Mandelli, G.E.; Santucci, A.; Vermi, W.; Giurisato, E. Tumor-associated macrophages in osteosarcoma: From mechanisms to therapy. Int. J. Mol. Sci. 2020, 21, 5207. [Google Scholar] [CrossRef] [PubMed]
  209. Zhang, F.; Stephan, S.B.; Ene, C.I.; Smith, T.T.; Holland, E.C.; Stephan, M.T. Nanoparticles that reshape the tumor milieu create a therapeutic window for effective T-cell therapy in solid malignancies. Cancer Res. 2018, 78, 3718–3730. [Google Scholar] [CrossRef] [PubMed]
  210. Xiao, H.; Jensen, P.E.; Chen, X. Elimination of osteosarcoma by necroptosis with graphene oxide-associated anti-HER2 antibodies. Int. J. Mol. Sci. 2019, 20, 4360. [Google Scholar] [CrossRef] [PubMed]
  211. Kopecka, J.; Porto, S.; Lusa, S.; Gazzano, E.; Salzano, G.; Pinzòn-Daza, M.L.; Giordano, A.; Desiderio, V.; Ghigo, D.; De Rosa, G.; et al. Zoledronic acid-encapsulating self-assembling nanoparticles and doxorubicin: A combinatorial approach to overcome simultaneously chemoresistance and immunoresistance in breast tumors. Oncotarget 2016, 7, 20753. [Google Scholar] [CrossRef] [PubMed]
  212. Ge, Y.-X.; Zhang, T.-W.; Zhou, L.; Ding, W.; Liang, H.-F.; Hu, Z.-C.; Chen, Q.; Dong, J.; Xue, F.-F.; Yin, X.-F.; et al. Enhancement of anti-PD-1/PD-L1 immunotherapy for osteosarcoma using an intelligent autophagy-controlling metal organic framework. Biomaterials 2022, 282, 121407. [Google Scholar] [CrossRef] [PubMed]
  213. Mochizuki, Y.; Tazawa, H.; Demiya, K.; Kure, M.; Kondo, H.; Komatsubara, T.; Sugiu, K.; Hasei, J.; Yoshida, A.; Kunisada, T.; et al. Telomerase-specific oncolytic immunotherapy for promoting efficacy of PD-1 blockade in osteosarcoma. Cancer Immunol. Immunother. 2021, 70, 1405–1417. [Google Scholar] [CrossRef]
  214. Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef]
  215. Okła, K.; Czerwonka, A.; Wawruszak, A.; Bobiński, M.; Bilska, M.; Tarkowski, R.; Bednarek, W.; Wertel, I.; Kotarski, J. Clinical relevance and immunosuppressive pattern of circulating and infiltrating subsets of myeloid-derived suppressor cells (MDSCs) in epithelial ovarian cancer. Front. Immunol. 2019, 10, 691. [Google Scholar] [CrossRef]
  216. Gielen, P.; Schulte, B.; Kers-Rebel, E.; Verrijp, K.; Petersen-Baltussen, H.; ter Laan, M.; Wesseling, P.; Adema, G. Increase of Both CD14+ and CD15+ MDSC Subpopulations in the Blood of Glioma Patients but Predominance of CD15+ MDSCs in Glioma Tissue. 2016. Available online: https://repository.ubn.ru.nl/bitstream/handle/2066/160839/160839.pdf?sequence=1#page=71 (accessed on 1 November 2016).
Figure 1. Immunosuppressive OS-TME: Immune-related cells including OS-M, CTL, Treg, MDSC, and OS-N closely work with each other, facilitating the formation of immunosuppressive OS-TME that benefits the proliferation of OS. The MDSC, OS-M, and Treg benefit angiogenesis via VEGF pathways. The OS-TME favors the cancer metastasis process from the primary bone site to the distant lung site. Created with biorender.com, access on 25 October 2023.
Figure 1. Immunosuppressive OS-TME: Immune-related cells including OS-M, CTL, Treg, MDSC, and OS-N closely work with each other, facilitating the formation of immunosuppressive OS-TME that benefits the proliferation of OS. The MDSC, OS-M, and Treg benefit angiogenesis via VEGF pathways. The OS-TME favors the cancer metastasis process from the primary bone site to the distant lung site. Created with biorender.com, access on 25 October 2023.
Pharmaceutics 16 00251 g001
Figure 2. Illustration of TAM therapies: traditional approaches and engineering cell approaches. Traditional therapies involve established methods inhibiting OS-M recruitment and function. These approaches aim to eliminate cancer cells and modulate the immune response. The corresponding elements in the figure highlight the key components and interactions involved in traditional approaches.
Figure 2. Illustration of TAM therapies: traditional approaches and engineering cell approaches. Traditional therapies involve established methods inhibiting OS-M recruitment and function. These approaches aim to eliminate cancer cells and modulate the immune response. The corresponding elements in the figure highlight the key components and interactions involved in traditional approaches.
Pharmaceutics 16 00251 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cheng, S.; Wang, H.; Kang, X.; Zhang, H. Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress. Pharmaceutics 2024, 16, 251. https://doi.org/10.3390/pharmaceutics16020251

AMA Style

Cheng S, Wang H, Kang X, Zhang H. Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress. Pharmaceutics. 2024; 16(2):251. https://doi.org/10.3390/pharmaceutics16020251

Chicago/Turabian Style

Cheng, Shigao, Huiyuan Wang, Xuejia Kang, and Hui Zhang. 2024. "Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress" Pharmaceutics 16, no. 2: 251. https://doi.org/10.3390/pharmaceutics16020251

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