1. Introduction
Osteosarcoma (OS) is a bone cancer that begins with a tumour secreted by the abnormal growth of the osteoid substances and immature bone [
1]. OS primarily affects adolescents and elders in the age ranges of 15–19 [
2] and over 65 [
3], respectively. Although OS is a rare type of childhood cancer, it accounts for 3-5% of children's carcinomas and nearly 0.2% of all malignant tumours [
4]. It has been revealed that a substantial (25%) portion of OS paediatric patients, while seeking medical help first, have metastases [
5]. Around 15–20% of OS metastasis cases presented with the pulmonary tree and had an inferior clinical outcome [
6]. Only about 20–30% of such metastatic cases live for a prolonged period, compared with 65–70% of localised cases [
7].
Chemotherapy [
8], immunotherapy [
9], inhibitory therapy [
10], and surgery [
11] are the standard treatments for OS. Without proper treatments, OS diseases will progress to malignant tumours [
12]. Over the course of five years, the overall and event-free survival rates are approximately 71% and 54%, respectively [
13]. Due to the recurrence and metastasis incidences [
14], the postmetastasis course over the five years for the overall and event-free survival rates are 38.1 ± 6.4% and 25 ± 5.3%, respectively [
15]. Due to the fact that OS is the most common malignant bone tumour in adolescents, the 5-year survival rate drops to 70% and further decreases to 20–30% for patients with metastases [
1]. Even after numerous trials done for nearly four decades, there has been no significant improvement in either localised or metastatic OS therapy [
16]. In conclusion, the main cause of mortality is pulmonary metastases [
15], which cause more than 90% of deaths [
17].
Immunotherapy demonstrated high efficacy in clinical trials for tumorigenesis therapy [
18], but after long-term treatments, it develops severe immunosuppressive and chemotherapeutic resistance in the tumour microenvironment (TME) [
8]. This is due to OS’s innate and acquired nature, which causes the therapy’s progression to stall [
19]. Scientists and clinicians have attempted to solve these problems for the past thirty years [
20], but 30% of OS patients still do not respond to these standard treatments [
21]. Therefore, combinational and targeted therapies [
22,
23] are developed for these problems and has a higher response rate [
24]. However, their evidence of efficacy, long-term use [
25], and personalised precision medicine [
26] remain inadequate [
27]. There is some evidence showing that immunosuppressive and chemotherapeutic resistance (chemoresistance) developed into immune evasion and multidrug resistance (MDR), respectively [
28,
29,
30]. Despite these well-known immunotherapies, immune evasion, and chemoresistance, OS metastasis remains the primary barrier to achieving therapy efficacy [
8]. Thus, more efficacious therapies and alternative therapeutic approaches are needed to intensify the treatments and achieve higher cure rates [
31]. However, not much research has successfully provided an in-depth understanding of the relationship between tumorigenesis, metastasis, immune evasion, and chemoresistance.
Because the four stages are intertwined and complex [
32], the focus of this review is on clarifying their relationship and encounter therapies. First of all, the status quo of OS therapies is presented, along with their historical development and clinical advancements. A comprehensive timeline is drawn to demonstrate significant discoveries and advancements in osteosarcoma studies. Besides, the summary of completed years for sarcoma clinical trials is tabulated to highlight the seminal discoveries and major clinical triumphs. In the content, the bidirectional relationship between these four OS mutation stages—tumorigenesis, metastasis, immune evasion, and chemoresistance—is clearly described. Further, short and precise definitions are given to each of them to reach a common understanding. Hereafter, their intertwined therapies could be discussed individually. Herein, it is notable that many clinically relevant therapies nowadays are combinational and multifunctional in order to cure the complex OS stages. Notably, intertwined therapy is a fact that should not be overlooked; however, it is prudent to discuss them by reconstructing them to elaborate precisely. Through this review, the stages and therapies of OS are precisely defined and clearly elucidated, which are expected to serve as guidance for scientists and clinicians.
2. Status Quo of Osteosarcoma Therapies
2.1. Historical Development
The significant discoveries and advancements in OS studies are drawn on a timeline as shown in
Figure 1. The first in vivo test was done by Evans and Long (1922), who investigated the characteristic effects of the OS using the fresh anterior lobe [
33]. After 42 years, the first gene cloned using human growth hormone (HGH) was done by Li and Liu (1964) [
34]. After many trials, Jaffe et al. (1978) intended to use high-dose methotrexate to treat OS [
35]. Rosen et al. (1982) pioneered preoperative adjuvant chemotherapy for OS to address the compliance issue associated with the previous high-dose therapy [
36]. Furthermore, Zapf et al. (1984) investigated the biological and immunological properties of OS using insulin-like growth factors (IGF) [
37]. The first in vitro test was done by Stashenko et al. (1987), who investigated a bone inhibitor using Interleukin (IL)-1β [
38]. Bengtsson et al. (1993) were the first to use the recombinant technology for HGH [
39]. Gentet et al. (1997) were the first to use the combined chemotherapy of etoposide and ifosfamide (EnI) that is commonly used in childhood OS now [
40]. McGary et al. (2002) were the first to use the Tyrosine Kinase Inhibitor STI571 in targeted OS therapy [
41]. Nardin et al. (2006) were the first to intend to target and activate macrophages by using liposomal muramyl tripeptide phosphatidylethanolamine in immunotherapy [
42]. The OS stem cell studies using salinomycin inhibitor were pioneered by Tang et al. (2011) [
43]. Finally, Gordon et al. (2019) were the first to successfully introduce an ABI-009 (nab-sirolimus) drug carrier for preliminary efficacy and safety results in the OS [
44].
2.2. Clinical Advancement
OS research is extremely hard and remains a global challenge. Despite the fact that many clinical trials had begun, the majority of them could not be completed. For the past twenty years, only ten clinical trials with federal government clinical trial identifiers (GCTI) have been successfully completed. Thus, the sarcoma types completed their clinical trials in years with API and primary tests as shown in
Table 1. For all these trials, there are only two main types of OS: soft and solid, as observed in the table. The following active pharmaceutical ingredients (API) are appropriate for OS: topotecan (Tpt) [
45], pazopanib (Pzp) [
46], placebo (Plb) [
47], gemcitabine (Gct) [
48], M6620 [
49,
50], regorafenib (Rgf) [
51,
52], glembatumumab vedotin (GV) [
53,
54], lenvatinib [
55], etoposide and ifosfamide (EnI) [
56], nab-rapamycin (Rpm) [
57,
58], cyclophosphamide (Cfa) [
59], simvastatin (Sim) [
60], myeloid growth factor (MGF) [
61], nab-paclitaxel [
62,
63], methotrexate (Mtx) [
64], and doxorubicin (Dox) [
65]. Besides, the primary tests, such as laboratory biomarker analyses [
66] and dose escalation studies [
67], were conducted for these successful triumphs. As a result, these two primary tests indicate the two main investigation parameters.
3. Osteosarcoma Bidirectional Mutation Stages
OS therapies are difficult because they progress and reverse through four mutation stages and are intertwined, including tumour microenvironment (TME), metastasis, immune evasion, and chemotherapeutic resistance, as shown in
Figure 2. The bidirectional complexity of progression and reversion in OS mutation stages is influenced by the exosomes of tumour, stem, mesenchymal, immune, fibroblast, and endothelial cells [
68]. There is mounting evidence that signal molecules such as neurotransmitters, enzymes, hormones, and nucleic acids [
69] are involved in the angiogenesis, growth, migration, metastasis, and apoptosis of the above mentioned cells, involving intercellular cell communication, body regulation, and immune responses [
70]. In this cellular communication, extracellular vesicles (EV) play a key role [
71], which could be derived by various cells such as OS cells, mesenchymal stem cells (MSC), adipose-derived MSC (ADMSC), cancer-associated stromal fibroblasts (CAF), and macrophages [
72]. These EV regulate the activity of recipient cells, including angiogenesis, proliferation, invasion, migration, metastasis, chemotherapeutic resistance, and apoptosis, by using their cargoes of proteins, DNA, and RNA [
73]. These three cargoes have distinct metabolic dynamics [
74] including a connection with the EV components’ biogenesis machinery, a cellular homeostasis regulator with cytoplasmic DNA sensor activation, and parental cell function efficiency at different states [
75]. The creation of biomarker vehicles [
76] that employ the aforementioned pro-tumorigenic components and signalling pathways to circulate immune responses from OS cancer diseases remains a significant clinical trial challenge [
77].
3.1. Tumorigenesis
TME is composed of EV secretion cells, MSC, and tumour cells. The EV cells are secreted by MSC and immune cells to alter macrophage phenotype-2 (M2) [
78] and regulate tumour progression via the Wnt signalling pathway [
79]. Furthermore, the EV cells are the paracrine factors secreted by human bone marrow MSC (BMSC), such as osteoclasts, osteoblasts, and endothelial cells, to regulate tumour cells via communication with the Hedgehog signalling pathway [
80]. As a result, the M2 and tumour cells regulate TME by EV secretions in order to promote angiogenesis, growth, and metastasis [
81,
82]. Basically, the tumorigenesis research in TME is concerned with how coexisting cells interact and communicate with one another.
3.2. Metastasis
The metastasis potential is affected by the communication between the stressed MSC and the micro RNA (miRNA) content of EV. Tumour cells metastasize in three ways: by secreting EV, by influencing TME, and by mediating the transformation of distant MSC. Direct EV secretion by osteoblasts and CAF can improve migrability. The induction of metastasis can be influenced by regulating tumour and MSC oncogenic phenotypes in TME. The pro-angiogenic factors from endothelial cells can mediate EV transformation to modulate cell invasiveness and promote metastasis. Metastasis could be activated either by modulating tumour-associated macrophage (TAM) cellular signalling to promote the M2 or by producing transforming growth factor beta (TGFβ)-2 to create an immunosuppressive and pro-TME [
83]. As a result, the tumour cells could be metastasized by inducing a pro-metastatic and tumorigenic phenotype and mediating transformation into local or distant cells.
3.3. Immune Evasion
The immune system is divided into innate and adaptive immunizations, which are always related to the bone microenvironment [
84]. The immune evasion occurred because the inefficient immune cells allowed the tumours to evade the immune surveillance systems or the host immune checkpoint through multiple mechanisms [
24]. This inefficiency induced a tolerance for the T-cell receptor (TCR), resulting in a dormant response to tumour recognition [
85]. Therefore, the tumour cancer cells in TME escaped immunotherapy. However, this peripheral tolerance of host-cell immune responses is protected by regulating T regulatory cells (T
reg) to prevent autoimmune disorders [
86]. In fact, two major mechanisms induce immune tolerance [
87]: T-cell-mediated inflammation suppression and no tumour signals received by the major histocompatibility complex (MHC) antigen presentation [
88]. Traditionally, the plasma protease thrombin cleaves glycoprotein A repetitions predominant (GARP) in tumour immune evasion to release active TGFβ [
89]. TGFβ is the main coordinator and mediator between both mechanisms mentioned in immune evasion [
90]. TGFβ increased programmed cell death protein (PD) ligand-1 expression on TAM [
91] to bind with PD-1 (CD279) for cytotoxic T lymphocyte-associated antigen (CTLA)-4 inhibition. CTLA4 (CD152) is a membrane glycoprotein of immunosuppressive T
reg that binds to costimulatory molecules CD80 and CD86 to inhibit early T cell (CD8+ and CD4+) activation [
92]. These T cells are anti-tumour cells that respond to cancer-associated fibroblasts (CAFs) for immune evasion regulation [
93].
3.4. Chemoresistance
Chemoresistance is chemotherapeutic resistance, resulting in a chemotherapeutic efficacy deficit [
94]. It always results in cytotoxic agents being minimally delivered or severely off-target, destroying therapeutic compliance effects [
95]. Chemoresistance in cancer cells can be either inherent or acquired, with the latter increasing proportionally with the duration of the therapy [
96]. Chemoresistance is commonly known as MDR, which is drug resistance to Mtx, Dox, and CDDP drugs [
97]. Drug accumulation in clone and stem cells altered TME, leading to mutation and decreased drug sensitivity [
98]. For instance, chemoresistance decreased Dox sensitivity, resulting in M2 induction, which caused tumour cells to spread without responsiveness to Dox [
99]. However, the sensitivity of drugs can be induced by the transfer of specific bioactive molecules, such as non-coding RNA and proteomic signatures [
100].
4. Recent Osteosarcoma Therapies
Because there have been numerous OS therapies over the last four decades, only the five most recent years are considered below. Despite the fact that many OS therapies have been developed, their individual and combinational mechanisms are dispersed [
101]. Therefore, a schematic is drawn to elucidate their recent medicines and therapy mechanisms in OS, as shown in
Figure 3. Medicines used to inhibit and suppress tumorigenesis, metastasis, immune evasion, and chemoresistance via communication mediums [
102]. Targeted therapies can be developed to intensify the therapies and achieve higher cure rates by having a thorough understanding of the roles of genes in communication axes and signalling pathways [
103].
4.1. Tumorigenesis Therapies
The tumorigenesis therapies are generally medicated in connection with the suppressive, regulative, and inhibitive treatment mechanisms [
104]. There is a summary of ten recent studies that have addressed tumorigenesis with medicines for their treatment mechanisms, as shown in
Table 2. For instance, six studies used suppressive mechanism treatments to halt tumorigenesis’ proliferation, migration, and invasion. Collagen beta (1-O) galactosyl transferase 2 (COLGALT2) inhibitor [
105,
106], transformer 2β (Tra2B) [
107,
108], and ArfGAP with GTPase domain 1 (AGAP1) [
109,
110] were used as the medicines to suppress ADMSC exosomes, miR-206, and miR-1307, respectively. In studies of chondrogenesis like osteoclast differentiation and bone resorption activity, the miR-148a and miR-21-5p EVs were used increase their genes to mimic umbilical vein endothelial cell (UVEC) formation in TME [
81,
111]. Furthermore, long non-coding RNA (lncRNA) leukaemia inhibitory factor receptor antisense RNA1 (LIFR-AS1) inhibitor was used to inhibit miR-29a in the the nuclear factor IA (NFIA) axis to suppress human peripheral-blood monocytes (THP-1)-induced macrophage-derived exosomes (MDE) [
112,
113]. All ten studies are related to the exosomal-derived genes and these could be used as diagnosis and prognosis markers in OS progression [
114].
4.2. Interfere Communication Mediators’ Therapies
Metastasis is stimulated and controlled by intercellular communication in endothelial cells [
115]. By interfering with their direct and indirect communication mediators against endothelial changes, both stages can be inhibited [
116]. There is a summary of twenty recent studies that have addressed metastasis with medicines that interfere with communication mediators in signalling pathways, as shown in
Table 3. For instance, twelve studies interfered with communication mediators using inhibitors, including AXL receptor tyrosine kinase (AXL) [
117,
118], miR-135a-5p [
119,
120], messenger RNA (mRNA) B-cell lymphoma-6 (BCL6) [
121,
122], TGFβ1 [
123,
124], T-cell immunoglobulin and mucin-domain containing protein-3 (Tim-3) [
125,
126], and suppressor of cytokine signalling-5 (SOCS5) [
127,
128]. These inhibitors interfered with miR-29a-3p, BMSC-derived exosomal lymphocyte cytosolic protein-1 (LCP1), miR-101 EV, CRISPR-associated protein-9 (Cas9), M2 mediation, and signal transducers and activators of transcription (STAT)-1 mediation by suppressing long intergenic non-protein coding RNA (linc)-00852 in the jumonji and AT-rich interaction domain containing 2 (JARID2) axis; neuregulin receptor degradation protein-1 (NRDP1) in the Janus kinase-2 (JAK2)/STAT3 signalling pathway; ADMSC-derived miR-101; CAF and α-smooth muscle actin expression and fibronectin (ASMAFN) differentiation; IL-10, TGFβ, and vascular endothelial growth factor (VEGF) secretions; and collagen type VI alpha 1 (COL6A1) from histone3 lysine27 acetylation (H3K27ac) activated in CAF conversion with IL-6 and IL-8 secretions; respectively.
The other eight studies interfered with communication mediators using other medicines, such as cancer susceptibility 15 (CASC15) or KLF3 antisense RNA 1 (KLF3-AS1) [
129,
130], programmed cell death 4 (PDCD4) [
131,
132], autophagy-related gene 5 (ATG5) [
133,
134], and Rab22a-NeoF1 fusion protein [
135,
136]. Other medicines interfered with Ras-associated binding 14 (RAB14), extracellular signal-regulated kinase-1/2 (ERK1/2) signalling pathway, oncogenic autophagy, and M2 with Arginylglycylaspartic acid (RGD) peptide internalisation in STAT3 by suppressing miR-338-3p; miR-208a EV; BMSC-derived EV; and protein tyrosine kinase-2 (PYK2) and Ras homolog family member A (RhoA); respectively. All of the medicines used in the 20 studies interfered with communication mediators related to EV secretions or protein expression.
4.3. Immune Evasion Therapies
The suppression and communication barriers of immune cells allow immune evasion [
137]. Tumour cells escape being destroyed by the immune system because the immune cells, such as neutrophils, monocytes, macrophages, dendritic cells, natural killer cells, and B and T lymphocytes, are suppressed [
138]. Besides, the tumour immune responses are barred from immune checkpoint activations, thereby causing immune evasion [
139]. Therefore, the immune evasion is eliminated by suppressing and inhibiting the both mechanisms. There is a summary of twenty recent studies with medicines and prevention mechanisms for immune evasion, as shown in
Table 4. For instance, twelve studies used medicines to suppress immune evasion mechanisms, such as mRNA N-cadherin [
140,
141], anti-IL-9 [
142,
143], ubiquitin-specific peptidase 12 (USP12) inhibitor [
144,
145], immunoglobulin G1 monoclonal antibody (IgG)-4
+ B-cells [
146,
147], latency-associated peptide domain (LAP) inhibitor [
148,
149], and anti-Wnt2 mAb [
93,
150]. These medicines suppressed PD-L1, PD-1, IL-4, IL-10, monocytic myeloid-derived suppressor cells (M-MDSC), NO synthase, and CAF in order to activate CD8
+ T cells and TCR. The remaining eight studies used medicines to inhibit immune evasion mechanisms, such as anti-αvβ8 integrin (also known as ADWA-11) [
151,
152], hexokinase-2 (HK2)-mediated phosphorylation of i-kappa-b-alpha (IκBα) [
153,
154], indoleamine 2,3-dioxygenase (IDO) inhibitor with NO [
155,
156], and TGFβ receptor II (TGFβRII) with anti-IgG1 (also known as bintrafusp alfa) [
157,
158]. These medicines inhibited the expression of TGFβ, TGFβ1, PD-L1, and glycolysis in order to activate CD8
+ T cells and T
reg cells. In all 20 studies, ignoring the immune evasion due to innate and adaptive immunizations, the medical therapies are focused on the activation of immune cells, such as CD8
+ T cells, TCR, and T
reg cells. As a result, immune evasion is prevented by checkpoint blockade therapy [
159], such as PD-L1, PD-1, IL-4, IL-10, M-MDSC, NO synthase, CAF, TGFβ, and TGFβ1 [
160].
4.4. Chemoresistance Therapies
Chemoresistance therapies are always accompanied by treatments for severe off-target effects to restore therapeutic compliance [
161]. The acquired nature of chemoresistance in cancer cells will minimise the cytotoxic agent’s delivery proportionally to the chemotherapeutic treatment duration [
162]. Hence, this becomes a primary challenge for therapeutic agents and cellular lesions in osteosarcoma therapy [
8]. Drug accumulation in cells, intracellular detoxification, apoptosis, DNA repair, signal transduction disruption, and tumour stem cell immunity all contribute to chemoresistance [
163]. The majority of therapeutic approaches involve inhibition of the oncogene’s expression to interfere with or mute the communication pathways or axis [
164]. Some use drug carriers to avoid rapid drug clearance and prolong release [
165,
166]. As a result, chemoresistance therapies should focus on oncogene inhibition, drug influx and efflux, and drug carriers [
167].
Chemoresistance therapies are divided into two types: inhibitor therapies and gene knockdown therapies. There is a summary of ten recent studies that used inhibitor therapies for different types of drug resistance and their chemoresistance prevention in osteosarcoma cells, as shown in
Table 5. For instance, there are four studies focused on reducing folate receptors for Mtx and Dox drug resistance. Dihydrofolate reductase (DHFR) [
168,
169] and folylpoly-γ-glutamate synthetase (FPGS) [
170,
171] inhibitors were used to induce cancer cell apoptosis and inhibit the interaction of spindle and kinetochore associated complex subunit 1 (SKA1) and RNA polymerase II subunit 3 (RPB3), respectively. DHFR reduced the affinity of methotrexate resistance by converting dihydrofolate to tetrahydrofolate in order to inhibit purine and thymidine synthesis, resulting in a deficit in DNA replication and apoptosis. Another two studies of cis-diamminedichloroplatinum (II) (CDDP or cisplatin) drug resistance used the heat shock protein (HSP)-90AA1 gene inhibitor [
172,
80] to deactivate autophagy activating kinase 1 (Ulk1) in FUN14 domain-containing protein 1 (FUNDC1) mediation for mitophagy activation to induce apoptosis. Mitophagy is mitochondrial removal through autophagy, which allows tumour cells to survive cellular stress by clearing damaged organelles and proteins. Two studies of P-glycoprotein (PGP) were inhibited by the inverse agonist XCT-790 or the anlotinib tyrosine kinase (ATK) inhibitor [
173,
174] for mRNA ATP-binding cassette subfamily B member 1 (ABCB1) in the oestrogen-related receptor alpha (ERRα) axis. Another two studies of ABCB1 in the ERRα axis were inhibited by insulin-like growth factor 1 (IGF1) [
175,
176] in order to reverse metabolic disorders. As a result, folate receptors, FUNDC1-mediated Ulk1, and ABCB1 in the ERRα axis are the key targets in chemoresistance therapies.
Despite the above key targets in chemoresistance therapies, the expression of siRNA oncogenes has been popularly used recently to interfere with or mute the communication pathways or axis [
177]. There is a summary of twenty recent studies that used siRNA gene knockdown therapies for different types of drug resistance and their chemoresistance prevention in osteosarcoma cells, as shown in
Table 6. Transmitting circular RNA (circ_) is used to prevent the Mtx, Dox, and CDDP drug resistance in the four, ten, and six studies, respectively. For Mtx instances, the circ_0000073 [
178,
179] and circ_0081001 [
180,
181] gene knockdowns inhibited the N-Ras pathway by sponging miR-145-5p and miR-151-3p and the transglutaminase-2 (TGM2) axis by miR-494-3p, respectively. For Dox instances, the gene knockdowns of circ_0004674 [
182,
183], circ_0001721 [
184,
185], circ_SAMD4A (sterile alpha motif domain) [
186,
187], circ_0002060 [
188,
189], and circ_0003496 [
190,
191] inhibited the fibrillin-1 axis by sponging miR-342-3p, the transcription factor 4 (TCF4) axis by miR-758, the Krüppel-like factor (KLF)-8 axis by miR-218-5p, the ABCB1 axis by miR-198, and the KLF12 axis by miR-370, respectively. For CDDP instances, the gene knockdowns of circ_CHI3L1.2 (chitinase 3-like 1.2) or lncRNA OPI5-AS [
192,
193], circ_TADA2A (transcriptional adaptor 2A) [
194,
195], and circ_103801 [
196,
197] inhibited the lysophosphatidic acid acyltransferase β (LPAATβ) axis by sponging miR-340-5p, the yes-associated protein (YAP) and trichorhinophalangeal syndrome 1 (TRPS1) axis by miR-129-5p, and the MDR-associated protein 1 and PGP, respectively. As a result, the communication pathways of chemoresistance in osteosarcoma cells would be more effectively prevented by the therapies targeting oncogene expression with their knockdowns.
5. Conclusions
Despite the innate and acquired nature of OS, its progression is intertwined, including cycles of tumorigenesis, metastasis, immune evasion, and chemoresistance. Firstly, tumorigenesis is the result of M2 alterations, which are progressed via signalling pathways by the MSC- and immune cell-secreted EV. Secondly, metastasis is potently affected by the communication between the stressed MSC and the miRNA content of EV. Thirdly, immune evasion occurred because tumour cells evaded the host immune checkpoint through the TCR tolerance mechanism, resulting in Treg in autoimmune disorders. Finally, chemoresistance causes cytotoxic agents to be delivered severely off-target, resulting in a chemotherapeutic efficacy deficit. These four stages of progression are treated by the combinational and multifunctional therapies listed below. Ten tumorigenesis therapy studies have been conducted using medicines such as COLGALT2 inhibitors, Tra2B, and AGAP1, miR-148a and miR-21-5p EVs, and the lncRNA LIFR-AS1 inhibitor. The mechanisms of tumorigenesis were being suppressed, regulated, and inhibited, such as proliferation, migration, invasion, chondrogenesis, and UVEC formation. Their targets include ADMSC exosomes, miR-206, miR-1307, miR-148a, miR-21-5p, and miR-29a in the NFIA axis. Metastasis therapies are treated with medicines related to EV secretions and protein expression for intercellular communication in endothelial cells. There have been twenty therapy studies using inhibitor and disruptor medicines to inhibit pro-tumorigenic expression and disrupt signalling pathways. AXL, miR-135a-5p, mRNA BCL6, TGFβ1, Tim-3, and SOCS5 are the inhibitor medicines. These medicines inhibit miR-29a-3p and linc-00852 in the JARID2 axis, LCP1 and NRDP1 in the JAK2/STAT3 signalling pathway, miR-101 EV, Cas9 in CAF and ASMAFN differentiation, IL-10, TGFβ, and VEGF secretions for M2, and CAF conversion with COL6A1 and H3K27ac in the STAT1 signalling pathway. CASC15, KLF3-AS1, PDCD4, ATG5, and Rab22a-NeoF1 are the disruptor medicines. These medicines disrupt RAB14 by miR-338-3p, the ERK1/2 signalling pathway by miR-208a EV, oncogenic autophagy by BMSC-derived EV, and M2 with RGD in STAT3 by PYK2 and RhoA. Immune evasion is treated by activating CD8+ T cells and connecting TCR and Treg cells for immune checkpoint activations and communication checkpoint regulations, respectively. Twenty therapy studies have been conducted using medicines to suppress and inhibit immune evasion mechanisms. These medicines are mRNA N-cadherin, anti-IL-9, USP12 inhibitor, IgG-4+ B-cells, LAP inhibitor, anti-Wnt2 mAb, anti-αvβ8 integrin, HK2-mediated IκBα, IDO inhibitor with NO, and TGFβRII with anti-IgG1. Medical targets include PD-L1, PD-1, IL-4, IL-10, M-MDSC, NO synthase, CAF, TGFβ, and TGFβ1. Chemoresistance therapies use oncogene inhibition as well as drug carriers for influx and efflux to repair immune therapies and disrupt communication pathways. There are thirty studies of chemoresistance therapies focused on Mtx, Dox, and CDDP drug resistance by using inhibitor therapies and gene knockdown therapies. The inhibitors are DHFR, FPGS, HSP-90AA1, XCT-790, ATKI, and IGF1. The inhibitor’s targets are folate receptors, FUNDC1-mediated Ulk1, and ABCB1 in the ERRα axis. Besides, the gene knockdowns are circ_0000073, circ_0081001, circ_0004674, circ_0001721, circ_SAMD4A, circ_0002060, circ_0003496, circ_CHI3L1.2 or lncRNA OPI5-AS, circ_TADA2A, and circ_103801. The gene knockdown’s targets are miR-145-5p and miR-151-3p in the N-Ras pathway, miR-494-3p in the TGM2 axis, miR-342-3p in the fibrillin-1 axis, miR-758 in the TCF4 axis, miR-218-5p in the KLF-8 axis, miR-198 in the ABCB1 axis, miR-370 in the KLF12 axis, miR-340-5p in the LPAATβ axis, miR-129-5p in the YAP/TRPS1 axis, and the MDR-associated protein 1 and PGP. In conclusion, all these OS therapies are individually elucidated to treat tumorigenesis, metastasis, immune evasion, and chemoresistance, but their OS mutation stages are bidirectional and intertwined, resulting in their being combinational and multifunctional.
6. Challenges and Future
OS is an unusual and complicated malignant tumour that necessitates an integrative and interdisciplinary therapeutic approach [
198]. It has been reported that a multidisciplinary approach requires collaboration and cooperation between paediatric or medical cancer specialists, surgeons, pathologists, radiologists, and radiotherapists [
5]. Thus, several models of OS neoplasm have diverse clinical outcomes [
199], making the diagnosis and treatment of OS cancer extremely challenging. Thereby, the therapeutic regimen for OS patients has not been systematized, harmonized, or standardized [
200]. It has been reported that thorough surgical eradication of all sites of primary and metastatic OS is obligatory, foretelling better clinical end-results and continuity of quality life [
5]. However, those OS cases have several primary and metastatic OS disease locations that are not manageable for total surgical resection and result in impecunious clinical consequences. Furthermore, preoperative chemotherapies cause chemoresistance, resulting in a two-fold increase in the cisplatin capability of mutational load in OS cases [
201]. As a result, chemotherapeutic regimens for recurring or replacement cases of chemotherapy resistance are constantly being improved [
202].
According to a recent study, the genetic framework and oncogenesis process of OS are largely unknown, which is impeding research efforts. The immune microenvironment of OS tumours has been extensively studied. It found that OS possesses noticeable diversity and a complicated all-around mode of process regarding malignancy continuation and metastasis [
138]. Another study reported that MDSCs massively invade OS tumours and promote anti-cancer immune-suppressive activities [
138,
203,
204]. Research studies said that preoperative chemotherapy agents, e.g., dox, CDDP, and ifosfamide, effectively brought down the MDSC count in OS cases and, after that, augmented both immune sensitivities and the overall immune system [
205]. Metformin has been shown in studies to effectively reduce OS tumour progression and size. Metformin also shows substantial activity in reducing polymorphnuclear MDSC; nevertheless, no considerable variability was observed for M-MDSC [
206]. Sodium-glucose cotransporter 2 (SGLT2) is a principal intercessor of epithelial glucose transport. It has been proclaimed that SGLT2 is vigorously and exaggeratedly exhibited in several malignant tumour cells, including OS [
207]. Antagonizing overexpressed SGLT2 appreciably hinders cancer advancement, e.g., breast cancer, cervical cancer, hepatocellular cancer, prostate cancer, and lung cancer [
208]. Although the anti-malignant pharmacodynamics of SGLT2 antagonists in OS malignancy remain imprecise [
207,
208,
209]. This narrative review advocates more research regarding this malignancy and safeguards our children and adults from the atrocities of this cancer.
Author Contributions
Formal analysis, Y.Y.L. and M.H.; resources, Y.Y.L. and M.H.; writing—original draft preparation, Y.Y.L.; writing—review and editing, and M.H., A.M.A.Z., and A.M.; supervision, A.M.A.Z. and A.M.; project administration, A.M.A.Z. and A.M.; funding acquisition, A.M.A.Z. and A.M. All authors have read and agreed to the published version of the manuscript.
Funding
The work was funded by the Ministry of Higher Education (MOHE) Malaysia, via the Fundamental Research Grant Scheme (FRGS), grant number FRGS/1/2021/STG01/UPNM/01/1.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
Not applicable.
Acknowledgments
The authors wish to express their gratitude to the staff at the National Defence University of Malaysia and the MOHE Malaysia for the technical support given and for the approved funds, respectively.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Moukengue, B.; Lallier, M.; Marchandet, L.; Baud’huin, M.; Verrecchia, F.; Ory, B.; Lamoureux, F. Origin and Therapies of Osteosarcoma. Cancers (Basel). 2022, 14, 3503. [CrossRef]
- Stiller, C.A.; Bielack, S.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. [CrossRef]
- Mirabello, L.; Troisi, R.J.; Savage, S.A. International Osteosarcoma Incidence Patterns in Children and Adolescents, Middle Ages and Elderly Persons. Int. J. Cancer 2009, 125, 229–234. [CrossRef]
- Surveillance, Epidemiology, and End Results (SEER) Program, SEER*Stat Database: Incidence—SEER Research Data, 9 Registries, Nov 2019 Sub (1975–2017). Available: https://seer.cancer.gov/ (accessed on 24 January 2023).
- Tsukamoto, S.; Errani, C.; Angelini, A.; Mavrogenis, A.F. Current Treatment Considerations for Osteosarcoma Metastatic at Presentation. Orthopedics 2020, 43. [CrossRef]
- Albuquerque M. Silva1, J.; Marchiori2, E.; Carvalho de Macedo1, F.; Ricardo Garcia da Silva1, P.; Brandão Amorim1, 3, V. Pulmonary Metastasis of Osteosarcoma: Multiple Presentations in a Single Patient. J. Bras. Pneumol. 2022, e20210478. [CrossRef]
- American Cancer Society. Survival Rates for Osteosarcoma. Available: https://www.cancer.org/cancer/osteosarcoma/detection-diagnosis-staging/survival-rates.html (accessed on 28 January 2023).
- 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. [CrossRef]
- Evdokimova, V.; Gassmann, H.; Radvanyi, L.; Burdach, S.E.G. Current State of Immunotherapy and Mechanisms of Immune Evasion in Ewing Sarcoma and Osteosarcoma. Cancers (Basel). 2022, 15, 272. [CrossRef]
- Li, E.; Zhong, S.; Ma, G.; Wang, Q.; Gao, Y. MIR503HG Overexpression Inhibits the Malignant Behaviors of Osteosarcoma Cells by Sponging MiR-103a-3p. Crit. Rev. Eukaryot. Gene Expr. 2022. [CrossRef]
- Clapp, B.; Portela, R.; Sharma, I.; Nakanishi, H.; Marrero, K.; Schauer, P.; Halfdanarson, T.R.; Abu Dayyeh, B.; Kendrick, M.; Ghanem, O.M. Risk of Non-Hormonal Cancer after Bariatric Surgery: Meta-Analysis of Retrospective Observational Studies. Br. J. Surg. 2022, 110, 24–33. [CrossRef]
- Yeo, S.Y.; Bratke, G.; Grüll, H. High Intensity Focused Ultrasound for Treatment of Bone Malignancies—20 Years of History. Cancers (Basel). 2022, 15, 108. [CrossRef]
- Smeland, S.; Bielack, S.S.; Whelan, J.; Bernstein, M.; Hogendoorn, P.; Krailo, M.D.; Gorlick, R.; Janeway, K.A.; Ingleby, F.C.; Anninga, J.; et al. Survival and Prognosis with Osteosarcoma: Outcomes in More than 2000 Patients in the EURAMOS-1 (European and American Osteosarcoma Study) Cohort. Eur. J. Cancer 2019, 109, 36–50. [CrossRef]
- Singh, R.; Valluri, A.; Didwania, P.; Lehrer, E.J.; Baliga, S.; Hiniker, S.; Braunstein, S.E.; Murphy, E.S.; Lazarev, S.; Tinkle, C.; et al. Efficacy and Safety of Stereotactic Body Radiation Therapy (SBRT) for Pediatric Malignancies: The LITE-SABR Systematic Review and Meta-Analysis. Adv. Radiat. Oncol. 2023, 101123. [CrossRef]
- Ahmed, G.; Zamzam, M.; Kamel, A.; Ahmed, S.; Salama, A.; Zaki, I.; Kamal, N.; Elshafiey, M. Effect of Timing of Pulmonary Metastasis Occurrence on the Outcome of Metastasectomy in Osteosarcoma Patients. J. Pediatr. Surg. 2019, 54, 775–779. [CrossRef]
- Khanna, C.; Fan, T.M.; Gorlick, R.; Helman, L.J.; Kleinerman, E.S.; Adamson, P.C.; Houghton, P.J.; Tap, W.D.; Welch, D.R.; Steeg, P.S.; et al. Toward a Drug Development Path That Targets Metastatic Progression in Osteosarcoma. Clin. Cancer Res. 2014, 20, 4200–4209. [CrossRef]
- Ritter, J.; Bielack, S.S. Osteosarcoma. Ann. Oncol. 2010, 21, vii320–vii325. [CrossRef]
- Guo, S.; Zhu, X.; Huang, Z.; Wei, C.; Yu, J.; Zhang, L.; Feng, J.; Li, M.; Li, Z. Genomic Instability Drives Tumorigenesis and Metastasis and Its Implications for Cancer Therapy. Biomed. Pharmacother. 2023, 157, 114036. [CrossRef]
- López-Otín, C.; Pietrocola, F.; Roiz-Valle, D.; Galluzzi, L.; Kroemer, G. Meta-Hallmarks of Aging and Cancer. Cell Metab. 2023, 35, 12–35. [CrossRef]
- Ni, M. [Update and Interpretation of 2021 National Comprehensive Cancer Network (NCCN) “Clinical Practice Guidelines for Bone Tumors”]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2021, 35, 1186–1191. [CrossRef]
- Tan, G.; Xu, J.; Yu, Q.; Yang, Z.; Zhang, H. The Safety and Efficiency of Photodynamic Therapy for the Treatment of Osteosarcoma: A Systematic Review of in Vitro Experiment and Animal Model Reports. Photodiagnosis Photodyn. Ther. 2022, 40, 103093. [CrossRef]
- Lim, Y.Y.; Miskon, A.; Zaidi, A.M.A.; Megat Ahmad, M.M.H.; Abu Bakar, M. Structural Characterization Analyses of Low Brass Filler Biomaterial for Hard Tissue Implanted Scaffold Applications. Materials (Basel). 2022, 15, 1421. [CrossRef]
- Lim, Y.Y.; Miskon, A.; Zaidi, A.M.A. Structural Strength Analyses for Low Brass Filler Biomaterial with Anti-Trauma Effects in Articular Cartilage Scaffold Design. Materials (Basel). 2022, 15, 4446. [CrossRef]
- Fierro Pineda, J.C.; Wedekind, M.F.; Glod, J.W. Immunotherapy Approaches for Rare Pediatric Solid Tumors: Advances and Future Directions. Curr. Opin. Pediatr. 2023, 35, 63–74. [CrossRef]
- Lim, Y.Y.; Miskon, A.; Zaidi, A.M.A.; Megat Ahmad, M.M.H.; Abu Bakar, M. Numerical Simulation Study on Relationship between the Fracture Mechanisms and Residual Membrane Stresses of Metallic Material. J. Funct. Biomater. 2022, 13, 20. [CrossRef]
- Lim, Y.Y.; Miskon, A.; Zaidi, A.M.A. CuZn Complex Used in Electrical Biosensors for Drug Delivery Systems. Materials (Basel). 2022, 15, 7672. [CrossRef]
- Ke, C.-H.; Chiu, Y.-H.; Huang, K.-C.; Lin, C.-S. Exposure of Immunogenic Tumor Antigens in Surrendered Immunity and the Significance of Autologous Tumor Cell-Based Vaccination in Precision Medicine. Int. J. Mol. Sci. 2022, 24, 147. [CrossRef]
- Fazaeli, H.; Sheikholeslami, A.; Ghasemian, F.; Amini, E.; Sheykhhasan, M. The Emerging Role of LncRNA FENDRR in Multiple Cancer: A Review Study. Curr. Mol. Med. 2022, 22. [CrossRef]
- Shahpouri, M.; Adili-Aghdam, M.A.; Mahmudi, H.; Jaymand, M.; Amoozgar, Z.; Akbari, M.; Hamblin, M.R.; Jahanban-Esfahlan, R. Prospects for Hypoxia-Based Drug Delivery Platforms for the Elimination of Advanced Metastatic Tumors: From 3D Modeling to Clinical Concepts. J. Control. Release 2023, 353, 1002–1022. [CrossRef]
- Zheng, Y.; Chang, X.; Huang, Y.; He, D. The Application of Antidepressant Drugs in Cancer Treatment. Biomed. Pharmacother. 2023, 157, 113985. [CrossRef]
- Khadembaschi, D.; Jafri, M.; Praveen, P.; Parmar, S.; Breik, O. Does Neoadjuvant Chemotherapy Provide a Survival Benefit in Maxillofacial Osteosarcoma: A Systematic Review and Pooled Analysis. Oral Oncol. 2022, 135, 106133. [CrossRef]
- Lim, Y.Y.; Zaidi, A.M.A.; Miskon, A. Composing On-Program Triggers and On-Demand Stimuli into Biosensor Drug Carriers in Drug Delivery Systems for Programmable Arthritis Therapy. Pharmaceuticals 2022, 15, 1330. [CrossRef]
- Evans, H.M.; Long, J.A. Characteristic Effects upon Growth, Oestrus and Ovulation Induced by the Intraperitoneal Administration of Fresh Anterior Hypophyseal Substance. Proc. Natl. Acad. Sci. 1922, 8, 38–39. [CrossRef]
- Li, C.H.; Liu, W.-K. Human Pituitary Growth Hormone. Experientia 1964, 20, 169–178. [CrossRef]
- Jaffe, N.; Frei, E.; Watts, H.; Traggis, D. High-Dose Methotrexate in Osteogenic Sarcoma: A 5-Year Experience. Cancer Treat. Rep. 1978, 62, 259–264.
- Rosen, G.; Caparros, B.; Huvos, A.G.; Kosloff, C.; Nirenberg, A.; Cacavio, A.; Marcove, R.C.; Lane, J.M.; Mehta, B.; Urban, C. Preoperative Chemotherapy for Osteogenic Sarcoma:Selection of Postoperative Adjuvant Chemotherapy Based on the Response of the Primary Tumor to Preoperative Chemotherapy. Cancer 1982, 49, 1221–1230. [CrossRef]
- Zapf, J.; Schmid, C.H.; Froesch, E.R. 1 Biological and Immunological Properties of Insulin-like Growth Factors (IGF) I and II. Clin. Endocrinol. Metab. 1984, 13, 3–30. [CrossRef]
- Stashenko, P.; Dewhirst, F.E.; Rooney, M.L.; Desjardins, L.A.; Heeley, J.D. Interleukin-1β Is a Potent Inhibitor of Bone Formation in Vitro. J. Bone Miner. Res. 2009, 2, 559–565. [CrossRef]
- Bengtsson, B.A.; Edén, S.; Lönn, L.; Kvist, H.; Stokland, A.; Lindstedt, G.; Bosaeus, I.; Tölli, J.; Sjöström, L.; Isaksson, O.G. Treatment of Adults with Growth Hormone (GH) Deficiency with Recombinant Human GH. J. Clin. Endocrinol. Metab. 1993, 76, 309–317. [CrossRef]
- Gentet, J.-C.; Brunat-Mentigny, M.; Demaille, M.C.; Pein, F.; Avet-Loiseau, H.; Berger, C.; De Lumley, L.; Pacquement, H.; Schmitt, C.; Sariban, E.; et al. Ifosfamide and Etoposide in Childhood Osteosarcoma. A Phase II Study of the French Society of Paediatric Oncology. Eur. J. Cancer 1997, 33, 232–237. [CrossRef]
- McGary, E.C.; Weber, K.; Mills, L.; Doucet, M.; Lewis, V.; Lev, D.C.; Fidler, I.J.; Bar-Eli, M. Inhibition of Platelet-Derived Growth Factor-Mediated Proliferation of Osteosarcoma Cells by the Novel Tyrosine Kinase Inhibitor STI571. Clin. Cancer Res. 2002, 8, 3584–3591.
- Nardin, A.; Lefebvre, M.; Labroquere, K.; Faure, O.; Abastado, J. Liposomal Muramyl Tripeptide Phosphatidylethanolamine: Targeting and Activating Macrophages for Adjuvant Treatment of Osteosarcoma. Curr. Cancer Drug Targets 2006, 6, 123–133. [CrossRef]
- Tang, Q.-L.; Zhao, Z.-Q.; Li, J.; Liang, Y.; Yin, J.-Q.; Zou, C.-Y.; Xie, X.-B.; Zeng, Y.-X.; Shen, J.-N.; Kang, T.; et al. Salinomycin Inhibits Osteosarcoma by Targeting Its Tumor Stem Cells. Cancer Lett. 2011, 311, 113–121. [CrossRef]
- Gordon, E.M.; Chua-Alcala, V.S.; Kim, K.; Baby, R.; Angel, N.; Quon, D.; Wong, S.; Chawla, S.P. A Phase I/II Investigation of Nivolumab and ABI-009 ( Nab -Sirolimus) in Advanced Undifferentiated Pleomorphic Sarcoma (UPS), Liposarcoma (LPS), Chondrosarcoma (CS), Osteosarcoma (OS), and Ewing Sarcoma: Preliminary Efficacy and Safety Results. J. Clin. Oncol. 2019, 37, 11057–11057. [CrossRef]
- Manji, A.; Samson, Y.; Deyell, R.J.; Johnston, D.L.; Lewis, V.A.; Zorzi, A.P.; Berman, J.N.; Brodeur-Robb, K.; Morrison, E.; Kee, L.; et al. Low-Dose Metronomic Topotecan and Pazopanib (TOPAZ) in Children with Relapsed or Refractory Solid Tumors: A C17 Canadian Phase I Clinical Trial. Cancers (Basel). 2022, 14, 2985. [CrossRef]
- Gartrell, J.; Panetta, J.C.; Baker, S.D.; Chen, Y.L.; Hawkins, D.S.; Ostrenga, A.; Scharschmidt, T.J.; Spunt, S.L.; Wang, D.; Weiss, A.R. The Effects of Pazopanib on Doxorubicin Pharmacokinetics in Children and Adults with Non-Rhabdomyosarcoma Soft Tissue Sarcoma: A Report from Children’s Oncology Group and NRG Oncology Study ARST1321. Cancer Chemother. Pharmacol. 2022, 89, 551–557. [CrossRef]
- Szkandera, J. Keeping up the ‘Race Pace’ in a Patient with Nonuterine Leiomyosarcoma. Futur. Oncol. 2022, 18, 12–16. [CrossRef]
- Liu, Z.; Wang, X.; Wang, J.; Zhang, P.; Li, C.; Wang, B.; Liu, G.; Yao, W. Gemcitabine Plus Anlotinib Is Effective and Safe Compared to Gemcitabine Plus Docetaxel in Advanced Soft Tissue Sarcoma. Front. Oncol. 2022, 12. [CrossRef]
- Villaruz, L.C.; Kelly, K.; Waqar, S.N.; Davis, E.J.; Shapiro, G.; LoRusso, P.; Dees, E.C.; Normolle, D.P.; Rhee, J.C.; Chu, E.; et al. NCI 9938: Phase I Clinical Trial of ATR Inhibitor Berzosertib (M6620, VX-970) in Combination with Irinotecan in Patients with Advanced Solid Tumors. J. Clin. Oncol. 2022, 40, 3012–3012. [CrossRef]
- Plummer, R.; Dean, E.; Arkenau, H.-T.; Redfern, C.; Spira, A.I.; Melear, J.M.; Chung, K.Y.; Ferrer-Playan, J.; Goddemeier, T.; Locatelli, G.; et al. A Phase 1b Study Evaluating the Safety and Preliminary Efficacy of Berzosertib in Combination with Gemcitabine in Patients with Advanced Non-Small Cell Lung Cancer. Lung Cancer 2022, 163, 19–26. [CrossRef]
- Brahmi, M.; Gautier, J.; Dufresne, A.; Marec-Berard, P.; Cropet, C.; Vizoso, S.; Bissuel, L.; Valentin, T.; Entz-Werle, N.; Bompas, E.; et al. REGOMAIN: A Randomized, Placebo-Controlled, Double-Blinded, Multicenter, Comparative Phase II Study of the Efficacy of Regorafenib as Maintenance Treatment in Patients (Pts) with High-Grade Bone Sarcomas (HGBS) at Diagnosis or Relapse and without Complete. J. Clin. Oncol. 2022, 40, TPS11585–TPS11585. [CrossRef]
- Vienot, A.; Vernerey, D.; Bouard, A.; Klajer, E.; Asgarov, K.; Kim, S.; Tournigand, C.; Louvet, C.; André, T.; Rousseau, B.; et al. SO-20 Stanniocalcin 1 (STC1) in Patients with Refractory Colorectal Cancer (CRC) Treated with Regorafenib: An Exploratory Analysis of the CORRECT Trial. Ann. Oncol. 2022, 33, S365. [CrossRef]
- Lee, S.; Cavaliere, A.; Gallezot, J.-D.; Keler, T.; Michelhaugh, S.K.; Belitzky, E.; Liu, M.; Mulnix, T.; Maher, S.E.; Bothwell, A.L.M.; et al. [89Zr]ZrDFO-CR011 PET Correlates with Response to Glycoprotein Nonmetastatic Melanoma B–Targeted Therapy in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2022, 21, 440–447. [CrossRef]
- Lazaratos, A.-M.; Annis, M.G.; Siegel, P.M. GPNMB: A Potent Inducer of Immunosuppression in Cancer. Oncogene 2022, 41, 4573–4590. [CrossRef]
- Albarrán, V.; Villamayor, M.L.; Chamorro, J.; Rosero, D.I.; Pozas, J.; San Román, M.; Calvo, J.C.; Pérez de Aguado, P.; Moreno, J.; Guerrero, P.; et al. Receptor Tyrosine Kinase Inhibitors for the Treatment of Recurrent and Unresectable Bone Sarcomas. Int. J. Mol. Sci. 2022, 23, 13784. [CrossRef]
- Pearson, A.D.; Gaspar, N.; Janeway, K.; Campbell-Hewson, Q.; Lawlor, E.R.; Copland, C.; Karres, D.; Norga, K.; Benzaghou, F.; Weiner, S.; et al. Paediatric Strategy Forum for Medicinal Product Development of Multi-Targeted Kinase Inhibitors in Bone Sarcomas. Eur. J. Cancer 2022, 173, 71–90. [CrossRef]
- Teo, M.Y.M.; Fong, J.Y.; Lim, W.M.; In, L.L.A. Current Advances and Trends in KRAS Targeted Therapies for Colorectal Cancer. Mol. Cancer Res. 2022, 20, 30–44. [CrossRef]
- Al Shihabi, A.; Davarifar, A.; Nguyen, H.T.L.; Tavanaie, N.; Nelson, S.D.; Yanagawa, J.; Federman, N.; Bernthal, N.; Hornicek, F.; Soragni, A. Personalized Chordoma Organoids for Drug Discovery Studies. Sci. Adv. 2022, 8. [CrossRef]
- Cash, T.; Jonus, H.C.; Tsvetkova, M.; Beumer, J.H.; Lee, J.Y.; Henry, C.; Aguilera, D.; Harvey, R.D.; Goldsmith, K.C. A Phase I Study of Simvastatin in Combination with Topotecan and Cyclophosphamide in Pediatric Patients with Relapsed and/or Refractory Solid and CNS Tumors. J. Clin. Oncol. 2020, 38, 10541–10541. [CrossRef]
- Duarte, J.A.; de Barros, A.L.B.; Leite, E.A. The Potential Use of Simvastatin for Cancer Treatment: A Review. Biomed. Pharmacother. 2021, 141, 111858. [CrossRef]
- Beird, H.C.; Bielack, S.S.; Flanagan, A.M.; Gill, J.; Heymann, D.; Janeway, K.A.; Livingston, J.A.; Roberts, R.D.; Strauss, S.J.; Gorlick, R. Osteosarcoma. Nat. Rev. Dis. Prim. 2022, 8, 77. [CrossRef]
- Pascual-Pasto, G.; Castillo-Ecija, H.; Unceta, N.; Aschero, R.; Resa-Pares, C.; Gómez-Caballero, A.; Vila-Ubach, M.; Muñoz-Aznar, O.; Suñol, M.; Burgueño, V.; et al. SPARC-Mediated Long-Term Retention of Nab-Paclitaxel in Pediatric Sarcomas. J. Control. Release 2022, 342, 81–92. [CrossRef]
- Digklia, A.; Kollar, A.; Kronig, M.-N.; Britschgi, C.; Rordorf, T.; Joerger, M.; Krasniqi, F.; Metaxas, Y.; Colombo, I.; Dietrich, D.; et al. 1495P SAKK 57/16 Nab-Paclitaxel and Gemcitabine in Soft Tissue Sarcoma (NAPAGE): Final Results from the Phase Ib/II Trial with >2y Median Follow Up. Ann. Oncol. 2022, 33, S1230. [CrossRef]
- Omar, S.; Albritton, K.; Heym, K.; Wang, J.; Ray, A. Multimodal Treatment of Sarcomas Linked to BCOR-CCNB3 Fusion in Pediatrics: A 3-Patient Case Series. Clin. Pediatr. Hematol. 2022, 29, 60–64. [CrossRef]
- Smida, M.; Ammar, A.; Fedhila, F.; Douira, W.; Sassi, S. Periosteal Preservation: A New Technique in Resection of Bone High-Grade Malignant Tumors in Children—about Eleven Cases. World J. Surg. Oncol. 2022, 20, 312. [CrossRef]
- Puppo, M.; Jaafar, M.; Diaz, J.-J.; Marcel, V.; Clézardin, P. MiRNAs and SnoRNAs in Bone Metastasis: Functional Roles and Clinical Potential. Cancers (Basel). 2022, 15, 242. [CrossRef]
- Sadeh, M.; Toledano, H.; Cohen, I.J. A Comprehensive Review of Neuropsychologic Studies Supports the Concept That Adequate Folinic Acid Rescue Prevents Post Methotrexate Neurotoxicity. J. Pediatr. Hematol. Oncol. 2023, 45, 1–11. [CrossRef]
- Li, S.; Wang, X. The Potential Roles of Exosomal Noncoding RNAs in Osteosarcoma. J. Cell. Physiol. 2021, 236, 3354–3365. [CrossRef]
- Abhange, K.; Makler, A.; Wen, Y.; Ramnauth, N.; Mao, W.; Asghar, W.; Wan, Y. Small Extracellular Vesicles in Cancer. Bioact. Mater. 2021, 6, 3705–3743. [CrossRef]
- Kluszczynska, K.; Czyz, M. Extracellular Vesicles-Based Cell-Cell Communication in Melanoma: New Perspectives in Diagnostics and Therapy. Int. J. Mol. Sci. 2023, 24, 965. [CrossRef]
- Song, H.; Zhao, J.; Cheng, J.; Feng, Z.; Wang, J.; Momtazi-Borojeni, A.A.; Liang, Y. Extracellular Vesicles in Chondrogenesis and Cartilage Regeneration. J. Cell. Mol. Med. 2021, 25, 4883–4892. [CrossRef]
- Jothimani, G.; Pathak, S.; Dutta, S.; Duttaroy, A.K.; Banerjee, A. A Comprehensive Cancer-Associated MicroRNA Expression Profiling and Proteomic Analysis of Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes. Tissue Eng. Regen. Med. 2022, 19, 1013–1031. [CrossRef]
- Kwok, Z.H.; Wang, C.; Jin, Y. Extracellular Vesicle Transportation and Uptake by Recipient Cells: A Critical Process to Regulate Human Diseases. Processes 2021, 9, 273. [CrossRef]
- Ross, J.; McIver, Z.; Lambert, T.; Piergentili, C.; Bird, J.E.; Gallagher, K.J.; Cruickshank, F.L.; James, P.; Zarazúa-Arvizu, E.; Horsfall, L.E.; et al. Pore Dynamics and Asymmetric Cargo Loading in an Encapsulin Nanocompartment. Sci. Adv. 2022, 8. [CrossRef]
- van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and Directions in Studying Cell–Cell Communication by Extracellular Vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [CrossRef]
- Xiang, Y.; Hu, C.; Wu, G.; Xu, S.; Li, Y. Nanomaterial-Based Microfluidic Systems for Cancer Biomarker Detection: Recent Applications and Future Perspectives. TrAC Trends Anal. Chem. 2023, 158, 116835. [CrossRef]
- Liang, Y.; Fang, D.; Gao, X.; Deng, X.; Chen, N.; Wu, J.; Zeng, M.; Luo, M. Circulating MicroRNAs as Emerging Regulators of COVID-19. Theranostics 2023, 13, 125–147. [CrossRef]
- Laviron, M.; Boissonnas, A. Ontogeny of Tumor-Associated Macrophages. Front. Immunol. 2019, 10. [CrossRef]
- Wei, Z.; Zheng, D.; Pi, W.; Qiu, Y.; Xia, K.; Guo, W. Isoquercitrin Restrains the Proliferation and Promotes Apoptosis of Human Osteosarcoma Cells by Inhibiting the Wnt/β-Catenin Pathway. J. Bone Oncol. 2023, 100468. [CrossRef]
- Lu, D.; Liu, R.; Zhou, Y.; Zhang, Z.; Jiang, X.; Xu, J.; Su, A.; Hu, Z. FOXO3a-Dependent up-Regulation of HSP90 Alleviates Cisplatin-Induced Apoptosis by Activating FUNDC1-Mediated Mitophagy in Hypoxic Osteosarcoma Cells. Cell. Signal. 2023, 101, 110500. [CrossRef]
- Raimondi, L.; De Luca, A.; Gallo, A.; Costa, V.; Russelli, G.; Cuscino, N.; Manno, M.; Raccosta, S.; Carina, V.; Bellavia, D.; et al. Osteosarcoma Cell-Derived Exosomes Affect Tumor Microenvironment by Specific Packaging of MicroRNAs. Carcinogenesis 2020, 41, 666–677. [CrossRef]
- DeNardo, D.G.; Ruffell, B. Macrophages as Regulators of Tumour Immunity and Immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [CrossRef]
- Wolf-Dennen, K.; Gordon, N.; Kleinerman, E.S. Exosomal Communication by Metastatic Osteosarcoma Cells Modulates Alveolar Macrophages to an M2 Tumor-Promoting Phenotype and Inhibits Tumoricidal Functions. Oncoimmunology 2020, 9. [CrossRef]
- Lopes, N.; Vivier, E.; Narni-Mancinelli, E. Natural Killer Cells and Type 1 Innate Lymphoid Cells in Cancer. Semin. Immunol. 2023, 66, 101709. [CrossRef]
- Wu, C.-C.; Beird, H.C.; Andrew Livingston, J.; Advani, S.; Mitra, A.; Cao, S.; Reuben, A.; Ingram, D.; Wang, W.-L.; Ju, Z.; et al. Immuno-Genomic Landscape of Osteosarcoma. Nat. Commun. 2020, 11, 1008. [CrossRef]
- Verma, A.; Mathur, R.; Farooque, A.; Kaul, V.; Gupta, S.; Dwarakanath, B.S. T-Regulatory Cells In Tumor Progression And Therapy. Cancer Manag. Res. 2019, Volume 11, 10731–10747. [CrossRef]
- Ray, S.K.; Mukherjee, S. Altering Landscape of Cancer Vaccines: Unique Platforms, Research on Therapeutic Applications and Recent Patents. Recent Pat. Anticancer. Drug Discov. 2023, 18, 133–146. [CrossRef]
- Bayatipoor, H.; Mehdizadeh, S.; Jafarpour, R.; Shojaei, Z.; Pashangzadeh, S.; Motallebnezhad, M. Role of NKT Cells in Cancer Immunotherapy—from Bench to Bed. Med. Oncol. 2022, 40, 29. [CrossRef]
- Metelli, A.; Wu, B.X.; Riesenberg, B.; Guglietta, S.; Huck, J.D.; Mills, C.; Li, A.; Rachidi, S.; Krieg, C.; Rubinstein, M.P.; et al. Thrombin Contributes to Cancer Immune Evasion via Proteolysis of Platelet-Bound GARP to Activate LTGF-β. Sci. Transl. Med. 2020, 12. [CrossRef]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ Drives Immune Evasion in Genetically Reconstituted Colon Cancer Metastasis. Nature 2018, 554, 538–543. [CrossRef]
- Shima, T.; Shimoda, M.; Shigenobu, T.; Ohtsuka, T.; Nishimura, T.; Emoto, K.; Hayashi, Y.; Iwasaki, T.; Abe, T.; Asamura, H.; et al. Infiltration of Tumor-associated Macrophages Is Involved in Tumor Programmed Death-ligand 1 Expression in Early Lung Adenocarcinoma. Cancer Sci. 2020, 111, 727–738. [CrossRef]
- Wang, X.; Chen, Z.; Li, B.; Fan, J.; Xu, W.; Xiao, J. Immunotherapy as a Promising Option for the Treatment of Advanced Chordoma: A Systemic Review. Cancers (Basel). 2022, 15, 264. [CrossRef]
- Huang, T.; Tan, X.; Huang, H.; et, al Targeting Cancer-Associated Fibroblast-Secreted WNT2 Restores Dendritic Cell-Mediated Antitumour Immunity. Gut 2022, 71, 333–344.
- Yue, P.; Han, B.; Zhao, Y. Focus on the Molecular Mechanisms of Cisplatin Resistance Based on Multi-Omics Approaches. Mol. Omi. 2023. [CrossRef]
- Fan, L.; Qu, Y.; Tong, W.; Lin, H.; Xiao, B. Application Value of a Selenium-Hydroxyapatite Nanodelivery System as Osteosarcoma Treatment. Mater. Express 2022, 12, 1033–1041. [CrossRef]
- Garcia-Ortega, D.Y.; Cabrera-Nieto, S.A.; Caro-Sánchez, H.S.; Cruz-Ramos, M. An Overview of Resistance to Chemotherapy in Osteosarcoma and Future Perspectives. Cancer Drug Resist. 2022, 5, 762–793. [CrossRef]
- Inui, T.; Nomoto, R.; Yokota, J.; Yamashita, T.; Okada, K.; Kishimoto, W.; Nakase, H.; Mizuguchi, H. Establishment of MDR1-Knockout Human Enteroids for Pharmaceutical Application. Drug Metab. Pharmacokinet. 2023, 48, 100476. [CrossRef]
- Cai, J.-X.; Liu, J.-H.; Wu, J.-Y.; Li, Y.-J.; Qiu, X.-H.; Xu, W.-J.; Xu, P.; Xiang, D.-X. Hybrid Cell Membrane-Functionalized Biomimetic Nanoparticles for Targeted Therapy of Osteosarcoma. Int. J. Nanomedicine 2022, Volume 17, 837–854. [CrossRef]
- Zhang, X.-B.; Zhang, R.-H.; Su, X.; Qi, J.; Hu, Y.-C.; Shi, J.-T.; Zhang, K.; Wang, K.-P.; Zhou, H.-Y. Exosomes in Osteosarcoma Research and Preclinical Practice. Am. J. Transl. Res. 2021, 13, 882–897.
- Weinman, M.A.; Ramsey, S.A.; Leeper, H.J.; Brady, J. V.; Schlueter, A.; Stanisheuski, S.; Maier, C.S.; Miller, T.; Ruby, C.E.; Bracha, S. Exosomal Proteomic Signatures Correlate with Drug Resistance and Carboplatin Treatment Outcome in a Spontaneous Model of Canine Osteosarcoma. Cancer Cell Int. 2021, 21, 245. [CrossRef]
- Homayoonfal, M.; Asemi, Z.; Yousefi, B. Potential Anticancer Properties and Mechanisms of Thymoquinone in Osteosarcoma and Bone Metastasis. Cell. Mol. Biol. Lett. 2022, 27, 21. [CrossRef]
- Cheng, B.; Yu, Q.; Wang, W. Intimate Communications within the Tumor Microenvironment: Stromal Factors Function as an Orchestra. J. Biomed. Sci. 2023, 30, 1. [CrossRef]
- Chaudhary, A.; Raza, S.S.; Haque, R. Transcriptional Factors Targeting in Cancer Stem Cells for Tumor Modulation. Semin. Cancer Biol. 2023, 88, 123–137. [CrossRef]
- Wu, T.; Yang, W.; Sun, A.; Wei, Z.; Lin, Q. The Role of CXC Chemokines in Cancer Progression. Cancers (Basel). 2022, 15, 167. [CrossRef]
- Wang, Y.; Chu, Y.; Li, K.; Zhang, G.; Guo, Z.; Wu, X.; Qiu, C.; Li, Y.; Wan, X.; Sui, J.; et al. Exosomes Secreted by Adipose-Derived Mesenchymal Stem Cells Foster Metastasis and Osteosarcoma Proliferation by Increasing COLGALT2 Expression. Front. Cell Dev. Biol. 2020, 8. [CrossRef]
- Kehayova, Y.S.; Wilkinson, J.M.; Rice, S.J.; Loughlin, J. Independent Osteoarthritis Risk-conferring Alleles Mediate the Same Epigenetic and Transcriptional Effect on a Shared Target Gene, COLGALT2. Arthritis Rheumatol. 2022. [CrossRef]
- Zhang, H.; Wang, J.; Ren, T.; Huang, Y.; Liang, X.; Yu, Y.; Wang, W.; Niu, J.; Guo, W. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal MiR-206 Inhibits Osteosarcoma Progression by Targeting TRA2B. Cancer Lett. 2020, 490, 54–65. [CrossRef]
- Tian, B.; Du, X.; Zheng, S.; Zhang, Y. The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. Int. J. Mol. Sci. 2022, 23, 16155. [CrossRef]
- Han, F.; Pu, P.; Wang, C.; Ding, X.; Zhu, Z.; Xiang, W.; Wang, W. Osteosarcoma Cell-Derived Exosomal MiR-1307 Promotes Tumorgenesis via Targeting AGAP1. Biomed Res. Int. 2021, 2021, 1–17. [CrossRef]
- Chen, P.-W.; Gasilina, A.; Yadav, M.P.; Randazzo, P.A. Control of Cell Signaling by Arf GTPases and Their Regulators: Focus on Links to Cancer and Other GTPase Families. Biochim. Biophys. Acta - Mol. Cell Res. 2022, 1869, 119171. [CrossRef]
- Vail, D.J.; Somoza, R.A.; Caplan, A.I. MicroRNA Regulation of Bone Marrow Mesenchymal Stem Cell Chondrogenesis: Toward Articular Cartilage. Tissue Eng. Part A 2022, 28, 254–269. [CrossRef]
- Zhang, H.; Yu, Y.; Wang, J.; Han, Y.; Ren, T.; Huang, Y.; Chen, C.; Huang, Q.; Wang, W.; Niu, J.; et al. Macrophages-Derived Exosomal LncRNA LIFR-AS1 Promotes Osteosarcoma Cell Progression via MiR-29a/NFIA Axis. Cancer Cell Int. 2021, 21, 192. [CrossRef]
- Bai, Z.; Wang, X.; Zhang, Z. Long Noncoding RNA LIFR-AS1: A New Player in Human Cancers. Biomed Res. Int. 2022, 2022, 1–11. [CrossRef]
- Alfieri, M.; Meo, L.; Ragno, P. Posttranscriptional Regulation of the Plasminogen Activation System by Non-Coding RNA in Cancer. Int. J. Mol. Sci. 2023, 24, 962. [CrossRef]
- Luo, X.; Li, Y.; Hua, Z.; Xue, X.; Wang, X.; Pang, M.; Xiao, C.; Zhao, H.; Lyu, A.; Liu, Y. Exosomes-Mediated Tumor Metastasis through Reshaping Tumor Microenvironment and Distant Niche. J. Control. Release 2023, 353, 327–336. [CrossRef]
- Zhang, W.; Yan, Y.; Peng, J.; Thakur, A.; Bai, N.; Yang, K.; Xu, Z. Decoding Roles of Exosomal LncRNAs in Tumor-Immune Regulation and Therapeutic Potential. Cancers (Basel). 2022, 15, 286. [CrossRef]
- Li, Q.; Wang, X.; Jiang, N.; Xie, X.; Liu, N.; Liu, J.; Shen, J.; Peng, T. Exosome-transmitted Linc00852 Associated with Receptor Tyrosine Kinase AXL Dysregulates the Proliferation and Invasion of Osteosarcoma. Cancer Med. 2020, 9, 6354–6366. [CrossRef]
- Zhang, H.; Du, Y.; Xin, P.; Man, X. The LINC00852/MiR-29a-3p/JARID2 Axis Regulates the Proliferation and Invasion of Prostate Cancer Cell. BMC Cancer 2022, 22, 1269. [CrossRef]
- Ge, X.; Liu, W.; Zhao, W.; Feng, S.; Duan, A.; Ji, C.; Shen, K.; Liu, W.; Zhou, J.; Jiang, D.; et al. Exosomal Transfer of LCP1 Promotes Osteosarcoma Cell Tumorigenesis and Metastasis by Activating the JAK2/STAT3 Signaling Pathway. Mol. Ther. - Nucleic Acids 2020, 21, 900–915. [CrossRef]
- Zhang, H.; Song, J.; Zhou, X. Long Noncoding RNA P53 Upregulated Regulator of P53 Levels Promotes Osteogenic Differentiation in Osteoporosis Progression Through Sponging MiR-135a-5p. J. Biomater. Tissue Eng. 2022, 12, 2085–2091. [CrossRef]
- Zhang, K.; Dong, C.; Chen, M.; Yang, T.; Wang, X.; Gao, Y.; Wang, L.; Wen, Y.; Chen, G.; Wang, X.; et al. Extracellular Vesicle-Mediated Delivery of MiR-101 Inhibits Lung Metastasis in Osteosarcoma. Theranostics 2020, 10, 411–425. [CrossRef]
- Mei, X.; Zhang, B.; Zhao, M.; Lu, Q. An Update on Epigenetic Regulation in Autoimmune Diseases. J. Transl. Autoimmun. 2022, 5, 100176. [CrossRef]
- Mazumdar, A.; Urdinez, J.; Boro, A.; Migliavacca, J.; Arlt, M.J.E.; Muff, R.; Fuchs, B.; Snedeker, J.G.; Gvozdenovic, A. Osteosarcoma-Derived Extracellular Vesicles Induce Lung Fibroblast Reprogramming. Int. J. Mol. Sci. 2020, 21, 5451. [CrossRef]
- Tripathi, R.; Sinha, N.R.; Kempuraj, D.; Balne, P.K.; Landreneau, J.R.; Juneja, A.; Webel, A.D.; Mohan, R.R. Evaluation of CRISPR/Cas9 Mediated TGIF Gene Editing to Inhibit Corneal Fibrosis in Vitro. Exp. Eye Res. 2022, 220, 109113. [CrossRef]
- Cheng, Z.; Wang, L.; Wu, C.; Huang, L.; Ruan, Y.; Xue, W. Tumor-Derived Exosomes Induced M2 Macrophage Polarization and Promoted the Metastasis of Osteosarcoma Cells Through Tim-3. Arch. Med. Res. 2021, 52, 200–210. [CrossRef]
- Gomes de Morais, A.L.; Cerdá, S.; de Miguel, M. New Checkpoint Inhibitors on the Road: Targeting TIM-3 in Solid Tumors. Curr. Oncol. Rep. 2022, 24, 651–658. [CrossRef]
- Zhang, Y.; Liu, Z.; Yang, X.; Lu, W.; Chen, Y.; Lin, Y.; Wang, J.; Lin, S.; Yun, J.-P. H3K27 Acetylation Activated-COL6A1 Promotes Osteosarcoma Lung Metastasis by Repressing STAT1 and Activating Pulmonary Cancer-Associated Fibroblasts. Theranostics 2021, 11, 1473–1492. [CrossRef]
- Wang, Y.; Han, Y.; Wang, L.; Zou, M.; Sun, Y.; Sun, H.; Guo, Q.; Peng, X. Mycoplasma Gallisepticum Escapes the Host Immune Response via Gga-MiR-365-3p/SOCS5/STATs Axis. Vet. Res. 2022, 53, 103. [CrossRef]
- Zhang, H.; Wang, J.; Ren, T.; Huang, Y.; Yu, Y.; Chen, C.; Huang, Q.; Guo, W. LncRNA CASC15 Is Upregulated in Osteosarcoma Plasma Exosomes and CASC15 Knockdown Inhibits Osteosarcoma Progression by Regulating MiR-338-3p/RAB14 Axis. Onco. Targets. Ther. 2020, Volume 13, 12055–12066. [CrossRef]
- Chen, C.; Liu, L. Silencing of LncRNA KLF3-AS1 Represses Cell Growth in Osteosarcoma via MiR-338-3p/MEF2C Axis. J. Clin. Lab. Anal. 2022, 36. [CrossRef]
- Qin, F.; Tang, H.; Zhang, Y.; Zhang, Z.; Huang, P.; Zhu, J. Bone Marrow-derived Mesenchymal Stem Cell-derived Exosomal MicroRNA-208a Promotes Osteosarcoma Cell Proliferation, Migration, and Invasion. J. Cell. Physiol. 2020, 235, 4734–4745. [CrossRef]
- Wang, Q.; Zhou, H.; Zhu, X.; Jiang, F.; Yu, Q.; Zhang, J.; Ji, Y. MiR-208 Inhibits Myocardial Tissues Apoptosis in Mice with Acute Myocardial Infarction by Targeting Inhibition of PDCD4. J. Biochem. Mol. Toxicol. 2022, 36. [CrossRef]
- Huang, Y.; Liu, W.; He, B.; Wang, L.; Zhang, F.; Shu, H.; Sun, L. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells Promote Osteosarcoma Development by Activating Oncogenic Autophagy. J. Bone Oncol. 2020, 21, 100280. [CrossRef]
- Yu, Z.; Tang, H.; Chen, S.; Xie, Y.; Shi, L.; Xia, S.; Jiang, M.; Li, J.; Chen, D. Exosomal LOC85009 Inhibits Docetaxel Resistance in Lung Adenocarcinoma through Regulating ATG5-Induced Autophagy. Drug Resist. Updat. 2022, 100915. [CrossRef]
- Zhong, L.; Liao, D.; Li, J.; Liu, W.; Wang, J.; Zeng, C.; Wang, X.; Cao, Z.; Zhang, R.; Li, M.; et al. Rab22a-NeoF1 Fusion Protein Promotes Osteosarcoma Lung Metastasis through Its Secretion into Exosomes. Signal Transduct. Target. Ther. 2021, 6, 59. [CrossRef]
- Zeng, C.; Zhong, L.; Liu, W.; Zhang, Y.; Yu, X.; Wang, X.; Zhang, R.; Kang, T.; Liao, D. Targeting the Lysosomal Degradation of Rab22a-NeoF1 Fusion Protein for Osteosarcoma Lung Metastasis. Adv. Sci. 2022, 2205483. [CrossRef]
- Ye, H.; Hu, X.; Wen, Y.; Tu, C.; Hornicek, F.; Duan, Z.; Min, L. Exosomes in the Tumor Microenvironment of Sarcoma: From Biological Functions to Clinical Applications. J. Nanobiotechnology 2022, 20, 403. [CrossRef]
- 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. [CrossRef]
- Liu, Z.; Zhou, Z.; Dang, Q.; Xu, H.; Lv, J.; Li, H.; Han, X. Immunosuppression in Tumor Immune Microenvironment and Its Optimization from CAR-T Cell Therapy. Theranostics 2022, 12, 6273–6290. [CrossRef]
- Wang, J.; Zhang, H.; Sun, X.; Wang, X.; Ren, T.; Huang, Y.; Zhang, R.; Zheng, B.; Guo, W. Exosomal PD-L1 and N-Cadherin Predict Pulmonary Metastasis Progression for Osteosarcoma Patients. J. Nanobiotechnology 2020, 18, 151. [CrossRef]
- Tomassetti, A. Editorial to the Special Issue “Activations of Cadherin Signaling in Cancer.” Int. J. Mol. Sci. 2022, 23, 7022. [CrossRef]
- Heim, L.; Yang, Z.; Tausche, P.; Hohenberger, K.; Chiriac, M.T.; Koelle, J.; Geppert, C.-I.; Kachler, K.; Miksch, S.; Graser, A.; et al. IL-9 Producing Tumor-Infiltrating Lymphocytes and Treg Subsets Drive Immune Escape of Tumor Cells in Non-Small Cell Lung Cancer. Front. Immunol. 2022, 13. [CrossRef]
- Feng, Y.; Yan, S.; Lam, S.K.; Ko, F.C.F.; Chen, C.; Khan, M.; Ho, J.C.-M. IL-9 Stimulates an Anti-Tumor Immune Response and Facilitates Immune Checkpoint Blockade in the CMT167 Mouse Model. Lung Cancer 2022, 174, 14–26. [CrossRef]
- Zhan, X.; He, Q.; Sheng, J.; Jiang, X.; Lin, L.; Huang, Y.; He, S.; Chen, Y.; Li, L.; Zeng, Z.; et al. USP12 Positively Regulates M-MDSC Function to Inhibit Antitumour Immunity through Deubiquitinating and Stabilizing P65. Immunology 2022, 167, 544–557. [CrossRef]
- Guo, J.; Zhao, J.; Sun, L.; Yang, C. Role of Ubiquitin Specific Proteases in the Immune Microenvironment of Prostate Cancer: A New Direction. Front. Oncol. 2022, 12. [CrossRef]
- Toney, N.J.; Opdenaker, L.M.; Cicek, K.; Frerichs, L.; Kennington, C.R.; Oberly, S.; Archinal, H.; Somasundaram, R.; Sims-Mourtada, J. Tumor-B-Cell Interactions Promote Isotype Switching to an Immunosuppressive IgG4 Antibody Response through Upregulation of IL-10 in Triple Negative Breast Cancers. J. Transl. Med. 2022, 20, 112. [CrossRef]
- Zhang, X.; Jin, X.; Guan, L.; Lin, X.; Li, X.; Li, Y. IgG4-Related Disease With Gastrointestinal Involvement: Case Reports and Literature Review. Front. Immunol. 2022, 13. [CrossRef]
- Cao, Y.; He, H.; Li, R.; Liu, X.; Chen, Y.; Qi, Y.; Yu, K.; Wang, J.; Lin, C.; Liu, H.; et al. Latency-Associated Peptide Identifies Immunoevasive Subtype Gastric Cancer With Poor Prognosis and Inferior Chemotherapeutic Responsiveness. Ann. Surg. 2022, 275, e163–e173. [CrossRef]
- Chen, S.-Y.; Mamai, O.; Akhurst, R.J. TGFβ: Signaling Blockade for Cancer Immunotherapy. Annu. Rev. Cancer Biol. 2022, 6, 123–146. [CrossRef]
- Mohapatra, P.; Chandrasekaran, N. Wnt/β-Catenin Targeting in Liver Carcinoma through Nanotechnology-Based Drug Repurposing: A Review. Biomed. Pharmacother. 2022, 155, 113713. [CrossRef]
- Duan, Z.; Lin, X.; Wang, L.; Zhen, Q.; Jiang, Y.; Chen, C.; Yang, J.; Lee, C.-H.; Qin, Y.; Li, Y.; et al. Specificity of TGF-Β1 Signal Designated by LRRC33 and Integrin AVβ8. Nat. Commun. 2022, 13, 4988. [CrossRef]
- Dodagatta-Marri, E.; Ma, H.-Y.; Liang, B.; Li, J.; Meyer, D.S.; Chen, S.-Y.; Sun, K.-H.; Ren, X.; Zivak, B.; Rosenblum, M.D.; et al. Integrin Avβ8 on T Cells Suppresses Anti-Tumor Immunity in Multiple Models and Is a Promising Target for Tumor Immunotherapy. Cell Rep. 2021, 36, 109309. [CrossRef]
- Guo, D.; Tong, Y.; Jiang, X.; Meng, Y.; Jiang, H.; Du, L.; Wu, Q.; Li, S.; Luo, S.; Li, M.; et al. Aerobic Glycolysis Promotes Tumor Immune Evasion by Hexokinase2-Mediated Phosphorylation of IκBα. Cell Metab. 2022, 34, 1312-1324.e6. [CrossRef]
- Zhang, Y.; Fu, J.; Liu, S.; Wang, L.; Qiu, J.; van Schaik, E.J.; Samuel, J.E.; Song, L.; Luo, Z.-Q. Coxiella Burnetii Inhibits Host Immunity by a Protein Phosphatase Adapted from Glycolysis. Proc. Natl. Acad. Sci. 2022, 119. [CrossRef]
- Du, L.; He, H.; Xiao, Z.; Xiao, H.; An, Y.; Zhong, H.; Lin, M.; Meng, X.; Han, S.; Shuai, X. GSH-Responsive Metal–Organic Framework for Intratumoral Release of NO and IDO Inhibitor to Enhance Antitumor Immunotherapy. Small 2022, 18, 2107732. [CrossRef]
- Fujiwara, Y.; Kato, S.; Nesline, M.K.; Conroy, J.M.; DePietro, P.; Pabla, S.; Kurzrock, R. Indoleamine 2,3-Dioxygenase (IDO) Inhibitors and Cancer Immunotherapy. Cancer Treat. Rev. 2022, 110, 102461. [CrossRef]
- Gulley, J.L.; Schlom, J.; Barcellos-Hoff, M.H.; Wang, X.; Seoane, J.; Audhuy, F.; Lan, Y.; Dussault, I.; Moustakas, A. Dual Inhibition of TGF-β and PD-L1: A Novel Approach to Cancer Treatment. Mol. Oncol. 2022, 16, 2117–2134. [CrossRef]
- Metropulos, A.E.; Munshi, H.G.; Principe, D.R. The Difficulty in Translating the Preclinical Success of Combined TGFβ and Immune Checkpoint Inhibition to Clinical Trial. eBioMedicine 2022, 86, 104380. [CrossRef]
- Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune Checkpoint Blockade Therapy for Cancer: An Overview of FDA-Approved Immune Checkpoint Inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [CrossRef]
- Martin, C.J.; Datta, A.; Littlefield, C.; Kalra, A.; Chapron, C.; Wawersik, S.; Dagbay, K.B.; Brueckner, C.T.; Nikiforov, A.; Danehy, F.T.; et al. Selective Inhibition of TGFβ1 Activation Overcomes Primary Resistance to Checkpoint Blockade Therapy by Altering Tumor Immune Landscape. Sci. Transl. Med. 2020, 12. [CrossRef]
- Chen, C.; Shi, Q.; Xu, J.; Ren, T.; Huang, Y.; Guo, W. Current Progress and Open Challenges for Applying Tyrosine Kinase Inhibitors in Osteosarcoma. Cell Death Discov. 2022, 8, 488. [CrossRef]
- Northcote-Smith, J.; Suntharalingam, K. Targeting Chemotherapy-Resistant Tumour Sub-Populations Using Inorganic Chemistry: Anti-Cancer Stem Cell Metal Complexes. Curr. Opin. Chem. Biol. 2023, 72, 102237. [CrossRef]
- Costa, A.R.; Duarte, A.C.; Costa-Brito, A.R.; Gonçalves, I.; Santos, C.R.A. Bitter Taste Signaling in Cancer. Life Sci. 2023, 121363. [CrossRef]
- Aprile, M.; Costa, V.; Cimmino, A.; Calin, G.A. Emerging Role of Oncogenic Long Noncoding RNA as Cancer Biomarkers. Int. J. Cancer 2023, 152, 822–834. [CrossRef]
- Zhang, Q.; Liu, N.; Wang, J.; Liu, Y.; Wang, K.; Zhang, J.; Pan, X. The Recent Advance of Cell-Penetrating and Tumor-Targeting Peptides as Drug Delivery Systems Based on Tumor Microenvironment. Mol. Pharm. 2023. [CrossRef]
- de Castro, K.C.; Coco, J.C.; dos Santos, É.M.; Ataide, J.A.; Martinez, R.M.; do Nascimento, M.H.M.; Prata, J.; da Fonte, P.R.M.L.; Severino, P.; Mazzola, P.G.; et al. Pluronic® Triblock Copolymer-Based Nanoformulations for Cancer Therapy: A 10-Year Overview. J. Control. Release 2023, 353, 802–822. [CrossRef]
- Wang, H.; Zhou, X.; Li, C.; Yan, S.; Feng, C.; He, J.; Li, Z.; Tu, C. The Emerging Role of Pyroptosis in Pediatric Cancers: From Mechanism to Therapy. J. Hematol. Oncol. 2022, 15, 140. [CrossRef]
- Marques da Costa, M.E.; Marchais, A.; Gomez-Brouchet, A.; Job, B.; Assoun, N.; Daudigeos-Dubus, E.; Fromigué, O.; Santos, C.; Geoerger, B.; Gaspar, N. In-Vitro and In-Vivo Establishment and Characterization of Bioluminescent Orthotopic Chemotherapy-Resistant Human Osteosarcoma Models in NSG Mice. Cancers (Basel). 2019, 11, 997. [CrossRef]
- Salem, I.M.; Mostafa, S.M.; Salama, I.; El-Sabbagh, O.I.; Hegazy, W.A.H.; Ibrahim, T.S. Design, Synthesis and Antitumor Evaluation of Novel Pyrazolo[3,4- d ]Pyrimidines Incorporating Different Amino Acid Conjugates as Potential DHFR Inhibitors. J. Enzyme Inhib. Med. Chem. 2023, 38, 203–215. [CrossRef]
- Yu, W.; Min, D.; Lin, F.; Zheng, S.; Tang, L.; He, A.; Hu, H.; Shen, Z. SKA1 Induces de Novo MTX-resistance in Osteosarcoma through Inhibiting FPGS Transcription. FEBS J. 2019, 286, 2399–2414. [CrossRef]
- Zhang, Y.; Zhang, C.; Man, X.; Men, Y.; Ren, X.; Li, X.; Han, L.; Sun, Z.; Yang, Y.; Hou, S.; et al. Functional Characterization of the SiFPGS2 Gene of Foxtail Millet in Folate Accumulation and Root Development. Plant Growth Regul. 2022. [CrossRef]
- Xiao, X.; Wang, W.; Li, Y.; Yang, D.; Li, X.; Shen, C.; Liu, Y.; Ke, X.; Guo, S.; Guo, Z. HSP90AA1-Mediated Autophagy Promotes Drug Resistance in Osteosarcoma. J. Exp. Clin. Cancer Res. 2018, 37, 201. [CrossRef]
- Chen, Y.; Zhang, K.; Li, Y.; Guo, R.; Zhang, K.; Zhong, G.; He, Q. Oestrogen-Related Receptor Alpha Mediates Chemotherapy Resistance of Osteosarcoma Cells via Regulation of ABCB1. J. Cell. Mol. Med. 2019, 23, 2115–2124. [CrossRef]
- Wang, G.; Cao, L.; Jiang, Y.; Zhang, T.; Wang, H.; Wang, Z.; Xu, J.; Mao, M.; Hua, Y.; Cai, Z.; et al. Anlotinib Reverses Multidrug Resistance (MDR) in Osteosarcoma by Inhibiting P-Glycoprotein (PGP1) Function In Vitro and In Vivo. Front. Pharmacol. 2022, 12. [CrossRef]
- He, Q.; Hao, P.; He, G.; Mai, H.; Liu, W.; Zhang, W.; Zhang, K.; Zhong, G.; Guo, R.; Yu, C.; et al. IGF2BP1-Regulated Expression of ERRα Is Involved in Metabolic Reprogramming of Chemotherapy Resistant Osteosarcoma Cells. J. Transl. Med. 2022, 20, 348. [CrossRef]
- Nwabo Kamdje, A.H.; Seke Etet, P.F.; Kipanyula, M.J.; Vecchio, L.; Tagne Simo, R.; Njamnshi, A.K.; Lukong, K.E.; Mimche, P.N. Insulin-like Growth Factor-1 Signaling in the Tumor Microenvironment: Carcinogenesis, Cancer Drug Resistance, and Therapeutic Potential. Front. Endocrinol. (Lausanne). 2022, 13. [CrossRef]
- Celik, B.; Cicek, K.; Leal, A.F.; Tomatsu, S. Regulation of Molecular Targets in Osteosarcoma Treatment. Int. J. Mol. Sci. 2022, 23, 12583. [CrossRef]
- Li, X.; Liu, Y.; Zhang, X.; Shen, J.; Xu, R.; Liu, Y.; Yu, X. Circular RNA Hsa_circ_0000073 Contributes to Osteosarcoma Cell Proliferation, Migration, Invasion and Methotrexate Resistance by Sponging MiR-145-5p and MiR-151-3p and Upregulating NRAS. Aging (Albany. NY). 2020, 12, 14157–14173. [CrossRef]
- Lilienthal, I.; Herold, N. Targeting Molecular Mechanisms Underlying Treatment Efficacy and Resistance in Osteosarcoma: A Review of Current and Future Strategies. Int. J. Mol. Sci. 2020, 21, 6885. [CrossRef]
- Wei, W.; Ji, L.; Duan, W.; Zhu, J. Circular RNA Circ_0081001 Knockdown Enhances Methotrexate Sensitivity in Osteosarcoma Cells by Regulating MiR-494-3p/TGM2 Axis. J. Orthop. Surg. Res. 2021, 16, 50. [CrossRef]
- Liu, S.; Duan, K.; Zhang, X.; Cao, X.; Wang, X.; Meng, F.; Liu, H.; Xu, B.; Wang, X. Circ_0081001 Down-Regulates MiR-494-3p to Enhance BACH1 Expression and Promotes Osteosarcoma Progression. Aging (Albany. NY). 2021, 13, 17274–17284. [CrossRef]
- Bai, Y.; Li, Y.; Bai, J.; Zhang, Y. Hsa_circ_0004674 Promotes Osteosarcoma Doxorubicin Resistance by Regulating the MiR-342-3p/FBN1 Axis. J. Orthop. Surg. Res. 2021, 16, 510. [CrossRef]
- Ma, X.-L.; Zhan, T.-C.; Hu, J.-P.; Zhang, C.-L.; Zhu, K.-P. Doxorubicin-Induced Novel CircRNA_0004674 Facilitates Osteosarcoma Progression and Chemoresistance by Upregulating MCL1 through MiR-142-5p. Cell Death Discov. 2021, 7, 309. [CrossRef]
- Guan, H.; Xu, H.; Chen, J.; Wu, W.; Chen, D.; Chen, Y.; Sun, J. Circ_0001721 Enhances Doxorubicin Resistance and Promotes Tumorigenesis in Osteosarcoma through MiR-758/TCF4 Axis. Cancer Cell Int. 2021, 21, 336. [CrossRef]
- Gao, Y.; Ma, H.; Gao, Y.; Tao, K.; Fu, L.; Ren, R.; Hu, X.; Kou, M.; Chen, B.; Shi, J.; et al. CircRNA Circ_0001721 Promotes the Progression of Osteosarcoma Through MiR-372-3p/MAPK7 Axis. Cancer Manag. Res. 2020, Volume 12, 8287–8302. [CrossRef]
- Wei, W.; Ji, L.; Duan, W.; Zhu, J. CircSAMD4A Contributes to Cell Doxorubicin Resistance in Osteosarcoma by Regulating the MiR-218-5p/KLF8 Axis. Open Life Sci. 2020, 15, 848–859. [CrossRef]
- Yanbin, Z.; Jing, Z. CircSAMD4A Accelerates Cell Proliferation of Osteosarcoma by Sponging MiR-1244 and Regulating MDM2 MRNA Expression. Biochem. Biophys. Res. Commun. 2019, 516, 102–111. [CrossRef]
- Ji, Y.; Liu, J.; Zhu, W.; Ji, J. Circ_0002060 Enhances Doxorubicin Resistance in Osteosarcoma by Regulating the MiR-198/ABCB1 Axis. Cancer Biother. Radiopharm. 2020, cbr.2020.4240. [CrossRef]
- Huang, Y.; Xie, J.; Li, E. Comprehensive Circular RNA Profiling Reveals Circ_0002060 as a Potential Diagnostic Biomarkers for Osteoporosis. J. Cell. Biochem. 2019, 120, 15688–15694. [CrossRef]
- Xie, C.; Liang, G.; Xu, Y.; Lin, E. Circular RNA Hsa_circ_0003496 Contributes to Tumorigenesis and Chemoresistance in Osteosarcoma Through Targeting (MicroRNA) MiR-370/Krüppel-Like Factor 12 Axis. Cancer Manag. Res. 2020, Volume 12, 8229–8240. [CrossRef]
- Lin, Z.; Xie, X.; Lu, S.; Liu, T. Noncoding RNAs in Osteosarcoma: Implications for Drug Resistance. Cancer Lett. 2021, 504, 91–103. [CrossRef]
- Zhang, Z.; Zhou, Q.; Luo, F.; Zhou, R.; Xu, J.; Xiao, J.; Dai, F.; Song, L. Circular RNA Circ-CHI3L1.2 Modulates Cisplatin Resistance of Osteosarcoma Cells via the MiR-340-5p/LPAATβ Axis. Hum. Cell 2021, 34, 1558–1568. [CrossRef]
- Song, L.; Zhou, Z.; Gan, Y.; Li, P.; Xu, Y.; Zhang, Z.; Luo, F.; Xu, J.; Zhou, Q.; Dai, F. Long Noncoding RNA OIP5-AS1 Causes Cisplatin Resistance in Osteosarcoma through Inducing the LPAATβ/PI3K/AKT/MTOR Signaling Pathway by Sponging the MiR-340-5p. J. Cell. Biochem. 2019, 120, 9656–9666. [CrossRef]
- Zhang, J.; Ma, X.; Zhou, R.; Zhou, Y. TRPS1 and YAP1 Regulate Cell Proliferation and Drug Resistance of Osteosarcoma via Competitively Binding to the Target of CircTADA2A – MiR-129-5p. Onco. Targets. Ther. 2020, Volume 13, 12397–12407. [CrossRef]
- Wu, Y.; Xie, Z.; Chen, J.; Chen, J.; Ni, W.; Ma, Y.; Huang, K.; Wang, G.; Wang, J.; Ma, J.; et al. Circular RNA CircTADA2A Promotes Osteosarcoma Progression and Metastasis by Sponging MiR-203a-3p and Regulating CREB3 Expression. Mol. Cancer 2019, 18, 73. [CrossRef]
- Pan, Y.; Lin, Y.; Mi, C. Cisplatin-resistant Osteosarcoma Cell-derived Exosomes Confer Cisplatin Resistance to Recipient Cells in an Exosomal Circ_103801-dependent Manner. Cell Biol. Int. 2021, 45, 858–868. [CrossRef]
- Soghli, N.; Qujeq, D.; Yousefi, T.; Soghli, N. The Regulatory Functions of Circular RNAs in Osteosarcoma. Genomics 2020, 112, 2845–2856. [CrossRef]
- Eaton, B.R.; Schwarz, R.; Vatner, R.; Yeh, B.; Claude, L.; Indelicato, D.J.; Laack, N. Osteosarcoma. Pediatr. Blood Cancer 2021, 68. [CrossRef]
- Prater S, M.B. Osteosarcoma. [Updated 2022 May 29]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan-. Available: https://www.ncbi.nlm.nih.gov/books/NBK549868/ (accessed on 28 January 2023).
- Czarnecka, A.M.; Synoradzki, K.; Firlej, W.; Bartnik, E.; Sobczuk, P.; Fiedorowicz, M.; Grieb, P.; Rutkowski, P. Molecular Biology of Osteosarcoma. Cancers (Basel). 2020, 12, 2130. [CrossRef]
- Synoradzki, K.J.; Bartnik, E.; Czarnecka, A.M.; Fiedorowicz, M.; Firlej, W.; Brodziak, A.; Stasinska, A.; Rutkowski, P.; Grieb, P. TP53 in Biology and Treatment of Osteosarcoma. Cancers (Basel). 2021, 13, 4284. [CrossRef]
- Brady, S.W.; Ma, X.; Bahrami, A.; Satas, G.; Wu, G.; Newman, S.; Rusch, M.; Putnam, D.K.; Mulder, H.L.; Yergeau, D.A.; et al. The Clonal Evolution of Metastatic Osteosarcoma as Shaped by Cisplatin Treatment. Mol. Cancer Res. 2019, 17, 895–906. [CrossRef]
- 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. [CrossRef]
- Bleve, A.; Consonni, F.M.; Porta, C.; Garlatti, V.; Sica, A. Evolution and Targeting of Myeloid Suppressor Cells in Cancer: A Translational Perspective. Cancers (Basel). 2022, 14, 510. [CrossRef]
- Deng, C.; Xu, Y.; Fu, J.; Zhu, X.; Chen, H.; Xu, H.; Wang, G.; Song, Y.; Song, G.; Lu, J.; et al. Reprograming the Tumor Immunologic Microenvironment Using Neoadjuvant Chemotherapy in Osteosarcoma. Cancer Sci. 2020, 111, 1899–1909. [CrossRef]
- Uehara, T.; Eikawa, S.; Nishida, M.; Kunisada, Y.; Yoshida, A.; Fujiwara, T.; Kunisada, T.; Ozaki, T.; Udono, H. Metformin Induces CD11b+-Cell-Mediated Growth Inhibition of an Osteosarcoma: Implications for Metabolic Reprogramming of Myeloid Cells and Anti-Tumor Effects. Int. Immunol. 2019, 31, 187–198. [CrossRef]
- Wright, E.M. SGLT2 and Cancer. Pflügers Arch. - Eur. J. Physiol. 2020, 472, 1407–1414. [CrossRef]
- Barbosa, A.M.; Martel, F. Targeting Glucose Transporters for Breast Cancer Therapy: The Effect of Natural and Synthetic Compounds. Cancers (Basel). 2020, 12, 154. [CrossRef]
- Wu, W.; Zhang, Z.; Jing, D.; Huang, X.; Ren, D.; Shao, Z.; Zhang, Z. SGLT2 Inhibitor Activates the STING/IRF3/IFN-β Pathway and Induces Immune Infiltration in Osteosarcoma. Cell Death Dis. 2022, 13, 523. [CrossRef]
|
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. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).