Abstract
Renal neoplasms are highlighted as one of the 10 most common types of cancer. Renal cell carcinoma (RCC) is the most common type of renal cancer, considered the seventh most common type of cancer in the Western world. The most frequently altered genes described as altered are VHL, PBRM1, SETD2, KDM5C, PTEN, BAP1, mTOR, TP53, TCEB1 (ELOC), SMARCA4, ARID1A, and PIK3CA. RCC therapies can be classified in three groups: monoclonal antibodies, tyrosine kinase inhibitors, and mTOR inhibitors. Besides, there are targeted agents to treat RCC. However, frequently patients present side effects and resistance. Even though many multidrug resistance mechanisms already have been reported to RCC, studies focused on revealing new biomarkers as well as more effective antitumor therapies with no or low side effects are very important. Some studies reported that natural products, such as honey, epigallocatechin-3-gallate (EGCG), curcumin, resveratrol, and englerin A showed antitumor activity against RCC. Moreover, nanoscience is another strategy to improve RCC treatment and reduce the side effects due to the improvement in pharmacokinetics and reduction of toxicities of chemotherapies. Taking this into account, we conducted a systemic review of recent research findings on RCC hallmarks, drug resistance, and adjuvant therapies. In conclusion, a range of studies reported that RCC is characterized by high incidence and increased mortality rates because of the development of resistance to standard therapies. Given the importance of improving RCC treatment and reducing adverse effects, nanoscience and natural products can be included in therapeutic strategies.
Keywords: Renal neoplasm, anticancer treatments, nanoscience, natural products, review
Cancer is a complex multifactorial disease considered the greatest problem of public health in recent decades and the second leading cause of death in the world, with an average of 9.8 million deaths per year (1).
Renal neoplasms are highlighted as one of the 10 most common types of cancer, mainly in the West. These cancers are classified into four main types: renal cell carcinoma (RCC), Wilms tumor, renal urothelial carcinoma, and renal sarcoma (2).
RCC is the most common type of renal neoplasm, considered the seventh most common type of cancer in the Western world, with an increase in incidence of 80%-90% of kidney cancers in adults, prevalence of 2%-3% of all malignancies in adults, and mortality of approximately 40% (3-5).
RCC is the third most prevalent type of genitourinary cancer and the most common malignancy in the kidneys, with 403,000 new diagnoses per year (2.2% of all tumors), and more than 175,000 deaths in the same period (1.8% of mortality from cancer) (6,7). The USA incidence is estimated at 76,080 new cases and 13,780 deaths in 2021 (3). The survival of patients with locally advanced and metastatic disease at 5 years is 69.6% and 12%, respectively (8,9). The GLOBOCAN study estimated an incidence of 2.2%-3.3% and mortality of 1.8%-2.6% in developed countries (7,10).
Moreover, several risk factors can be associated with this group of neoplasms, such as predisposition to chronic kidney disease, estrogen therapy, exposure to asbestos, petroleum and heavy metal products, diabetes, sedentary lifestyle, family history, poorly controlled hypertension, smoking, obesity, alcohol intake, and diet (8,11-21).
RCC can trigger local symptoms such as hematuria, pain in the lower back, palpable abdominal mass, and systemic symptoms, for example, weight loss, fever, and abdominal pain (22,23). Hematuria, low back pain, and palpable abdominal mass are considered the classic triad in diagnostic approaches to RCC. Furthermore, between 50%-80% of cases are diagnosed incidentally through abdominal ultrasound, contributing to the early detection of asymptomatic tumors (24-26).
Although more cases have been diagnosed early due to the increase in imaging exam applications, RCC still constitutes a great challenge to public health considering that it is characterized as a tumor frequently asymptomatic and with reduced clinical manifestation, when compared to the other types of cancers (9).
Renal Cell Carcinoma Staging and Subtypes
RCC is considered a complex disease characterized as a heterogeneous group of tumors with different genomic, histological, and clinical characteristics. In addition, other factors such as the extent of the disease, the presentation of different clinical phenotypes, and different responses to treatment make it even more complex (23,27-29).
RCC staging is determined considering the anatomical site of the primary tumor (T) (Figure 1), regional lymph node (N), and metastasis distance (M) according to American Joint Committee on Cancer (AJCC), known as TNM (30,31). Regarding the primary tumor (T): in the absence of evidence of a primary tumor is classified as T0, a tumor 7 cm or less and limited to the kidney as T1, if >7 cm and limited to the kidney as T2, if it reaches larger vessels or the perirenal tissue without going beyond Gerota’s fascia as T3, if it invades Gerota’s fascia as T4, and if it cannot be evaluated as TX. Regarding regional lymph nodes (N): if there are no metastases in lymph nodes is classified asN0, if there is lymph node metastasis as N1, if lymph nodes could not be evaluated as NX. Regarding distant metastasis (M): in the absence of distant metastases is classified as M0, if there are distant metastases as M1, if the metastasis cannot be evaluated as MX. Thus, these parameters can receive numerical graduations (T0 to T4; N0 to N1 and M0 to M1), alphabetical (a, b, c), and “X” when the category cannot be evaluated, giving more details on each of these analyzed aspects (30,32) (Table I).
There are three main subtypes based on the appearance of RCC, such as clear cell (ccRCC), papillary (pRCC), and chromophobic (crRCC). These subtypes represent approximately 90% of all RCCs. The remaining 10% comprise rare and benign subtypes (Table II) (5).
Genetic and Metabolic Hallmarks of RCC
Although the histological classification of renal tumors is an important tool in the diagnosis and evaluation of the prognosis of patients (33), molecular characteristics that differentiate the subtypes of this disease have been increasingly used. It is important to more precisely characterize the subtypes, as well as to improve the prognosis and treatment (34).
Taking this into account, studies have focused on the molecular mechanisms of RCC. Thus, The Cancer Genome Atlas (TCGA) includes several genomic studies, focusing on individual tumor subtypes, using data generated from multiple platforms (35).
Regarding the molecular profile of RCCs, recurrent alterations are described into specific subtypes. For example, the most prevalent RCC subtype, ccRCC, is associated with some mutations. The most frequent genes described as altered are VHL, PBRM1, SETD2, KDM5C, PTEN, BAP1, mTOR, TP53, TCEB1 (ELOC), SMARCA4, ARID1A, and PIK3CA. In addition to punctual alterations, alterations in chromosomes 3, 5, 10, and 14 are also frequently described in this subtype (34,36-40).
Approximately 80% of ccRCCs present inactivation of the Von Hoppel Lindau (VHL) gene by mutation or methylation (34,41). The VHL gene is a tumor suppressor and, in many cases, one of its alleles is inactivated by some kind of mutation, and the second is affected by a deletion in the 3p25-26 region in approximately 90% of cases of ccRCC (42-44), thereby playing an important role in both hereditary and sporadic disease (41). This mutation causes elevated levels of factors that induce hypoxia and increases the levels of vascular endothelial growth factor (VEGF), thereby facilitating tumor-associated angiogenesis. VEGF mediates neoangiogenesis that allows tumor nutrition and growth (45).
ccRCC is the subtype frequently present in hereditary von Hippel-Lindau syndrome, an autosomal dominant condition with germline mutations in the VHL gene. In this syndrome, approximately 40% of those affected will develop RCC, which is usually characterized by the presence of small tumors that develop in the kidneys (46,47).
The pRCC type, subdivided into types 1 and 2, also has specific genetic characteristics. Type 1 tumors are characterized by variants in MET gene, a proto-oncogene, which encodes a cell surface protein for hepatocyte growth factor, while type 2 tumors are characterized by alterations in the CDKN2A, SETD2, and FH genes (48). Changes in PBRM1, BAP1, and SETD2 are also found in the pRCC type 2 subtype, however, in lower frequencies than those observed in the ccRCC (35,37,48). In addition, methylation patterns have also been associated with subtype 2, characterizing a more aggressive disease with a lower survival rate (48).
The molecular characteristics of RCC of the chromophobe subtype have also been analyzed. Since it is a rare subtype, the identified alterations presented a lower frequency (approximately 10%) than those found in other subtypes. The genes found mutated in these cases were PTEN and TP53. However, it was also possible to identify alterations in mTOR, NRAS, and FLCN (variants germline related to hereditary syndrome). Furthermore, gene fusions with the gene TERT have been reported frequently in this subtype, and methylation profiles have been related to a more aggressive disease (49). Moreover, chromosomal changes are frequent in this subtype, such as loss of chromosomes Y, 1, 2, 6, 10, 13, 17, and 21 (23). In addition, a previous study reported that FH, FLNN, SDHB, and SDHD are also linked to hereditary RCC recurrence (50).
Renal Cell Carcinoma Treatments
RCC tumors have a rounded shape and variable size from few centimeters to complete occupancy of the abdomen. Small renal masses that increase over time and show increased contrast in computed tomography (CT) scans, should be considered extremely suspicious for renal neoplasms, although there are many inherent uncertainties (23). Therefore, the most suitable therapy for cases of localized disease is total or partial surgical resection of the tumor. Total resection is performed when the most conservative surgery is impossible, in cases of locally advanced disease, which occasionally will require resection of adjacent organs (8). Although it is an established and well-recognized therapy approach, studies show a certain heterogeneity between detected renal masses, where approximately 20% have a benign profile, 60% are considered indolent tumors, and 20% potentially aggressive tumors (51,52). These findings suggest that a more detailed characterization of these tumors may contribute to the choice of a less aggressive strategy in certain cases and intervention surgery in others.
Patients with metastatic disease present a heterogeneous group. Hence, different initial therapies can be used, and the most recurrent metastases are found in the lymph nodes, lungs, bones, and liver (8,53). Approximately 25% of patients diagnosed with RCC present with metastasis at diagnosis and 20-40% will develop metastasis after treatment of the primary tumor. Patients with metastatic disease have a survival average of 6 months to 1 year, and less than 20% of these patients survive more than 2 years (54,55).
Regarding the prognosis of patients with RCC, therapeutic strategies such as targeted therapy, inhibitors of tyrosine kinase and monoclonal antibodies, as well as the age of the patient and early diagnosis are essential factors for a good prognosis (56). RCC therapies can be categorized into three groups: monoclonal antibodies [such as bevacizumab (anti-VEGF) and nivolumab (anti-PD-1)], tyrosine kinase inhibitor (TKIs) (sorafenib, sunitinib, pazopanib, axitinib, and cabozantinib), and mTOR inhibitors (mTORi; temsirolimus and everolimus). Moreover, further investigations are been carried out to revel additional target agents, for example, TKIs regorafenib, cediranib, tivozanib, dovitinib, and lenvatinib (57).
RCC is a vascular tumor that is highly resistant to chemotherapy and radiotherapy. Some randomized studies have shown no benefits in using adjunctive systemic therapies such as interleukin-2 and interferon immunotherapies alpha (58), radiotherapies, and hormone treatments in patients with this neoplasm (23). Therefore, further advances in the understanding of biology of RCC are required to enable the use of new drugs (targeted therapy) in patients with RCC, such as therapies that inhibit the VEGF pathway, mTOR protein inhibitors, and PDGFR. Although these new therapies have increased overall survival and are specific to these patients, metastatic disease, in most cases, is still incurable and requires constant patient follow-up (59). In this scenario, numerous clinical and preclinical studies exploring the potential of therapies for the inhibition of immunological checkpoints have shown that these therapies can substantially contribute to the survival of patients with advanced renal neoplasms. These new therapeutic strategies work by blocking immune checkpoints, which normally prevent the development of an immune response against normal cells. This happens because some neoplasms can acquire these checkpoints, preventing tumor cells from being recognized by the immune system, and consequently inactivating the immune cells that can destroy them (60). Thus, the mechanisms of action of these immune checkpoint inhibitor therapies involve the removal of inhibitory signal activation of T cells, which enable tumor cells to overcome the mechanisms of immune system regulators (61-63). As a result, immunotherapies with checkpoint inhibitors (anti PD-1 and anti CTLA-4) are emerging in urologic cancer as promising treatment options. Clinical studies are ongoing, and thus far the results show that certain patients have a good response to treatment; however, more personalized strategies need to be used to better stratify the patients, taking into account their clinical and mainly molecular characteristics (64-67) (Figure 2).
The most used drugs for the treatment of advanced RCC are sorafenib, sunitinib, and temsirolimus. Sorafenib, a new oral small-molecule multikinase inhibitor, inhibits tumor growth and angiogenesis by targeting the RAF/MEK/ERK pathway and receptor tyrosine kinases with a response rate of 10%. Sunitinib acts as an antiangiogenic agent, inhibiting angiogenesis in tumors. This activity is achieved by inhibiting the receptor of vascular endothelial growth factor (VEGFR), which in turn inhibits angiogenic growth factors. Temsirolimus has an anticancer activity acting directly on the mTOR, a key mediator of tissue growth, proliferation, and angiogenesis, and its inhibition may also lead to growth reduction and stabilization, making it an immunosuppressant (58).
RCC tumors are highly infiltrated by immune cells, especially T cells. Taking advantage of these characteristics of the microenvironment, immunotherapy has advanced in the last decade, with VEGF TKIs, and immune checkpoint blockade. However, they also have other targets, which explains toxicity and possible additive antitumor effects (59).
Side Effects of Renal Cell Carcinoma Therapies
Antiangiogenic agents are considered standard therapy for RCC. Although these therapies are considered inhibitors of angiogenesis, they also present other mechanisms. This aspect is important to determine the efficacy and side effects of the therapy. These therapeutic agents can be divided into three groups: VEGF, Tyrosine kinases, and mTOR inhibitors (68).
Bevacizumab, also known as avastin, is a recombinant human IgG1 monoclonal antibody (MAb) that recognizes all isoforms of human VEGF and binds to it and prevents VEGF from binding to VEGF receptors on endothelial cells. This treatment has demonstrated a range of side effects (68,69).
Some adverse effects have been associated with these agents, such as hypertension caused by the decrease in the production of vasodilator substances, stimulated by VEGF and VEGFR-2, increasing peripheral vascular resistance. Proteinuria is another side effect since inhibition of VGFR affects the glomerular filtration barrier due to its cytoprotective function of endothelial cells. Regarding wound healing and bleeding, agents inhibiting VEGF can affect endothelial turnover in response to trauma and trigger high clotting, leading to thromboembolic events (68,69).
Tyrosine kinases participate in the process of growth and development of cancer through signaling pathways, such as VEGF. TKIs inhibit the activity of receptor tyrosine kinases 1, 2, and 3; however, it is non-specific, which leads to enhancement of other receptor tyrosine kinases. Thus, this treatment affects transmission of multiple signaling pathways, which leads to a wide range of side effects (70).
The side effects caused by TKIs are manifested according to the signaling pathway that the TKIs will act on; when the TKIs affect the VEGF pathway, the side effects are identical to those of VEGF inhibitors. When they affect other signaling pathways, the effects can be those observed in Table III (69).
The inhibition of mTOR activity is very important in RCC therapies because, together with tyrosine kinase receptors, mTOR regulates protein biosynthetic pathways that promote cell growth and angiogenesis. The most used mTOR therapies are temsirolimus and everolimus (70).
The most observed side effects of mTOR inhibition are related to metabolic abnormalities such as hyperglycemia, hypertriglyceridemia, hypercholesterolemia, and hypophospha-temia. As reported for TKIs, these inhibitors present no selective profile, they also affect other signaling pathways, which can lead to side effects similar to those of TKIs. Due to the great complexity of mTOR-related signaling pathways, the pathophysiology of side effects caused by these inhibitors is still not completely clear. However, some side effects can be explained.
Resistance to Systemic Therapies in Renal Cell Carcinoma
Response Evaluation Criteria in Solid Tumors (RECIST) are criteria used to measure the response to anticancer therapy. Cancer resistance is determined if RECIST achieves a result of 20% or more in the sum of some parameters, such as measurable lesions, the development of new tumors, or an unequivocal progression of non-measurable disease, for example, small lung nodules or bone lesions (71).
Two types of cancer resistance to targeted therapeutics can be found, intrinsic (primary) and acquired (secondary) resistance. When the therapies immediately fail, the tumor is classified as intrinsic resistant due to the presence of resistant cells before the treatment via inherited resistance or evolutionary clonal selection. However, when cancer cells resume proliferation after the first regression, during the treatment, resistance is classified as acquired. Some resistance pathways have already been revealed (72). However, more studies should be performed to further explain these cancer cell strategies to resist therapies.
A previous study reported some processes associated with primary resistance in RCCs, such as apoptosis inhibition, epigenetic modifications of histone proteins, and ATP-binding cassette (ABC) drug transporters. Primary resistance is also associated with apoptosis blockage via a rise in B-cell lymphoma-2 (Bcl-2) and/or Bcl-XL proteins and a decrease in CD95 expression (73,74). Moreover, another dysfunctional pathway that has been associated with primary resistance is the VEGF pro-angiogenic signaling pathway (75).
Regarding acquired resistance, activation of alternative pro-angiogenic pathways, resistance mediated by the tumor microenvironment, increased invasiveness and metastasis, lysosomal sequestering, single-nucleotide polymorphisms, and microRNAs play important roles (76).
Studies have reported that 26% of patients treated with sorafenib and sunitinib exhibit primary resistance to the treatment. Most of these patients present poor results, independently of subsequent therapy (77). Besides, some patients that are susceptible to therapies targeting the VEGF pathway, frequently develop secondary or acquired resistance after chronic treatment (Figure 3).
Primary and Acquired Resistance to Systemic Therapeutic Agents: Acquired Mechanisms
Lysosomal sequestration via TKIs. Reduced influx of TKIs by RCC is one of the mechanisms of multidrug resistance (78). Lysosomal sequestration of TKIs has been shown to play an important role in cellular adaptation to the treatment (79). Moreover, TKIs resistance and increased metastasis can be related to epithelial-to-mesenchymal transition (80).
Studies reported that lysosomes have 50 different acid hydrolases activated by the acidic pH around 4.6 to 5.0, which is maintained through the use of vacuolar ATPases. Since sunitinib is a hydrophobic weak base, it might favorably accumulate in acidic lysosomes. TKIs are weak bases, which are sequestered by being trapped in their protonated form and they do not reach their local target. Resistance to sunitinib, erlotinib, and pazopanib can be associated with this mechanism (81).
Inactivation of Von Hoppel Lindau protein. The inactivation of VHL proteins triggers over-expression and activation of receptor tyrosine kinases MET and AXL. Both MET and AXL signaling pathways have been associated with clinical resistance to VEGF-targeted therapeutics agents (72). High levels of MET or AXL oncoproteins are associated with poor clinical prognosis. Given the fact that MET and AXL up-regulation present an important role in association with VEGF for RCC development and progression, agents that are able to inhibit these molecules can be an excellent strategy to selective RCC therapy (82).
In addition, the increase in VEGF action is related to the connection of receptor tyrosine with the angiopoietin 1 and 2 (Ang 1 and Ang 2) pathway (83). HIFs are responsible for increasing VEGF, interleukins 6 and 8 (IL-6 and IL-8), hepatocyte growth factor (HGFR/C-MET), fibroblast growth factor (FGF2) and some other growth factors (72,83). IL-6 and IL-8 also have an important role in tumor angiogenesis, and their levels are increased during treatment with sunitinib and pazopanib. The activation of chemokine receptor 2 contributes to the transcription and translation of VEGF mRNA leading to increased levels of VEGF protein, which activates VEGFR-2 in an autocrine manner (84).
Increased invasiveness and metastasis via angiogenic switch. Some studies have reported that alterations in genes associated with angiogenesis and the rise in pericyte coverage of tumor vessels result in the recruitment of pro-angiogenic inflammatory cells from the bone marrow and metastatic activity in RCC (75).
Moreover, studies have shown that the use of sunitinib facilitates the formation of tubules and the proliferation of endothelial cells through FGF. This growth factor also activates alternative pathways such as MAPK, ERK, PI3K, and AKT (84).
Antiangiogenic mechanisms are up-regulated by hypoxia-inducible factors and correlate with poor prognosis and resistance to VEGF receptor inhibitors in preclinical models of RCC and other cancers (82,85). Besides, RCC cells express HLA-G and HLA-E on their surface that decrease the immune response and promote early tolerance (86).
Growth increase via alternative pathways activation. A study reported some molecular patterns associated with primary resistance in ccRCCs, such as the absence of HIF-α protein and wild-type VHL alleles; VHL-deficient tumors, expressing detectable HIF-1α and HIF-2α; and VHL-deficient tumors expressing HIF-2α exclusively (87). Wild-type tumors and those that expressed both HIF-1α and HIF-2α showed an increase in AKT/mTOR and MAPK pathways and were more sensitive to TKI. In contrast, tumors that expressed only HIF-2α showed increased c-Myc activity, triggering proliferation and increased resistance. These results suggest that HIF-1α and HIF-2α activate distinct oncogenes in ccRCC (87).
Studies reported that loss of PTEN is correlated with sunitinib resistance in renal cells. However, the up-regulation of PTEN or the inhibition of AKT/mTOR pathway increases the response of PTEN-deficient ccRCC cells to sunitinib via apoptosis activation (88). Moreover, PTEN down-regulation is associated with poor sensitivity to bevacizumab (89).
ATP-binding cassette (ABC) efflux transporters. Membrane structures were associated with multidrug resistance, for example, ATP-binding cassette (ABC) drug transporters [P-glycoprotein (Pgp, ABCB1), and multidrug resistance associated protein (MRP) 1 (ABCC1)] (90).
Alterations in gene expression levels. The action of histone deacetylases and methyltransferases, enzymes that control epigenetic modifications, have been reported to be modified in RCC (37). Histone methyltransferase EZH2 over-expression is associated with tumor angiogenesis via blocking anti-angiogenic factors by promoter gene methylation, causing low response to sunitinib (91).
Tumoral heterogeneity. Heterogeneity can be found even among cells in the same tumor, including variations in gene, microRNA, and protein expression (89). For example, post-sunitinib metastatic lesions show FLT4, KMT2D, and BMP5 mutations, which were not found in the primary tumor (92).
Previous studies reported that after treatment with sorafenib, RCC cancer cells developed mutations and morphologic heterogeneity compared to untreated subjects (93). Moreover, studies suggest that VEGF-targeted therapy can induce polyclonal outgrowth of tumor cell subclones that can result in poor treatment response (93).
Tumor microenvironment. The tumor microenvironment is composed of tumor cells, extracellular matrix (ECM), signaling molecules, and stromal cells, such fibroblasts, vascular endothelial cells, pericytes, and immune cells. Myeloid-derived suppressor cells (MDSCs) are significantly found in the tumor microenvironment since they are potent immunosuppressors. Due to this fact, MDSCs are highly recruited by tumors to trigger low response to anti-angiogenic drugs via increasing pro-angiogenic factors that can activate VEGF-independent angiogenesis (94).
It has been shown that patients that received sunitinib treatment presented a reduction in MDSCs in peripheral blood; on the other hand, the tumor tissue did not show a decrease in MDSCs (95). Moreover, pericytes, which are considered stromal cells, are also involved in aberrant tumor angiogenesis and drug resistance.
Resistance mediated by the action of microRNAs. RCC can exhibit different patterns of miRNA expression that can result in therapy resistance. Sunitinib-resistant RCC tumors present an increase in the expression of miRNA-942, miRNA-133a, miRNA-628-5p, and miRNA-484 when compared to sunitinib-sensitive tumors.
The up-regulation of miRNA-942 in an mRCC cell line can increase the production of MMP-9 and VEGF that result in the migration of endothelial cells and sunitinib resistance (96).
Adjuvant Therapies to Improve RCC Treatment Focus on Natural Products
Even though many multidrug resistance mechanisms already have been reported in RCC, studies focusing on new biomarkers as well as selected antitumor therapies with no or low side effects and more effective are very important.
Currently, the standard therapy for RCC is partial nephrectomy (97). Alternative treatment options are available, such as stereotactic body radiotherapy, microwave ablation, cryoablation, radiofrequency ablation, and active surveillance (98). Unfortunately, there is no adjuvant treatment for RCC. However, investigations in this field are very important, due to the fact that the 5-year relapse rate for intermediate- and high-risk early-stage RCC is 30% to 40% (99). Metastatic RCC can be treated successfully with immune therapy and targeted therapy (100). Pazopanib and sunitinib are currently the standard first-line treatment for metastatic RCC, both with similar efficacy, although the safety profile favors the use of pazopanib (101). Various adjuvant trials with immune therapy have been conducted. However, they reported no benefit in disease-free survival, and clinical trials with targeted agents have not reported results yet (102). Taking this into account, new treatments against RCC that present a selective profile and are more effective should be developed. Thus, some natural products that exhibit several bioactivities have been investigated in this area.
Honey
Previous studies have indicated that honey may have antitumor activity in human renal carcinoma cell lines (ACHN), mainly by activating apoptosis (103). Samarghandian et al. (104) reported that treatment of ACHN cells with different concentrations of honey for three days decreased cell viability in a concentration and time-dependent manner. The IC50 values of honey against the ACHN cell lines were 1.7±0.04% and 2.1±0.03% μg/ml after two and three days post treatment, respectively. However, further studies are necessary to understand the exact molecular mechanism behind the antitumor activity of honey in this context.
Epigallocatechin-3-gallate (EGCG)
Epigallocatechin-3-gallate (EGCG), a component derived from Camellia sinensis (green tea) that is also found in apple, shows cytotoxic effects in RCC; it inhibited tumor growth and invasiveness in RCC by up-regulating expression of TFPI-2 through inhibition of DNA methyltransferase (DNMT) activity (105). Additional studies indicated that EGCG sensitized human 786-0 renal cell carcinoma cell lines to apoptosis by down-regulating c-FLIP, Mcl-1, and Bcl-2 proteins in a caspase-dependent pathway, while inhibited proliferation and migration of these cells by down-regulating matrix metalloproteinase-2 and matrix metalloproteinase-9 (106,107).
Curcumin
Curcumin, (1E,6E)-1,7-Bis(4-hydroxy-3-methoxyphenyl) hepta-1,6-diene-3,5-dione, is the primary bioactive substance found in turmeric (Curcuma longa). This molecule presents anti-cancer effects in melanoma cell lines and RCC by inhibition of Signal transducer and activator of transcription 3 (STAT3) phosphorylation with specificity for the Jak-2-STAT3 pathway (108). Several studies also showed that curcumin promoted apoptosis in vitro in various human cancer cell lines, including RCC (26,109,110), by decreasing activation of the PI3K/AKT signaling pathway. Besides apoptosis, curcumin can also inhibit the proliferation of RCC and activate autophagy via Akt/mTOR suppression and beclin-1 activation (111).
Resveratrol
Resveratrol, (trans-3,5,4’-trihydroxystilbene), a polyphenolic compound found in grapes, can induce apoptosis and cell cycle arrest, and inhibit proliferation of RCC via JAK-1, c-Src, and STAT3/5 over-expression and PTPε and SHP-2 tyrosine phosphatase activation (112). Other findings suggest that resveratrol induces differential expression of genes that are related to the inhibition of RCC cell growth and induction of RCC cell death, and these effects depend directly on resveratrol concentration (113). Resveratrol also seems to have inhibitory effects on the expression of VEGF gene and the proliferation of RCC cells (786-0) (83).
Englerin A
Englerin A is a compound derived from the Phyllanthus engleri, a Southern Africa native tree known to have medicinal properties (114). It has been shown that Englerin A induces cell death in RCC via induction of apoptosis, inhibition of cell migration, activation of autophagy, and cell cycle arrest by PI3/AKT/ERK inhibition and PKCθ activation (115,116). The main mechanism of action of this compound is through activation of canonical transient receptor potential channels (TRPCs, especially TRPC4 and TRPC5), which are found in the membranes of renal cells (117). Englerin A acts on these channels, elevates the intracellular concentration of calcium, and induces cell death (118). Englerin A can also prevent the migration and invasion of RCC cells by TGF-β1 transformation (119). Studies investigating the in vivo effects of Englerin A have not been conducted yet, however, trials on mouse models suggest that the levels of this compound needed for anti-tumor effects may be fatal (120).
Despite of the many treatment options currently available, RCC is still one of the deadliest forms of cancer, which demonstrates a great need for new drugs that are effective against this disease. After reviewing the scientific literature on the subject, it is understood that there are bioactive natural products with anti-tumor effects in RCC, acting through mechanisms such as induction of apoptosis, inhibition of tumor growth, and activation of autophagy (Table IV). Therefore, it is evident that natural products may have a broad range of potential applications in the future; however, more clinical studies regarding these molecules are necessary.
Nanoparticles Can Enhance Renal Cell Carcinoma Treatment
Nanotechnology and nanoscience are fundamentally related regarding to develop new materials and devices with improved properties in a nanometric scale between 1 and 100 nm (121,122). At this size, their proprieties and characteristics are distinct from their bulk form mostly due to the surface and quantum confinement effects (123). The effects related to the surface properties are associated with the increase in the area/volume ratio, which can enhance the specific area and porosity (124). Besides, quantum confinement is involved in the optical and electronic characteristics (125).
Diverse nanomaterials have been developed and improved to increase the performance in different applications. The unique properties and characteristics of nanomaterials (NMs) and nanoparticles (NPs) make them excellent agents for application in different areas, such as water remediation (126,127), medicine (128,129), development of sensors (130), drug delivery (131), and cancer treatment (132). Figure 4 shows some characteristics of nanoparticles.
Nanoparticles, due to their extraordinary ability to co-encapsulate different therapeutic agents, can also be employed to overcome drug resistance in cancer. Nanoparticles have excellent properties, such as reactive and surface area, that can be used to improve the interactions between drugs and cells as well as overcome cancer resistance. In support, NPs showed benefits in cancer therapy: greater pharmacokinetics, precise targeting, and reduced side effects (133). For instance, polymeric nanoparticles (NPs) have been extensively studied due their physico-chemical properties allowing encapsulation of known drugs. Biocompatible polymers are used mostly for their ability to transport drugs directly to the targeted tissue through surface modifications (134). Also, micelles are suitable as a template to incorporate gold NPs, regulating their sizes with a pH-sensitive triblock copolymer micelle, i.e., bringing relevant applications to circumvent drug resistance by acting in biological processes such as ion transport and targeted drug delivery (135). It is well-known that conventional therapies have several drawbacks relating to their efficacy and side effects, such as damage in healthy cells and tissues. In this manner, the size and shape furnished by the nanotechnology, allows a decrease in the oxidative stress and delivery of the drug to the target organ/tissue (136).
RCC is fundamentally originated from the renal cortex and presents a high metastatic rate (137). Nevertheless, due to the side effects of cancer treatment, such as nausea and blood clots, new research develops alternatives to improve the treatment efficiency and appease the effects (138). NPs are an excellent alternative for cancer cell treatment due to the improvement in the pharmacokinetics and reduction of toxicities of chemotherapies (133) (Figure 5).
Magnetic nanoparticles (MNPs), such as magnetite (Fe3O4) and hematite (Fe2O3), are widely used in different areas due to their excellent properties: biocompatibility, reactivity, and high surface area (139). To evaluate the anticarcinogenic activity of nanoparticles, Abbas and co-workers (140) synthesized an α-Fe2O3 (NPLAA@IONP-PEG) employing polyethylene glycol (PEG) and ascorbic acid (LAA), and tested it against the HEK-293 human embryonic kidney cell line. LAA@IONP-PEG decreased cell viability in a dose-dependent manner. The high cytotoxicity against cancer cells of the nanocomposite was attributed to the small size and the presence of PEG and LAA (141). It was shown that LAA@IONP-PEG presents an antioxidant capability.
Furthermore, Nagajyothi et al. (142) synthesized iron oxide NPs (α-Fe2O3) and analyzed their catalytic and anticancer behavior. The in vitro experiments indicated that α-Fe2O3 inhibited the growth of the RCC line Caki-2in a dose-dependent manner. However, only the highest concentration (0.8 mg ml–1) (Figure 6) showed a cytotoxic effect on normal cells.
Inorganic nanoparticles, such silver (AgNPs) and gold (AuNPs) nanoparticles, are an important alternative employed in cell treatments due to their excellent properties, such as antibacterial and antimicrobial activities (143,144). Chen and coauthors (135) reported the mechanisms and toxicity effects of AgNPs against HEK293T and A498 cell lines. The results showed that low concentrations (1-8 μg ml–1) had no significant effect on cell viability and ROS production compared to the untreated control. However, AgNPs increased autophagy even at low concentrations; increasing the LC3II level and autophagy-associated genes.
Inorganic nanoparticles are also a viable alternative for gene delivery. Shi et al. (2020) developed a polyethylene glycol modified with manganese dioxide (PEG-MnO2), which they loaded with osteopontin siRNA, as the gene drug (siRNA OPN) (PEG-MnO2-OPN siRNA), and used for magnetic resonance imaging (MRI) for guided gene delivery (145). In vitro assays demonstrated that the PEG-MnO2 was cytotoxic against the RCC line 786-O and umbilical vein EA hy926 cell line. Furthermore, contrast of MRI showed that the nanocomposite resulted in a significant improvement, even at low concentrations, due to the presence of Mn2+ ions and the anti-tumor effect of nanoparticles. Nonetheless, Chai and co-authors (146) synthesized folate grafted PEI600-CyD (H1) nanoparticles for the delivery of AIM2 gene (H1/pAIM2) for renal carcinoma cell treatment (786-O and OSRC-2). The nanocomposite significantly decreased tumor volume and weight. The in vitro results showed that the NPs enhanced the effect of AIM2 against cancer cell lines, decreasing cell migration and invasion.
Soliman and co-workers (147) synthesized NPs with succinyl chloride (SC) and loaded them with gemcitabine (GT) and 5-Fluoracil (5FU), called GT-SC-5FU. In vitro experiments showed that GT-SC-5FU significantly decreased the viability of the RCC line SNU-349 in a dose-dependent manner. Nonetheless, GT-SC-5FU presented higher cytotoxicity when compared to individual drugs (GT and 5FU), suggesting a drug synergistic effect in the SNU-349 cell line.
Green synthesis can be used to produce high quality inorganic nanoparticles with good yields (148). Along with this, biological synthesis (green approaches) of metal NPs are an excellent alterative to produce ecological and environmentally friendly nanomaterials with diverse benefits (149). Liu et al. (150) used an easy and green technique for development of gold nanoparticles, Curcuma wenyujin extraction, which was called CWAuNP. Treatment of the RCC lines A498 and Sw-156 increasing concentrations of gold nanoparticles (5-50 μg/ml) significantly decreased cell viability. In addition, the production of reactive oxygen species (ROS) increased and the mitochondrial membrane potential decreased.
Furthermore, Li and co-workers (151) synthesized copper nanoparticles (Cu-NPs) using the medicinal plant Ziziphus zizyphus (Cu-ZZ NPs) and evaluated the anti-cancer activity against A498 cells. Cu-ZZ NPs significantly decreased viability and increased mitochondrial membrane potential (MMP) in a concentration dependent manner (10-50 μg ml–1). These results suggest that Cu-ZZ NPs has a dose-dependent anti-tumor activity due to the DNA damage.
Considering their excellent characteristic, zinc oxide nanoparticles (ZnO-NPs) produced through a green synthesis approach can be used in different applications (152). Lokapur and co-workers (153) evaluated the toxicity of ZnO-NPs produced using Holigarna grahamii (HG) against the A498 cell line. The results showed that by increasing the concentration of nanoparticles the cytotoxicity effect increased and cell viability decreased to approximately 50%. Nevertheless, no significant toxicity was observed against health cells.
Zhou and Chen (154) developed supramolecular nanoparticles with cisplatin (CIS-PT-NPs) and evaluated their anticancer properties, such as antiproliferative activity and toxicity, against the RCC lines Caki-1 and A498 and compared them to those of CIS-PT (no nanometric material). MTT assay showed a higher cytotoxic activity of CIS-PT-NPs in comparison to CIS-PT; treatment resulted in a significant and dose-dependent decrease in cell viability. However, none of the materials (CIS-PT and CIS-PT-NPs) presented toxicity against non-cancerous cells (NIH-3T3). The drug release profile showed that CIS-PT liberate the drug faster than the nanoparticles (90% after 25 h). However, the kinetic baum profile of CIS-PT-NPs demonstrated a continuous drug release, enhancing cancer treatment.
Conclusion
In conclusion, RCC present increased incidence and mortality rates. This can be because this type of cancer frequently exhibits resistance to treatment. Furthermore, most anticancer agents used to treat RCC have side effects, since they present a non-selective profile. In this scenario, studies have been performed to reveal new therapies with low or null side effects. Nanoscience and natural products can be highlighted in this area. However, further clinical studies regarding these new RCC anticancer agents are necessary.
Conflicts of Interest
The Authors have no conflicts of interest to report in relation to this study.
Authors’ Contributions
Wrote the manuscript: Josiéle da Silva Prade, Ruan Soares de Souza, Camila Medianeira da Silva D’Ávila, Thayline Correia da Silva, Isadora Cassel Livinalli, Ana Clara Zanini Bertoncelli, Fernanda Krapf Saccol, Tallys de Oliveira Mendes, Larissa Gindri Wenning, Theodoro da Rosa Sallesa, Cristiano Rodrigo Bohn Rhodena, Francine Carla Cadoná. Provided figures and tables: Theodoro da Rosa Sallesa, Cristiano Rodrigo Bohn Rhodena, Francine Carla Cadoná, Ruan Soares de Souza, Camila Medianeira da Silva D’Ávila, Thayline Correia da Silva, Isadora Cassel Livinalli. Conception and design of the study: Francine Carla Cadoná, Cristiano Rodrigo Bohn Rhodena, Josiéle da Silva Prade.
Acknowledgements
The Authors would like to express their gratitude to the Franciscan University research team for all their help and support.
References
1
Pereira CAG
,
de Araújo Pontes BL
,
Valente TR
,
Moura AF
,
de Mesquita RB
&
Mont’Alverne DGB
. Influence of gastric and hematological cancers on the quality of life and the functionality of oncological patients. Rev Bras Cancerol.
68(1)
e051332
2022.
DOI:
10.32635/2176-9745.RBC.2022v68n1.1332
2
Calais da Silva F
. Recomendações clínicas no tratamento do carcinoma de células renais. 1ª ed. Grupo Português Génito-Urinário: Sociedade Portuguesa de Oncologia.
3
Siegel RL
,
Miller KD
,
Fuchs HE
&
Jemal A
. Cancer Statistics, 2021. CA Cancer J Clin.
71(1)
7
- 33
2021.
DOI:
10.3322/CAAC.21654
4
Voog E
,
Campillo-Gimenez B
,
Elkouri C
,
Priou F
,
Rolland F
,
Laguerre B
,
Elhannani C
,
Merrer J
,
Pfister C
,
Sevin E
,
L’Haridon T
,
Hasbini A
,
Moise L
,
Le Rol A
,
Malhaire JP
,
Delva R
,
Vauléon E
,
Cojocarasu O
,
Deguiral P
,
Cumin I
,
Cheneau C
,
Schlürmann F
,
Delecroix V
,
Boughalem E
,
Mollon D
,
Ligeza-Poisson C
,
Abadie-Lacourtoisie S
,
Monpetit E
,
Chatellier T
,
Desclos H
,
Coquan E
,
Joly F
,
Tessereau JY
,
Dupuy S
,
Lagadec DD
,
Marhuenda F
&
Grudé F
. Long survival of patients with metastatic clear cell renal cell carcinoma. Results of real life study of 344 patients. Int J Cancer.
146(6)
1643
- 1651
2020.
DOI:
10.1002/IJC.32578
5
Muglia VF
&
Prando A
. Renal cell carcinoma: histological classification and correlation with imaging findings. Radiol Bras.
48(3)
166
- 174
2015.
DOI:
10.1590/0100-3984.2013.1927
6
Galceran J
,
REDECAN Working Group
,
Ameijide A
,
Carulla M
,
Mateos A
,
Quirós JR
,
Rojas D
,
Alemán A
,
Torrella A
,
Chico M
,
Vicente M
,
Díaz JM
,
Larrañaga N
,
Marcos-gragera R
,
Sánchez MJ
,
Perucha J
,
Franch P
,
Navarro C
,
Ardanaz E
,
Bigorra J
,
Rodrigo P
&
Bonet RP
. Cancer incidence in Spain, 2015. Clin Transl Oncol.
19(7)
799
- 825
2017.
DOI:
10.1007/s12094-016-1607-9
7
Bray F
,
Ferlay J
,
Soerjomataram I
,
Siegel RL
,
Torre LA
&
Jemal A
. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin.
68(6)
394
- 424
2018.
DOI:
10.3322/caac.21492
8
Ljungberg B
,
Albiges L
,
Abu-Ghanem Y
,
Bensalah K
,
Dabestani S
,
Fernández-Pello S
,
Giles RH
,
Hofmann F
,
Hora M
,
Kuczyk MA
,
Kuusk T
,
Lam TB
,
Marconi L
,
Merseburger AS
,
Powles T
,
Staehler M
,
Tahbaz R
,
Volpe A
&
Bex A
. European Association of Urology Guidelines on Renal Cell Carcinoma: The 2019 update. Eur Urol.
75(5)
799
- 810
2019.
DOI:
10.1016/j.eururo.2019.02.011
9
Huang JJ
&
Hsieh JJ
. The therapeutic landscape of renal cell carcinoma: from the dark age to the golden age. Semin Nephrol.
40(1)
28
- 41
2020.
DOI:
10.1016/j.semnephrol.2019.12.004
10
Ferlay J
,
Soerjomataram I
,
Dikshit R
,
Eser S
,
Mathers C
,
Rebelo M
,
Parkin DM
,
Forman D
&
Bray F
. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer.
136(5)
E359
- E386
2015.
DOI:
10.1002/IJC.29210
11
Kotecha RR
,
Motzer RJ
&
Voss MH
. Towards individualized therapy for metastatic renal cell carcinoma. Nat Rev Clin Oncol.
16(10)
621
- 633
2019.
DOI:
10.1038/s41571-019-0209-1
12
Sanchez A
,
Furberg H
,
Kuo F
,
Vuong L
,
Ged Y
,
Patil S
,
Ostrovnaya I
,
Petruzella S
,
Reising A
,
Patel P
,
Mano R
,
Coleman J
,
Russo P
,
Liu CH
,
Dannenberg AJ
,
Chan TA
,
Motzer R
,
Voss MH
&
Hakimi AA
. Transcriptomic signatures related to the obesity paradox in patients with clear cell renal cell carcinoma: a cohort study. Lancet Oncol.
21(2)
283
- 293
2020.
DOI:
10.1016/S1470-2045(19)30797-1
13
Bao C
,
Yang X
,
Xu W
,
Luo H
,
Xu Z
,
Su C
&
Qi X
. Diabetes mellitus and incidence and mortality of kidney cancer: A meta-analysis. J Diabetes Complications.
27(4)
357
- 364
2013.
DOI:
10.1016/J.JDIACOMP.2013.01.004
14
Al-Mutawa A
,
Al-Sabah S
,
Anderson AK
&
Al-Mutawa M
. Evaluation of nutritional status post laparoscopic sleeve gastrectomy – 5-year outcomes. Obes Surg.
28(6)
1473
- 1483
2018.
DOI:
10.1007/s11695-017-3041-7
15
Albiges L
,
Hakimi AA
,
Xie W
,
McKay RR
,
Simantov R
,
Lin X
,
Lee JL
,
Rini BI
,
Srinivas S
,
Bjarnason GA
,
Ernst S
,
Wood LA
,
Vaishamayan UN
,
Rha SY
,
Agarwal N
,
Yuasa T
,
Pal SK
,
Bamias A
,
Zabor EC
,
Skanderup AJ
,
Furberg H
,
Fay AP
,
de Velasco G
,
Preston MA
,
Wilson KM
,
Cho E
,
McDermott DF
,
Signoretti S
,
Heng DYC
&
Choueiri TK
. Body mass index and metastatic renal cell carcinoma: Clinical and biological correlations. J Clin Oncol.
34(30)
3655
- 3663
2016.
DOI:
10.1200/JCO.2016.66.7311
16
Campeggi A
,
Xylinas E
,
Ploussard G
,
Ouzaid I
,
Fabre A
,
Allory Y
,
Vordos D
,
Abbou CC
,
Salomon L
&
De La Taille A
. Impact of body mass index on perioperative morbidity, oncological, and functional outcomes after extraperitoneal laparoscopic radical prostatectomy. Urology.
80(3)
576
- 584
2012.
DOI:
10.1016/J.UROLOGY.2012.04.066
17
Jensen BW
,
Meyle KD
,
Madsen K
,
Sørensen TIA
&
Baker JL
. Early life body size in relation to risk of renal cell carcinoma in adulthood: a Danish observational cohort study. Eur J Epidemiol.
35(3)
251
- 258
2020.
DOI:
10.1007/S10654-020-00605-8
18
Xu WH
,
Qu YY
,
Wang J
,
Wang HK
,
Wan FN
,
Zhao JY
,
Zhang HL
&
Ye DW
. Elevated CD36 expression correlates with increased visceral adipose tissue and predicts poor prognosis in ccRCC patients. J Cancer.
10(19)
4522
- 4531
2019.
DOI:
10.7150/jca.30989
19
Capitanio U
,
Bensalah K
,
Bex A
,
Boorjian SA
,
Bray F
,
Coleman J
,
Gore JL
,
Sun M
,
Wood C
&
Russo P
. Epidemiology of renal cell carcinoma. Eur Urol.
75(1)
74
- 84
2019.
DOI:
10.1016/j.eururo.2018.08.036
20
Kabaria R
,
Klaassen Z
&
Terris MK
. Renal cell carcinoma: links and risks. Int J Nephrol Renovasc Dis.
9
45
- 52
2016.
DOI:
10.2147/IJNRD.S75916
21
Navai N
&
Wood CG
. Environmental and modifiable risk factors in renal cell carcinoma. Urol Oncol Semin Orig Investig.
30(2)
220
- 224
2012.
DOI:
10.1016/j.urolonc.2011.10.001
22
Figliuolo G
,
Alarcón KMG
,
Costa DM
&
Silva FLT
. Estudo epidemiológico sobre câncer renal para conhecimento de sua incidência no estado do amazonas. Urominas.
3(7)
19
- 24
2016.
24
Chang YH
,
Chuang CK
,
Pang ST
,
Wu C Te
,
Chuang KL
,
Chuang HC
&
Liao SK
. Prognostic value of TNM stage and tumor necrosis for renal cell carcinoma. Kaohsiung J Med Sci.
27(2)
59
- 63
2011.
DOI:
10.1016/J.KJMS.2010.12.004
25
Kane CJ
,
Mallin K
,
Ritchey J
,
Cooperberg MR
&
Carroll PR
. Renal cell cancer stage migration. Cancer.
113(1)
78
- 83
2008.
DOI:
10.1002/CNCR.23518
28
Wilder-Smith A
&
Freedman DO
. Isolation, quarantine, social distancing and community containment: pivotal role for old-style public health measures in the novel coronavirus (2019-nCoV) outbreak. J Travel Med.
27(2)
taaa020
2020.
DOI:
10.1093/jtm/taaa020
29
Smith CG
,
Moser T
,
Mouliere F
,
Field-Rayner J
,
Eldridge M
,
Riediger AL
,
Chandrananda D
,
Heider K
,
Wan JCM
,
Warren AY
,
Morris J
,
Hudecova I
,
Cooper WN
,
Mitchell TJ
,
Gale D
,
Ruiz-Valdepenas A
,
Klatte T
,
Ursprung S
,
Sala E
,
Riddick ACP
,
Aho TF
,
Armitage JN
,
Perakis S
,
Pichler M
,
Seles M
,
Wcislo G
,
Welsh SJ
,
Matakidou A
,
Eisen T
,
Massie CE
,
Rosenfeld N
,
Heitzer E
&
Stewart GD
. Comprehensive characterization of cell-free tumor DNA in plasma and urine of patients with renal tumors. Genome Med.
12(1)
23
2020.
DOI:
10.1186/s13073-020-00723-8
30
Edge SB
&
Compton CC
. The American Joint Committee on Cancer: the 7th Edition of the AJCC Cancer Staging Manual and the Future of TNM. Ann Surg Oncol.
17(6)
1471
- 1474
2010.
DOI:
10.1245/S10434-010-0985-4
31
Amin MB
,
Greene FL
,
Edge SB
,
Compton CC
,
Gershenwald JE
,
Brookland RK
,
Meyer L
,
Gress DM
,
Byrd DR
&
Winchester DP
. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin.
67(2)
93
- 99
2017.
DOI:
10.3322/CAAC.21388
32
Guinan P
,
Sobin LH
,
Algaba F
,
Badellino F
,
Kameyama S
,
Maclennan G
&
Novick A
. TNM staging of renal cell carcinoma: Workgroup No. 3. Union International Contre le Cancer (UICC) and the American Joint Committee on Cancer (AJCC). Cancer.
80(5)
992
- 993
1997.
DOI:
10.1002/(sici)1097-0142(19970901)80:5<992::aid-cncr26>3.0.co;2-q
33
Jonasch E
,
Futreal PA
,
Davis IJ
,
Bailey ST
,
Kim WY
,
Brugarolas J
,
Giaccia AJ
,
Kurban G
,
Pause A
,
Frydman J
,
Zurita AJ
,
Rini BI
,
Sharma P
,
Atkins MB
,
Walker CL
&
Rathmell WK
. State of the science: an update on renal cell carcinoma. Mol Cancer Res.
10(7)
859
- 880
2012.
DOI:
10.1158/1541-7786.MCR-12-0117
34
Sato Y
,
Yoshizato T
,
Shiraishi Y
,
Maekawa S
,
Okuno Y
,
Kamura T
,
Shimamura T
,
Sato-otsubo A
,
Nagae G
,
Suzuki H
,
Nagata Y
,
Yoshida K
,
Kon A
,
Suzuki Y
,
Chiba K
,
Tanaka H
,
Niida A
,
Fujimoto A
,
Tsunoda T
,
Morikawa T
,
Maeda D
,
Kume H
,
Sugano S
,
Fukayama M
,
Aburatani H
,
Sanada M
,
Miyano S
,
Homma Y
&
Ogawa S
. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet.
45(8)
860
- 867
2013.
DOI:
10.1038/ng.2699
35
Chen F
,
Zhang Y
,
Bossé D
,
Lalani AA
,
Hakimi AA
,
Hsieh JJ
,
Choueiri TK
,
Gibbons DL
,
Ittmann M
&
Creighton CJ
. Pan-urologic cancer genomic subtypes that transcend tissue of origin. Nat Commun.
8(1)
199
2017.
DOI:
10.1038/s41467-017-00289-x
36
Varela I
,
Tarpey P
,
Raine K
,
Huang D
,
Ong CK
,
Stephens P
,
Davies H
,
Jones D
,
Lin ML
,
Teague J
,
Bignell G
,
Butler A
,
Cho J
,
Dalgliesh GL
,
Galappaththige D
,
Greenman C
,
Hardy C
,
Jia M
,
Latimer C
,
Lau KW
,
Marshall J
,
McLaren S
,
Menzies A
,
Mudie L
,
Stebbings L
,
Largaespada DA
,
Wessels LF
,
Richard S
,
Kahnoski RJ
,
Anema J
,
Tuveson DA
,
Perez-Mancera PA
,
Mustonen V
,
Fischer A
,
Adams DJ
,
Rust A
,
Chan-on W
,
Subimerb C
,
Dykema K
,
Furge K
,
Campbell PJ
,
Teh BT
,
Stratton MR
&
Futreal PA
. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature.
469(7331)
539
- 542
2011.
DOI:
10.1038/nature09639
37
Cancer Genome Atlas Research Network
. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature.
499(7456)
43
- 49
2013.
DOI:
10.1038/nature12222
38
Gerlinger M
,
Horswell S
,
Larkin J
,
Rowan AJ
,
Salm MP
,
Varela I
,
Fisher R
,
McGranahan N
,
Matthews N
,
Santos CR
,
Martinez P
,
Phillimore B
,
Begum S
,
Rabinowitz A
,
Spencer-Dene B
,
Gulati S
,
Bates PA
,
Stamp G
,
Pickering L
,
Gore M
,
Nicol DL
,
Hazell S
,
Futreal PA
,
Stewart A
&
Swanton C
. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet.
46(3)
225
- 233
2014.
DOI:
10.1038/ng.2891
39
da Costa WH
,
da Cunha IW
,
Fares AF
,
Bezerra SM
,
Shultz L
,
Clavijo DA
,
da Silva DV
,
Netto GJ
,
Guimaraes GC
&
Cassio Zequi SD
. Prognostic impact of concomitant loss of PBRM1 and BAP1 protein expression in early stages of clear cell renal cell carcinoma. Urol Oncol Semin Orig Investig.
36(5)
243.e1
- 243.e8
2018.
DOI:
10.1016/J.UROLONC.2018.01.002
40
Hsieh JJ
,
Le V
,
Cao D
,
Cheng EH
&
Creighton CJ
. Genomic classifications of renal cell carcinoma: a critical step towards the future application of personalized kidney cancer care with pan-omics precision. J Pathol.
244(5)
525
- 537
2018.
DOI:
10.1002/PATH.5022
41
Nickerson ML
,
Jaeger E
,
Shi Y
,
Durocher JA
,
Mahurkar S
,
Zaridze D
,
Matveev V
,
Janout V
,
Kollarova H
,
Bencko V
,
Navratilova M
,
Szeszenia-Dabrowska N
,
Mates D
,
Mukeria A
,
Holcatova I
,
Schmidt LS
,
Toro JR
,
Karami S
,
Hung R
,
Gerard GF
,
Linehan WM
,
Merino M
,
Zbar B
,
Boffetta P
,
Brennan P
,
Rothman N
,
Chow WH
,
Waldman FM
&
Moore LE
. Improved identification of von Hippel-Lindau gene alterations in clear cell renal tumors. Clin Cancer Res.
14(15)
4726
- 4734
2008.
DOI:
10.1158/1078-0432.CCR-07-4921
42
Hossen MS
,
Samad A
,
Ahammad F
,
Sasa GBK
,
Jiang Z
&
Ding X
. System biology approaches identified novel biomarkers and their signaling pathways involved in renal cell carcinoma with different human diseases. Biosci Rep.
42(11)
BSR20221108
2022.
DOI:
10.1042/BSR20221108
43
Beroukhim R
,
Brunet JP
,
Di Napoli A
,
Mertz KD
,
Seeley A
,
Pires MM
,
Linhart D
,
Worrell RA
,
Moch H
,
Rubin MA
,
Sellers WR
,
Meyerson M
,
Linehan WM
,
Kaelin WG Jr
&
Signoretti S
. Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney. Cancer Res.
69(11)
4674
- 4681
2009.
DOI:
10.1158/0008-5472.CAN-09-0146
44
Brugarolas J
. Molecular genetics of clear-cell renal cell carcinoma. J Clin Oncol.
32(18)
1968
- 1976
2014.
DOI:
10.1200/JCO.2012.45.2003
45
Mehanna SH
,
Gurtat LB
,
Sprada LS
&
Mattei MPM
. Cancer and angiogenesis: the role of vascular endothelial growth factor (VEGF) and target therapy. Rev Méd Paraná.
79(1)
106
- 111
2021.
46
Latif F
,
Tory K
,
Gnarra J
,
Yao M
,
Duh F
,
Orcutt ML
,
Stackhouse T
,
Kuzmin I
,
Modi W
,
Geil L
,
Schmidt L
,
Zhou F
,
Li H
,
Wei MH
,
Chen F
,
Glenn G
,
Choyke P
,
Walther MM
,
Weng Y
,
Duan DR
,
Dean M
,
Glavač D
,
Richards FM
,
Crossey PA
,
Ferguson-Smith MA
,
Le Paslier D
,
Chumakov L
,
Cohen D
,
Chinault AC
,
Maher ER
,
Linehan WM
,
Zbar B
&
Lerman MI
. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science.
260(5112)
1317
- 1320
1993.
DOI:
10.1126/SCIENCE.8493574
48
Cancer Genome Atlas Research Network
,
Linehan WM
,
Spellman PT
,
Ricketts CJ
,
Creighton CJ
,
Fei SS
,
Davis C
,
Wheeler DA
,
Murray BA
,
Schmidt L
,
Vocke CD
,
Peto M
,
Al Mamun AA
,
Shinbrot E
,
Sethi A
,
Brooks S
,
Rathmell WK
,
Brooks AN
,
Hoadley KA
,
Robertson AG
,
Brooks D
,
Bowlby R
,
Sadeghi S
,
Shen H
,
Weisenberger DJ
,
Bootwalla M
,
Baylin SB
,
Laird PW
,
Cherniack AD
,
Saksena G
,
Haake S
,
Li J
,
Liang H
,
Lu Y
,
Mills GB
,
Akbani R
,
Leiserson MD
,
Raphael BJ
,
Anur P
,
Bottaro D
,
Albiges L
,
Barnabas N
,
Choueiri TK
,
Czerniak B
,
Godwin AK
,
Hakimi AA
,
Ho TH
,
Hsieh J
,
Ittmann M
,
Kim WY
,
Krishnan B
,
Merino MJ
,
Mills Shaw KR
,
Reuter VE
,
Reznik E
,
Shelley CS
,
Shuch B
,
Signoretti S
,
Srinivasan R
,
Tamboli P
,
Thomas G
,
Tickoo S
,
Burnett K
,
Crain D
,
Gardner J
,
Lau K
,
Mallery D
,
Morris S
,
Paulauskis JD
,
Penny RJ
,
Shelton C
,
Shelton WT
,
Sherman M
,
Thompson E
,
Yena P
,
Avedon MT
,
Bowen J
,
Gastier-Foster JM
,
Gerken M
,
Leraas KM
,
Lichtenberg TM
,
Ramirez NC
,
Santos T
,
Wise L
,
Zmuda E
,
Demchok JA
,
Felau I
,
Hutter CM
,
Sheth M
,
Sofia HJ
,
Tarnuzzer R
,
Wang Z
,
Yang L
,
Zenklusen JC
,
Zhang J
,
Ayala B
,
Baboud J
,
Chudamani S
,
Liu J
,
Lolla L
,
Naresh R
,
Pihl T
,
Sun Q
,
Wan Y
,
Wu Y
,
Ally A
,
Balasundaram M
,
Balu S
,
Beroukhim R
,
Bodenheimer T
,
Buhay C
,
Butterfield YS
,
Carlsen R
,
Carter SL
,
Chao H
,
Chuah E
,
Clarke A
,
Covington KR
,
Dahdouli M
,
Dewal N
,
Dhalla N
,
Doddapaneni HV
,
Drummond JA
,
Gabriel SB
,
Gibbs RA
,
Guin R
,
Hale W
,
Hawes A
,
Hayes DN
,
Holt RA
,
Hoyle AP
,
Jefferys SR
,
Jones SJ
,
Jones CD
,
Kalra D
,
Kovar C
,
Lewis L
,
Li J
,
Ma Y
,
Marra MA
,
Mayo M
,
Meng S
,
Meyerson M
,
Mieczkowski PA
,
Moore RA
,
Morton D
,
Mose LE
,
Mungall AJ
,
Muzny D
,
Parker JS
,
Perou CM
,
Roach J
,
Schein JE
,
Schumacher SE
,
Shi Y
,
Simons JV
,
Sipahimalani P
,
Skelly T
,
Soloway MG
,
Sougnez C
,
Tam A
,
Tan D
,
Thiessen N
,
Veluvolu U
,
Wang M
,
Wilkerson MD
,
Wong T
,
Wu J
,
Xi L
,
Zhou J
,
Bedford J
,
Chen F
,
Fu Y
,
Gerstein M
,
Haussler D
,
Kasaian K
,
Lai P
,
Ling S
,
Radenbaugh A
,
Van Den Berg D
,
Weinstein JN
,
Zhu J
,
Albert M
,
Alexopoulou I
,
Andersen JJ
,
Auman JT
,
Bartlett J
,
Bastacky S
,
Bergsten J
,
Blute ML
,
Boice L
,
Bollag RJ
,
Boyd J
,
Castle E
,
Chen YB
,
Cheville JC
,
Curley E
,
Davies B
,
DeVolk A
,
Dhir R
,
Dike L
,
Eckman J
,
Engel J
,
Harr J
,
Hrebinko R
,
Huang M
,
Huelsenbeck-Dill L
,
Iacocca M
,
Jacobs B
,
Lobis M
,
Maranchie JK
,
McMeekin S
,
Myers J
,
Nelson J
,
Parfitt J
,
Parwani A
,
Petrelli N
,
Rabeno B
,
Roy S
,
Salner AL
,
Slaton J
,
Stanton M
,
Thompson RH
,
Thorne L
,
Tucker K
,
Weinberger PM
,
Winemiller C
,
Zach LA
&
Zuna R
. Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med.
374(2)
135
- 145
2016.
DOI:
10.1056/NEJMoa1505917
49
Davis CF
,
Ricketts CJ
,
Wang M
,
Yang L
,
Cherniack AD
,
Shen H
,
Buhay C
,
Kang H
,
Kim SC
,
Fahey CC
,
Hacker KE
,
Bhanot G
,
Gordenin DA
,
Chu A
,
Gunaratne PH
,
Biehl M
,
Seth S
,
Kaipparettu BA
,
Bristow CA
,
Donehower LA
,
Wallen EM
,
Smith AB
,
Tickoo SK
,
Tamboli P
,
Reuter V
,
Schmidt LS
,
Hsieh JJ
,
Choueiri TK
,
Hakimi AA
,
TheCancer Genome Atlas Research Network
,
Chin L
,
Meyerson M
,
Kucherlapati R
,
Park WY
,
Robertson AG
,
Laird PW
,
Henske EP
,
Kwiatkowski DJ
,
Park PJ
,
Morgan M
,
Shuch B
,
Muzny D
,
Wheeler DA
,
Linehan WM
,
Gibbs RA
,
Rathmell WK
&
Creighton CJ
. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell.
26(3)
319
- 330
2014.
DOI:
10.1016/j.ccr.2014.07.014
50
Haas NB
&
Nathanson KL
. Hereditary kidney cancer syndromes. Adv Chronic Kidney Dis.
21(1)
81
- 90
2014.
DOI:
10.1053/j.ackd.2013.10.001
51
Frank I
,
Blute ML
,
Cheville JC
,
Lohse CM
,
Weaver AL
&
Zincke H
. Solid renal tumors: an analysis of pathological features related to tumor size. J Urol.
170(6)
2217
- 2220
2003.
DOI:
10.1097/01.JU.0000095475.12515.5E
52
Remzi M
,
Özsoy M
,
Klingler HC
,
Susani M
,
Waldert M
,
Seitz C
,
Schmidbauer J
&
Marberger M
. Are small renal tumors harmless? Analysis of histopathological features according to tumors 4 cm or less in diameter. J Urol.
176(3)
896
- 899
2006.
DOI:
10.1016/J.JURO.2006.04.047
53
Motzer RJ
,
Hutson TE
,
Cella D
,
Reeves J
,
Hawkins R
,
Guo J
,
Nathan P
,
Staehler M
,
De Souza P
,
Merchan JR
,
Boleti E
,
Fife K
,
Jin J
,
Jones R
,
Uemura H
,
De Giorgi U
,
Harmenberg U
,
Wang J
,
Sternberg CN
,
Deen K
,
Mccann L
,
Hackshaw MD
,
Crescenzo R
,
Pandite LN
&
Choueiri TK
. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med.
369(8)
722
- 731
2013.
DOI:
10.1056/NEJMOA1303989
55
Janzen NK
,
Kim HL
,
Figlin RA
&
Belldegrun AS
. Surveillance after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent disease. Urol Clin North Am.
30(4)
843
- 852
2003.
DOI:
10.1016/S0094-0143(03)00056-9
56
Chiong E
,
Tay MH
,
Tan MH
,
Kumar S
,
Sim HG
,
Teh BT
,
Umbas R
&
Chau NM
. Management of kidney cancer in Asia: resource-stratified guidelines from the Asian Oncology Summit 2012. Lancet Oncol.
13(11)
e482
- e491
2012.
DOI:
10.1016/S1470-2045(12)70433-3
57
Duran I
,
Lambea J
,
Maroto P
,
González-Larriba JL
,
Flores L
,
Granados-Principal S
,
Graupera M
,
Sáez B
,
Vivancos A
&
Casanovas O
. Resistance to targeted therapies in renal cancer: the importance of changing the mechanism of action. Target Oncol.
12(1)
19
- 35
2017.
DOI:
10.1007/s11523-016-0463-4
58
Atzpodien J
,
Schmitt E
,
Gertenbach U
,
Fornara P
,
Heynemann H
,
Maskow A
,
Ecke M
,
Wöltjen HH
,
Jentsch H
,
Wieland W
,
Wandert T
,
Reitz M
&
German Cooperative Renal Carcinoma Chemo-Immunotherapy Trials Group (DGCIN)
. Adjuvant treatment with interleukin-2- and interferon-alpha2a-based chemoimmunotherapy in renal cell carcinoma post tumour nephrectomy: results of a prospectively randomised trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN). Br J Cancer.
92(5)
843
- 846
2005.
DOI:
10.1038/sj.bjc.6602443
59
Singer EA
,
Gupta GN
,
Marchalik D
&
Srinivasan R
. Evolving therapeutic targets in renal cell carcinoma. Curr Opin Oncol.
25(3)
273
- 280
2013.
DOI:
10.1097/CCO.0B013E32835FC857
60
Reis AP dos
&
Machado JAN
. Imunoterapia no câncer-inibidores do checkpoint imunológico. Arq Asma Alerg e Imunol.
4
72
- 77
2020.
61
Sharma P
&
Allison JP
. The future of immune checkpoint therapy. Science.
348(6230)
56
- 61
2015.
DOI:
10.1126/SCIENCE.AAA8172
62
Topalian SL
,
Drake CG
&
Pardoll DM
. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell.
27(4)
450
- 461
2015.
DOI:
10.1016/j.ccell.2015.03.001
63
Pardoll DM
. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer.
12(4)
252
- 264
2012.
DOI:
10.1038/nrc3239
64
Özdemir BC
,
Siefker-Radtke AO
,
Campbell MT
&
Subudhi SK
. Current and future applications of novel immunotherapies in urological oncology: a critical review of the literature. Eur Urol Focus.
4(3)
442
- 454
2018.
DOI:
10.1016/j.euf.2017.10.001
65
Pal SK
,
Sonpavde G
,
Agarwal N
,
Vogelzang NJ
,
Srinivas S
,
Haas NB
,
Signoretti S
,
Mcgregor BA
,
Jones J
,
Lanman RB
,
Banks KC
&
Choueiri TK
. Evolution of circulating tumor DNA profile from first-line to subsequent therapy in metastatic renal cell carcinoma. Eur Urol.
72(4)
557
- 564
2017.
DOI:
10.1016/J.EURURO.2017.03.046
66
Bergerot PG
,
Hahn AW
,
Bergerot CD
,
Jones J
&
Pal SK
. The role of circulating tumor dna in renal cell carcinoma. Curr Treat Options Oncol.
19
1
- 11
2018.
DOI:
10.1007/S11864-018-0530-4/TABLES/1
67
Miao D
,
Margolis CA
,
Gao W
,
Voss MH
,
Li W
,
Martini DJ
,
Norton C
,
Bossé D
,
Wankowicz SM
,
Cullen D
,
Horak C
,
Wind-Rotolo M
,
Tracy A
,
Giannakis M
,
Hodi FS
,
Drake CG
,
Ball MW
,
Allaf ME
,
Snyder A
,
Hellmann MD
,
Ho T
,
Motzer RJ
,
Signoretti S
,
Kaelin WG Jr
,
Choueiri TK
&
Van Allen EM
. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science.
359(6377)
801
- 806
2018.
DOI:
10.1126/science.aan5951
68
Liu Y
,
Li Y
,
Wang Y
,
Lin C
,
Zhang D
,
Chen J
,
Ouyang L
,
Wu F
,
Zhang J
&
Chen L
. Recent progress on vascular endothelial growth factor receptor inhibitors with dual targeting capabilities for tumor therapy. J Hematol Oncol.
15(1)
89
2022.
DOI:
10.1186/s13045-022-01310-7
69
Schmidinger M
&
Bellmunt J
. Plethora of agents, plethora of targets, plethora of side effects in metastatic renal cell carcinoma. Cancer Treat Rev.
36(5)
416
- 424
2010.
DOI:
10.1016/j.ctrv.2010.01.003
70
Battelli C
&
Cho DC
. mTOR inhibitors in renal cell carcinoma. Therapy.
8(4)
359
- 367
2011.
DOI:
10.2217/thy.11.32
71
Therasse P
,
Arbuck SG
,
Eisenhauer EA
,
Wanders J
,
Kaplan RS
,
Rubinstein L
,
Verweij J
,
Van Glabbeke M
,
Van Oosterom AT
,
Christian MC
&
Gwyther SG
. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst.
92(3)
205
- 216
2000.
DOI:
10.1093/jnci/92.3.205
72
Makhov P
,
Joshi S
,
Ghatalia P
,
Kutikov A
,
Uzzo RG
&
Kolenko VM
. Resistance to systemic therapies in clear cell renal cell carcinoma: mechanisms and management strategies. Mol Cancer Ther.
17(7)
1355
- 1364
2018.
DOI:
10.1158/1535-7163.MCT-17-1299
73
Gobé G
,
Rubin M
,
Williams G
,
Sawczuk I
&
Buttyan R
. Apoptosis and expression of Bcl-2, Bcl-XL, and Bax in renal cell carcinomas. Cancer Invest.
20(3)
324
- 332
2002.
DOI:
10.1081/CNV-120001177
74
Ramp U
,
Dejosez M
,
Mahotka C
,
Czarnotta B
,
Kalinski T
,
Wenzel M
,
Lorenz I
,
Müller M
,
Krammer P
,
Gabbert HE
&
Gerharz CD
. Deficient activation of CD95 (APO-1/Fas)-mediated apoptosis: a potential factor of multidrug resistance in human renal cell carcinoma. Br J Cancer.
82(11)
1851
- 1859
2000.
DOI:
10.1054/bjoc.2000.1155
75
Bergers G
&
Hanahan D
. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer.
8(8)
592
- 603
2008.
DOI:
10.1038/nrc2442
76
Ishibashi K
,
Haber T
,
Breuksch I
,
Gebhard S
,
Sugino T
,
Kubo H
,
Hata J
,
Koguchi T
,
Yabe M
,
Kataoka M
,
Ogawa S
,
Hiraki H
,
Yanagida T
,
Haga N
,
Thüroff JW
,
Prawitt D
,
Brenner W
&
Kojima Y
. Overriding TKI resistance of renal cell carcinoma by combination therapy with IL-6 receptor blockade. Oncotarget.
8(33)
55230
- 55245
2017.
DOI:
10.18632/oncotarget.19420
77
Park K
,
Lee JL
,
Park I
,
Park S
,
Ahn Y
,
Ahn JH
,
Ahn S
,
Song C
,
Hong JH
,
Kim CS
&
Ahn H
. Comparative efficacy of vascular endothelial growth factor (VEGF) tyrosine kinase inhibitor (TKI) and mammalian target of rapamycin (mTOR) inhibitor as second-line therapy in patients with metastatic renal cell carcinoma after the failure of first-line VEGF TKI. Med Oncol.
29(5)
3291
- 3297
2012.
DOI:
10.1007/s12032-012-0227-7
78
Gottesman MM
,
Fojo T
&
Bates SE
. Multidrug resistance in cancer: role of ATP–dependent transporters. Nat Rev.
2(1)
48
- 58
2002.
DOI:
10.1038/nrc706
79
Gotink KJ
,
Broxterman HJ
,
Labots M
,
de Haas RR
,
Dekker H
,
Honeywell RJ
,
Rudek MA
,
Beerepoot LV
,
Musters RJ
,
Jansen G
,
Griffioen AW
,
Assaraf YG
,
Pili R
,
Peters GJ
&
Verheul HM
. Lysosomal sequestration of sunitinib: a novel mechanism of drug resistance. Clin Cancer Res.
17(23)
7337
- 7346
2011.
DOI:
10.1158/1078-0432.CCR-11-1667
80
Bielecka ZF
,
Czarnecka AM
,
Solarek W
,
Kornakiewicz A
&
Szczylik C
. Mechanisms of acquired resistance to tyrosine kinase inhibitors in clear – cell renal cell carcinoma (ccRCC). Curr Signal Transduct Ther.
8(3)
218
- 228
2014.
DOI:
10.2174/1574362409666140206223014
81
Gotink KJ
,
Broxterman HJ
,
Labots M
,
de Haas RR
,
Dekker H
,
Honeywell RJ
,
Rudek MA
,
Beerepoot LV
,
Musters RJ
,
Jansen G
,
Griffioen AW
,
Assaraf YG
,
Pili R
,
Peters GJ
&
Verheul HM
. Lysosomal sequestration of sunitinib: a novel mechanism of drug resistance. Clin Cancer Res.
17(23)
7337
- 7346
2011.
DOI:
10.1158/1078-0432.CCR-11-1667
82
Choueiri TK
,
Halabi S
,
Sanford BL
,
Hahn O
,
Michaelson MD
,
Walsh MK
,
Feldman DR
,
Olencki T
,
Picus J
,
Small EJ
,
Dakhil S
,
George DJ
&
Morris MJ
. Cabozantinib versus sunitinib as initial targeted therapy for patients with metastatic renal cell carcinoma of poor or intermediate risk: the Alliance A031203 CABOSUN trial. J Clin Oncol.
35(6)
591
- 597
2017.
DOI:
10.1200/JCO.2016.70.7398
83
Sternberg CN
,
Davis ID
,
Mardiak J
,
Szczylik C
,
Lee E
,
Wagstaff J
,
Barrios CH
,
Salman P
,
Gladkov OA
,
Kavina A
,
Zarbá JJ
,
Chen M
,
Mccann L
,
Pandite L
,
Roychowdhury DF
&
Hawkins RE
. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized Phase III trial. J Clin Oncol.
41(11)
1957
- 1964
2023.
DOI:
10.1200/JCO.22.02622
84
Jin J
,
Xie Y
,
Zhang J-S
,
Wang J-Q
,
Dai S-J
,
He W
,
Li S-Y
,
Ashby CR
,
Chen Z-S
&
He Q
. Sunitinib resistance in renal cell carcinoma: From molecular mechanisms to predictive biomarkers. Drug Resist Updat.
67
100929
2023.
DOI:
10.1016/J.DRUP.2023.100929
85
Ciamporcero E
,
Miles KM
,
Adelaiye R
,
Ramakrishnan S
,
Shen L
,
Ku S
,
Pizzimenti S
,
Sennino B
,
Barrera G
&
Pili R
. Combination strategy targeting VEGF and HGF/c-met in human renal cell carcinoma models. Mol Cancer Ther.
14(1)
101
- 110
2015.
DOI:
10.1158/1535-7163.MCT-14-0094
86
Zhang Y
,
Kwok-Shing Ng P
,
Kucherlapati M
,
Chen F
,
Liu Y
,
Tsang YH
,
de Velasco G
,
Jeong KJ
,
Akbani R
,
Hadjipanayis A
,
Pantazi A
,
Bristow CA
,
Lee E
,
Mahadeshwar HS
,
Tang J
,
Zhang J
,
Yang L
,
Seth S
,
Lee S
,
Ren X
,
Song X
,
Sun H
,
Seidman J
,
Luquette LJ
,
Xi R
,
Chin L
,
Protopopov A
,
Westbrook TF
,
Shelley CS
,
Choueiri TK
,
Ittmann M
,
Van Waes C
,
Weinstein JN
,
Liang H
,
Henske EP
,
Godwin AK
,
Park PJ
,
Kucherlapati R
,
Scott KL
,
Mills GB
,
Kwiatkowski DJ
&
Creighton CJ
. A pan-cancer proteogenomic Atlas of PI3K/AKT/mTOR pathway alterations. Cancer Cell.
31(6)
820
- 832.e3
2017.
DOI:
10.1016/j.ccell.2017.04.013
87
Gordan JD
,
Lal P
,
Dondeti VR
,
Letrero R
,
Parekh KN
,
Oquendo CE
,
Greenberg RA
,
Flaherty KT
,
Rathmell WK
,
Keith B
,
Simon MC
&
Nathanson KL
. HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma. Cancer Cell.
14(6)
435
- 446
2008.
DOI:
10.1016/j.ccr.2008.10.016
88
Makhov PB
,
Golovine K
,
Kutikov A
,
Teper E
,
Canter DJ
,
Simhan J
,
Uzzo RG
&
Kolenko VM
. Modulation of Akt/mTOR signaling overcomes sunitinib resistance in renal and prostate cancer cells. Mol Cancer Ther.
11(7)
1510
- 1517
2012.
DOI:
10.1158/1535-7163.MCT-11-0907
89
Tsavachidou-Fenner D
,
Tannir N
,
Tamboli P
,
Liu W
,
Petillo D
,
Teh B
,
Mills GB
&
Jonasch E
. Gene and protein expression markers of response to combined antiangiogenic and epidermal growth factor targeted therapy in renal cell carcinoma. Ann Oncol.
21(8)
1599
- 1606
2010.
DOI:
10.1093/annonc/mdp600
90
Sarkadi B
,
Homolya L
,
Szakács G
&
Váradi A
. Human multidrug resistance ABCB and ABCG transporters: Participation in a chemoimmunity defense system. Physiol Rev.
86(4)
1179
- 1236
2006.
DOI:
10.1152/PHYSREV.00037.2005
91
Adelaiye R
,
Ciamporcero E
,
Miles KM
,
Sotomayor P
,
Bard J
,
Tsompana M
,
Conroy D
,
Shen L
,
Ramakrishnan S
,
Ku SY
,
Orillion A
,
Prey J
,
Fetterly G
,
Buck M
,
Chintala S
,
Bjarnason GA
&
Pili R
. Sunitinib dose escalation overcomes transient resistance in clear cell renal cell carcinoma and is associated with epigenetic modifications. Mol Cancer Ther.
14(2)
513
- 522
2015.
DOI:
10.1158/1535-7163.MCT-14-0208
92
Dietz S
,
Sültmann H
,
Du Y
,
Reisinger E
,
Riediger AL
,
Volckmar AL
,
Stenzinger A
,
Schlesner M
,
Jäger D
,
Hohenfellner M
,
Duensing S
,
Grüllich C
&
Pahernik S
. Patient-specific molecular alterations are associated with metastatic clear cell renal cell cancer progressing under tyrosine kinase inhibitor therapy. Oncotarget.
8(43)
74049
- 74057
2017.
DOI:
10.18632/oncotarget.18200
93
Stewart GD
,
O’Mahony FC
,
Laird A
,
Eory L
,
Lubbock AL
,
Mackay A
,
Nanda J
,
O’Donnell M
,
Mullen P
,
Mcneill SA
,
Riddick AC
,
Berney D
,
Bex A
,
Aitchison M
,
Overton IM
,
Harrison DJ
&
Powles T
. Sunitinib treatment exacerbates intratumoral heterogeneity in metastatic renal cancer. Clin Cancer Res.
21(18)
4212
- 4223
2015.
DOI:
10.1158/1078-0432.CCR-15-0207
94
Shojaei F
,
Wu X
,
Qu X
,
Kowanetz M
,
Yu L
,
Tan M
,
Meng YG
&
Ferrara N
. G-CSF-initiated myeloid cell mobilization and angiogenesis mediate tumor refractoriness to anti-VEGF therapy in mouse models. Proc Natl Acad Sci USA.
106(16)
6742
- 6747
2009.
DOI:
10.1073/pnas.0902280106
95
Ko JS
,
Rayman P
,
Ireland J
,
Swaidani S
,
Li G
,
Bunting KD
,
Rini B
,
Finke JH
&
Cohen PA
. Direct and differential suppression of myeloid-derived suppressor cell subsets by sunitinib is compartmentally constrained. Cancer Res.
70(9)
3526
- 3536
2010.
DOI:
10.1158/0008-5472.CAN-09-3278
96
Prior C
,
Perez-Gracia JL
,
Garcia-Donas J
,
Rodriguez-Antona C
,
Guruceaga E
,
Esteban E
,
Suarez C
,
Castellano D
,
del Alba AG
,
Lozano MD
,
Carles J
,
Climent MA
,
Arranz JA
,
Gallardo E
,
Puente J
,
Bellmunt J
,
Gurpide A
,
Lopez-Picazo JM
,
Hernandez AG
,
Mellado B
,
Martínez E
,
Moreno F
,
Font A
&
Calvo A
. Identification of tissue microRNAs predictive of sunitinib activity in patients with metastatic renal cell carcinoma. PLoS One.
9(1)
e86263
2014.
DOI:
10.1371/journal.pone.0086263
97
Siegel RL
,
Miller KD
&
Jemal A
. Cancer statistics, 2015. CA Cancer J Clin.
65(1)
5
- 29
2015.
DOI:
10.3322/CAAC.21254
98
Prins FM
,
Kerkmeijer LG
,
Pronk AA
,
Vonken EP
,
Meijer RP
,
Bex A
&
Barendrecht MM
. Renal cell carcinoma: Alternative nephron-sparing treatment options for small renal masses, a systematic review. J Endourol.
31(10)
963
- 975
2017.
DOI:
10.1089/end.2017.0382
99
Gerlinger M
,
Catto JW
,
Orntoft TF
,
Real FX
,
Zwarthoff EC
&
Swanton C
. Intratumour heterogeneity in urologic cancers: from molecular evidence to clinical implications. Eur Urol.
67(4)
729
- 737
2015.
DOI:
10.1016/J.EURURO.2014.04.014
100
Pantuck AJ
,
Zisman A
&
Belldegrun AS
. The changing natural history of renal cell carcinoma. J Urol.
166
1611
- 1623
2001.
DOI:
10.1016/s0022-5347(05)65640-6
101
Rohrmann K
,
Staehler M
,
Haseke N
,
Bachmann A
,
Stief CG
&
Siebels M
. Immunotherapy in metastatic renal cell carcinoma. World J Urol.
23(3)
196
- 201
2005.
DOI:
10.1007/s00345-004-0470-4
102
Linehan WM
,
Rubin JS
&
Bottaro DP
. VHL loss of function and its impact on oncogenic signaling networks in clear cell renal cell carcinoma. Int J Biochem Cell Biol.
41(4)
753
- 756
2009.
DOI:
10.1016/j.biocel.2008.09.024
104
Samarghandian S
,
Afshari JT
&
Davoodi S
. Honey induces apoptosis in renal cell carcinoma. Pharmacogn Mag.
7(25)
46
- 52
2011.
DOI:
10.4103/0973-1296.75901
105
Cohen HT
&
McGovern FJ
. Renal-cell carcinoma. N Engl J Med.
353(23)
2477
- 2490
2005.
DOI:
10.1056/NEJMra043172
106
Voss MH
,
Hsieh JJ
&
Motzer RJ
. Novel approaches targeting the vascular endothelial growth factor axis in renal cell carcinoma. Cacner J.
19(4)
299
- 306
2013.
DOI:
10.1097/PPO.0b013e31829d5cff
107
Faivre S
,
Kroemer G
&
Raymond E
. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov.
5(8)
671
- 688
2006.
DOI:
10.1038/nrd2062
108
Sabatini DM
. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer.
6(9)
729
- 734
2006.
DOI:
10.1038/nrc1974
109
Cancer Genome Atlas Research Network
. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature.
499(7456)
43
- 49
2013.
DOI:
10.1038/nature12222
110
Hamid O
&
Carvajal RD
. Anti-programmed death-1 and anti-programmed death-ligand 1 antibodies in cancer therapy. Expert Opin Biol Ther.
13(6)
847
- 861
2013.
DOI:
10.1517/14712598.2013.770836
111
Motzer RJ
,
Escudier B
,
McDermott DF
,
George S
,
Hammers HJ
,
Srinivas S
,
Tykodi SS
,
Sosman JA
,
Procopio G
,
Plimack ER
,
Castellano D
,
Choueiri TK
,
Gurney H
,
Donskov F
,
Bono P
,
Wagstaff J
,
Gauler TC
,
Ueda T
,
Tomita Y
,
Schutz FA
,
Kollmannsberger C
,
Larkin J
,
Ravaud A
,
Simon JS
,
Xu LA
,
Waxman IM
,
Sharma P
&
CheckMate 025 Investigators
. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med.
373(19)
1803
- 1813
2015.
DOI:
10.1056/NEJMoa1510665
112
Perrone D
,
Fuggetta MP
,
Ardito F
,
Cottarelli A
,
De Filippis A
,
Ravagnan G
,
De Maria S
&
Lo Muzio L
. Resveratrol (3,5,4’-trihydroxystilbene) and its properties in oral diseases. Exp Ther Med.
14(1)
3
- 9
2017.
DOI:
10.3892/etm.2017.4472
113
Motzer RJ
,
Hutson TE
,
Tomczak P
,
Michaelson MD
,
Bukowski RM
,
Rixe O
,
Oudard S
,
Negrier S
,
Szczylik C
,
Kim ST
,
Chen I
,
Bycott PW
,
Baum CM
&
Figlin RA
. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med.
356(2)
115
- 124
2007.
DOI:
10.1056/NEJMoa065044
114
Rini BI
,
Escudier B
,
Tomczak P
,
Kaprin A
,
Szczylik C
,
Hutson TE
,
Michaelson MD
,
Gorbunova VA
,
Gore ME
,
Rusakov IG
,
Negrier S
,
Ou Y
,
Castellano D
,
Lim HY
,
Uemura H
,
Tarazi J
,
Cella D
,
Chen C
,
Rosbrook B
,
Kim S
&
Motzer RJ
. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet.
378(9807)
1931
- 1939
2011.
DOI:
10.1016/S0140-6736(11)61613-9
115
Choueiri TK
,
Escudier B
,
Powles T
,
Mainwaring PN
,
Rini BI
,
Donskov F
,
Hammers H
,
Hutson TE
,
Lee JL
,
Peltola K
,
Roth BJ
,
Bjarnason GA
,
Géczi L
,
Keam B
,
Maroto P
,
Heng DY
,
Schmidinger M
,
Kantoff PW
,
Borgman-Hagey A
,
Hessel C
,
Scheffold C
,
Schwab GM
,
Tannir NM
,
Motzer RJ
&
METEOR Investigators
. Cabozantinib versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med.
373(19)
1814
- 1823
2015.
DOI:
10.1056/NEJMoa1510016
116
Eisen T
,
Joensuu H
,
Nathan PD
,
Harper PG
,
Wojtukiewicz MZ
,
Nicholson S
,
Bahl A
,
Tomczak P
,
Pyrhonen S
,
Fife K
,
Bono P
,
Boxall J
,
Wagner A
,
Jeffers M
,
Lin T
&
Quinn DI
. Regorafenib for patients with previously untreated metastatic or unresectable renal-cell carcinoma: a single-group phase 2 trial. Lancet Oncol.
13(10)
1055
- 1062
2012.
DOI:
10.1016/S1470-2045(12)70364-9
117
Mulders P
,
Hawkins R
,
Nathan P
,
De Jong I
,
Osanto S
,
Porfiri E
,
Protheroe A
,
Van Herpen CM
,
Mookerjee B
,
Pike L
,
Jürgensmeier JM
&
Gore ME
. Cediranib monotherapy in patients with advanced renal cell carcinoma: Results of a randomised phase II study. Eur J Cancer.
48(4)
527
- 537
2012.
DOI:
10.1016/j.ejca.2011.12.022
118
Motzer RJ
,
Nosov D
,
Eisen T
,
Bondarenko I
,
Lesovoy V
,
Lipatov O
,
Tomczak P
,
Lyulko O
,
Alyasova A
,
Harza M
,
Kogan M
,
Alekseev BY
,
Sternberg CN
,
Szczylik C
,
Cella D
,
Ivanescu C
,
Krivoshik A
,
Strahs A
,
Esteves B
,
Berkenblit A
&
Hutson TE
. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J Clin Oncol.
31(30)
3791
- 3799
2013.
DOI:
10.1200/JCO.2012.47.4940
119
Motzer RJ
,
Porta C
,
Vogelzang NJ
,
Sternberg CN
,
Szczylik C
,
Zolnierek J
,
Kollmannsberger C
,
Rha SY
,
Bjarnason GA
,
Melichar B
,
De Giorgi U
,
Grünwald V
,
Davis ID
,
Lee JL
,
Esteban E
,
Urbanowitz G
,
Cai C
,
Squires M
,
Marker M
,
Shi MM
&
Escudier B
. Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial. Lancet Oncol.
15(3)
286
- 296
2014.
DOI:
10.1016/S1470-2045(14)70030-0
120
Hsieh JJ
,
Purdue MP
,
Signoretti S
,
Swanton C
,
Albiges L
,
Schmidinger M
,
Heng DY
,
Larkin J
&
Ficarra V
. Renal cell carcinoma. Nat Rev Dis Primers.
3
17009
2017.
DOI:
10.1038/nrdp.2017.9
121
Bayda S
,
Adeel M
,
Tuccinardi T
,
Cordani M
&
Rizzolio F
. The history of nanoscience and nanotechnology: from chemical-physical applications to nanomedicine. Molecules.
25(1)
112
2019.
DOI:
10.3390/molecules25010112
122
Salles TDR
,
Rodrigues HDB
,
Bruckmann FDS
,
Alves LCS
,
Mortari SR
&
Rhoden CRB
. Graphene oxide optimization synthesis for application on laboratory of Universidade Franciscana. Discip Sci.
21(3)
15
- 26
2020.
DOI:
10.37779/nt.v21i3.3632
123
Bruckmann FDS
,
Nunes FB
,
Salles TDR
,
Franco C
,
Cadoná FC
&
Bohn Rhoden CR
. Biological applications of silica-based nanoparticles. Magnetochemistry.
8(10)
131
2022.
DOI:
10.3390/magnetochemistry8100131
124
Mohanapandian K
,
Kamala SSP
,
Periasamy P
,
Priya NS
,
Selvakumar B
&
Senthilkannan K
. Cu2+ substituted Cr2O3 nanostructures prepared by microwave-assisted method: an investigation of its structural, morphological, optical, and dielectric properties. J Sol-Gel Sci Technol.
99(3)
546
- 556
2021.
DOI:
10.1007/S10971-021-05596-W
125
Nunes FB
,
Da Silva Bruckmann F
,
Da Rosa Salles T
&
Rhoden CRB
. Study of phenobarbital removal from the aqueous solutions employing magnetite-functionalized chitosan. Environ Sci Pollut Res.
30(5)
12658
- 12671
2022.
DOI:
10.1007/s11356-022-23075-9
126
Rhoden CRB
,
da Silva Bruckmann F
,
da Rosa Salles T
,
Junior CGK
&
Mortari SR
. Study from the influence of magnetite onto removal of hydrochlorothiazide from aqueous solutions applying magnetic graphene oxide. J Water Process Eng.
43
102262
2021.
DOI:
10.1016/J.JWPE.2021.102262
127
Bruckmann FS
,
Schnorr C
,
Oviedo LR
,
Knani S
,
Silva LFO
,
Silva WL
,
Dotto GL
&
Bohn Rhoden CR
. Adsorption and photocatalytic degradation of pesticides into nanocomposites: a review. Molecules.
27(19)
6261
2022.
DOI:
10.3390/molecules27196261
128
de Oliveira ÉC
,
da Silva Bruckmann F
,
Schopf PF
,
Viana AR
,
Mortari SR
,
Sagrillo MR
,
de Vasconcellos NJS
,
da Silva Fernandes L
&
Rhoden CRB
. In vitro and in vivo safety profile assessment of graphene oxide decorated with different concentrations of magnetite. J Nanoparticle Res.
24
150
2022.
DOI:
10.1007/S11051-022-05529-W
129
da Silva Bruckmann F
,
Viana AR
,
Lopes LQS
,
Santos RCV
,
Muller EI
,
Mortari SR
&
Rhoden CRB
. Synthesis, characterization, and biological activity evaluation of magnetite-functionalized eugenol. J Inorg Organomet Polym Mater.
32(4)
1459
- 1472
2022.
DOI:
10.1007/s10904-021-02207-7
130
Liu G
,
Lu M
,
Huang X
,
Li T
&
Xu D
. Application of gold-nanoparticle colorimetric sensing to rapid food safety screening. Sensors (Basel).
18(12)
4166
2018.
DOI:
10.3390/s18124166
131
Fortuni B
,
Inose T
,
Ricci M
,
Fujita Y
,
Van Zundert I
,
Masuhara A
,
Fron E
,
Mizuno H
,
Latterini L
,
Rocha S
&
Uji-I H
. Polymeric engineering of nanoparticles for highly efficient multifunctional drug delivery systems. Sci Rep.
9(1)
2666
2019.
DOI:
10.1038/s41598-019-39107-3
132
Bruckmann FDS
,
Rossato Viana A
,
Tonel MZ
,
Fagan SB
,
Garcia WJDS
,
Oliveira AHD
,
Dorneles LS
,
Roberto Mortari S
,
Silva WLD
,
Silva IZD
&
Rhoden CRB
. Influence of magnetite incorporation into chitosan on the adsorption of the methotrexate and in vitro cytotoxicity. Environ Sci Pollut Res Int.
29(46)
70413
- 70434
2022.
DOI:
10.1007/s11356-022-20786-x
133
Gavas S
,
Quazi S
&
Karpiński TM
. Nanoparticles for cancer therapy: Current progress and challenges. Nanoscale Res Lett.
16(1)
173
2021.
DOI:
10.1186/s11671-021-03628-6
134
Palazzolo S
,
Bayda S
,
Hadla M
,
Caligiuri I
,
Corona G
,
Toffoli G
&
Rizzolio F
. The clinical translation of organic nanomaterials for cancer therapy: a focus on polymeric nanoparticles, micelles, liposomes and exosomes. Curr Med Chem.
25(34)
4224
- 4268
2018.
DOI:
10.2174/0929867324666170830113755
135
Chen Y
,
Wang M
,
Zhang T
,
Du E
,
Liu Y
,
Qi S
,
Xu Y
&
Zhang Z
. Autophagic effects and mechanisms of silver nanoparticles in renal cells under low dose exposure. Ecotoxicol Environ Saf.
166
71
- 77
2018.
DOI:
10.1016/J.ECOENV.2018.09.070
136
Rezvantalab S
,
Drude NI
,
Moraveji MK
,
Güvener N
,
Koons EK
,
Shi Y
,
Lammers T
&
Kiessling F
. PLGA-based nanoparticles in cancer treatment. Front Pharmacol.
9
1260
2018.
DOI:
10.3389/fphar.2018.01260
137
Xie D
,
Mao Y
,
Du N
,
Ji H
&
Li J
. Macrophages promote growth, migration and epithelial-mesenchymal transition of renal cell carcinoma by regulating GSDMD/IL-1β axis. Cytokine.
159
156021
2022.
DOI:
10.1016/J.CYTO.2022.156021
138
Zheng J
,
Karmakar B
,
El-Kott AF
,
Elsaid FG
,
Shati AA
,
Negm S
,
Alsayegh AA
&
El-Saber Batiha G
. Characterization and cytotoxicity and antihuman renal cell carcinoma potentials of starch capped-copper oxide nanoparticles synthesized by ultrasonic irradiation: Introducing a novel chemotherapeutic drug. J Saudi Chem Soc.
26(6)
101543
2022.
DOI:
10.1016/J.JSCS.2022.101543
139
da Rosa Salles T
,
da Silva Bruckamann F
,
Viana AR
,
Krause LMF
,
Mortari SR
&
Rhoden CRB
. Magnetic nanocrystalline cellulose: Azithromycin adsorption and in vitro biological activity against melanoma cells. J Polym Environ.
30(7)
2695
- 2713
2022.
DOI:
10.1007/S10924-022-02388-3
140
Abbas G
,
Singh KB
,
Kumar N
,
Shukla A
,
Kumar D
&
Pandey G
. Efficient anticarcinogenic activity of α-Fe2O3 nanoparticles: In-vitro and computational study on human renal carcinoma cells HEK-293. Mater Today Commun.
26
102175
2021.
DOI:
10.1016/J.MTCOMM.2021.102175
141
Huber DL
. Synthesis, properties, and applications of iron nanoparticles. Small.
1(5)
482
- 501
2005.
DOI:
10.1002/SMLL.200500006
142
Nagajyothi PC
,
Pandurangan M
,
Kim DH
,
Sreekanth TVM
&
Shim J
. Green synthesis of iron oxide nanoparticles and their catalytic and in vitro anticancer activities. J Clust Sci.
28(1)
245
- 257
2017.
DOI:
10.1007/s10876-016-1082-z
143
Bruckmann FDS
,
Pimentel AC
,
Viana AR
,
Salles TDR
,
Krause LMF
,
Mortari SR
,
Silva IZD
&
Rhoden CRB
. Synthesis, characterization and cytotoxicity evaluation of magnetic nanosilica in L929 cell line. Discip Sci.
21(3)
01
- 14
2020.
DOI:
10.37779/nt.v21i3.3631
144
Zhang XF
,
Liu ZG
,
Shen W
&
Gurunathan S
. Silver nanoparticles: Synthesis, characterization, properties, applications, and therapeutic approaches. Int J Mol Sci.
17(9)
1534
2016.
DOI:
10.3390/ijms17091534
145
Shi M
,
Wang S
,
Zheng S
,
Hou P
,
Dong L
,
He M
,
Wu C
,
Zhang X
,
Zuo F
,
Xu K
&
Li J
. Activatable MRI-monitoring gene delivery for the theranostic of renal carcinoma. Colloids Surfaces B Biointerfaces.
185
110625
2020.
DOI:
10.1016/J.COLSURFB.2019.110625
146
Chai D
,
Liu N
,
Li H
,
Wang G
,
Song J
,
Fang L
,
Lu Z
,
Yao H
&
Zheng J
. H1/pAIM2 nanoparticles exert anti-tumour effects that is associated with the inflammasome activation in renal carcinoma. J Cell Mol Med.
22(11)
5670
- 5681
2018.
DOI:
10.1111/jcmm.13842
147
Soliman MS
,
Moin A
,
Hussain T
,
Gowda D V
,
Dixit SR
&
Lila ASA
. Development and optimization of dual drug-loaded nanoparticles for the potent anticancer effect on renal carcinoma. J Drug Deliv Sci Technol.
59
101846
2020.
DOI:
10.1016/J.JDDST.2020.101846
148
Ying S
,
Guan Z
,
Ofoegbu PC
,
Clubb P
,
Rico C
,
He F
&
Hong J
. Green synthesis of nanoparticles: Current developments and limitations. Environ Technol Innov.
26
102336
2022.
DOI:
10.1016/J.ETI.2022.102336
149
Rónavári A
,
Igaz N
,
Adamecz DI
,
Szerencsés B
,
Molnar C
,
Kónya Z
,
Pfeiffer I
&
Kiricsi M
. Green silver and gold nanoparticles: Biological synthesis approaches and potentials for biomedical applications. Molecules.
26(4)
844
2021.
DOI:
10.3390/molecules26040844
150
Liu R
,
Pei Q
,
Shou T
,
Zhang W
,
Hu J
&
Li W
. Apoptotic effect of green synthesized gold nanoparticles from Curcuma wenyujin extract against human renal cell carcinoma A498 cells. Int J Nanomedicine.
14
4091
- 4103
2019.
DOI:
10.2147/IJN.S203222
151
Li X
,
Tang G
,
Guo X
&
Men T
. Characterization and apoptotic effect of copper nanoparticles biosynthesized from Ziziphus zizyphus leaf on human renal cell carcinoma A498 cells. Appl Nanosci.
11(1)
139
- 148
2021.
DOI:
10.1007/s13204-020-01570-0
152
Khalafi T
,
Buazar F
&
Ghanemi K
. Phycosynthesis and enhanced photocatalytic activity of zinc oxide nanoparticles toward organosulfur pollutants. Sci Rep.
9(1)
6866
2019.
DOI:
10.1038/s41598-019-43368-3
153
Lokapur V
,
Jayakar V
,
M.S D
,
Chalannavar RK
,
Lasrado L
&
Shantaram M
. ZnO nanoparticles with spectroscopically controlled morphology, bioinspired from Holigarna grahamii (Wight) Kurz and delving its antioxidant and anticancer potential on A498 cell line. Mater Today Commun.
31
103338
2022.
DOI:
10.1016/j.mtcomm.2022.103338
154
Zhou Z
&
Chen X
. Precise Engineering of cisplatin prodrug into supramolecular nanoparticles: Enhanced on in vitro antiproliferative activity and treatment and care of in vivo renal injury. .
DOI:
10.21203/rs.3.rs-254827/v1