Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
J Exp Clin Cancer Res ; 43(1): 242, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39169402

ABSTRACT

Renal cell carcinoma (RCC) is one of the most common tumors that afflicts the urinary system, accounting for 90-95% of kidney cancer cases. Although its incidence has increased over the past decades, its pathogenesis is still unclear. Tumor-associated macrophages (TAMs) are the most prominent immune cells in the tumor microenvironment (TME), comprising more than 50% of the tumor volume. By interacting with cancer cells, TAMs can be polarized into two distinct phenotypes, M1-type and M2-type TAMs. In the TME, M2-type TAMs, which are known to promote tumorigenesis, are more abundant than M1-type TAMs, which are known to suppress tumor growth. This ratio of M1 to M2 TAMs can create an immunosuppressive environment that contributes to tumor cell progression and survival. This review focused on the role of TAMs in RCC, including their polarization, impacts on tumor proliferation, angiogenesis, invasion, migration, drug resistance, and immunosuppression. In addition, we discussed the potential of targeting TAMs for clinical therapy in RCC. A deeper understanding of the molecular biology of TAMs is essential for exploring innovative therapeutic strategies for the treatment of RCC.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Tumor-Associated Macrophages , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Tumor Microenvironment/immunology
2.
Technol Cancer Res Treat ; 23: 15330338241275403, 2024.
Article in English | MEDLINE | ID: mdl-39149973

ABSTRACT

Early diagnosis is crucial for enhancing the survival rate of renal cell cancer patients, and exosomes present potential advantages in this area. Their small size, high mobility, and lipid bilayer structure enable exosomes to cross biological membranes easily, protecting the bioactive cargo within from degradation. Exosomes significantly influence the invasion and metastasis of RCC, and they also contribute to tumor drug resistance and immune evasion.


Subject(s)
Carcinoma, Renal Cell , Exosomes , Kidney Neoplasms , Humans , Exosomes/metabolism , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/diagnosis , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Biomarkers, Tumor , Nanoparticles/chemistry , Drug Delivery Systems , Drug Carriers/chemistry , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
3.
BMC Med Genomics ; 17(1): 215, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160519

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a rare autosomal dominant syndrome caused by a germline mutation in the fumarate hydratase (FH) gene that manifests with cutaneous leiomyomas, uterine fibroids, and renal cell cancer (RCC). Patients with HLRCC-associated RCC (HLRCC-RCC) have aggressive clinical courses, but there is no standardized therapy for advanced HLRCC-RCC. In this study, we described a case of aggressive HLRCC in a 33-year-old female who exhibited a novel heterozygous germline insertion mutation in exon 8 of the FH gene (c.1126 C > T; p.Q376*). The patient underwent laparoscopic resection of the right kidney, but metastases appeared within 3 months after surgery. Histological staining of the resected tumor revealed high expression levels of programmed cell death-ligand 1 (PD-L1). Therefore, the patient was treated with immunotherapy. The patient achieved a partial response to immunotherapy, and the treatment of metastatic lesions has continued to improve. A thorough literature review pinpointed 76 historical cases of HLRCC-RCC that had undergone immunotherapy. From this pool, 46 patients were selected for this study to scrutinize the association between mutations in the FH gene and the effectiveness of immunotherapy. Our results indicate that immunotherapy could significantly improve the overall survival (OS) of patients with HLRCC-RCC. However, no influence of different mutations in the FH germline gene on the therapeutic efficacy of immunotherapy was observed. Therefore, our study suggested that immunotherapy was an effective therapeutic option for patients with HLRCC regardless of the type of FH germline mutation.


Subject(s)
Fumarate Hydratase , Immunotherapy , Leiomyomatosis , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms , Humans , Female , Leiomyomatosis/genetics , Leiomyomatosis/pathology , Leiomyomatosis/therapy , Fumarate Hydratase/genetics , Adult , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Neoplastic Syndromes, Hereditary/complications , Neoplastic Syndromes, Hereditary/pathology , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Uterine Neoplasms/therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Germ-Line Mutation , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy
4.
FP Essent ; 543: 12-17, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39163010

ABSTRACT

Kidney cysts and tumors often are identified during imaging for unrelated issues. Kidney cysts can be attributable to heritable polycystic kidney diseases. These cysts are rare in children. In adults, they affect approximately 50% of individuals older than 50 years. Kidney cysts are categorized on imaging using the Bosniak Classification of Cystic Renal Masses, which determines the likelihood that cysts are malignant or benign. Asymptomatic Bosniak class I and II cysts require no further evaluation or follow-up; however, symptomatic large simple cysts might require treatment. Bosniak class III and IV cysts might be malignant and require excision. Kidney tumors also occur in children and adults. In children, the most common is Wilms tumor, but after age 10 years renal cell carcinoma (RCC) is more common. In adults, kidney tumors may be malignant or benign. RCC accounts for 85% of kidney tumors in adults, often with metastatic disease. In patients with kidney tumors, biopsy typically is avoided to prevent spread of malignant cells. Tumors that appear suspicious for cancer on imaging are managed directly, which can include total or partial nephrectomy, ablation therapy, and adjuvant therapies, along with chemotherapy and radiotherapy depending on tumor stage. For some patients, evaluation may involve consideration of genetic testing for hereditary cancer syndromes. Patients with these syndromes should undergo periodic screening for RCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Diseases, Cystic , Kidney Neoplasms , Wilms Tumor , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/therapy , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/therapy , Wilms Tumor/diagnosis , Wilms Tumor/therapy , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/therapy , Adult , Child
6.
World J Urol ; 42(1): 474, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112814

ABSTRACT

PURPOSE: To examine associations between ablative therapy (AT) and partial nephrectomy (PN) and the occurrence of local recurrence (LR), distant metastatic recurrence (DMR) and all-cause mortality in a nation-wide real-world population-based cohort of patients with nonmetastatic renal cell carcinoma (nmRCC). METHODS: Data on 2751 AT- or PN-treated nmRCC tumours diagnosed during 2005-2018, representing 2701 unique patients, were obtained from the National Swedish Kidney Cancer Register. Time to LR/DMR or death with/without LR/DMR was analysed using Cox regression models. RESULTS: During a mean of 4.8 years follow-up, LR was observed for 111 (4.0%) tumours, DMR for 108 (3.9%) tumours, and death without LR/DMR for 206 (7.5%) tumours. AT-treated tumours had a 4.31 times higher risk of LR (P < 0.001) and a 1.91 times higher risk of DMR (P = 0.018) than PN-treated, with no significant differences in risk of death without LR/DMR. During a mean of 3.2 and 2.5 years of follow-up after LR/DMR, respectively, 24 (21.6%) of the LR cases and 56 (51.9%) of the DMR cases died, compared to 7.5% in patients without LR/DMR. There were no significant differences between AT- and PN-treated regarding risks of early death after occurrence of LR or DMR. CONCLUSION: AT treatment of patients with nmRCC implied significantly higher risks of LR and DMR compared with PN treatment. To minimize the risks of LR and DMR, these results suggest that PN is preferred over AT as primary treatment, supporting the EAU guidelines to recommended AT mainly to frail and/or comorbid patients.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Neoplasm Recurrence, Local , Nephrectomy , Nephrons , Humans , Carcinoma, Renal Cell/surgery , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Kidney Neoplasms/therapy , Male , Female , Aged , Middle Aged , Neoplasm Recurrence, Local/epidemiology , Nephrectomy/methods , Risk Assessment , Organ Sparing Treatments , Ablation Techniques/methods , Sweden/epidemiology
7.
Front Immunol ; 15: 1428584, 2024.
Article in English | MEDLINE | ID: mdl-39091498

ABSTRACT

Renal cell carcinoma (RCC) is considered radio- and chemo-resistant. Immune checkpoint inhibitors (ICIs) have demonstrated significant clinical efficacy in advanced RCC. However, the overall response rate of RCC to monotherapy remains limited. Given its immunomodulatory effects, a combination of radiotherapy (RT) with immunotherapy is increasingly used for cancer treatment. Heavy ion radiotherapy, specifically the carbon ion radiotherapy (CIRT), represents an innovative approach to cancer treatment, offering superior physical and biological effectiveness compared to conventional photon radiotherapy and exhibiting obvious advantages in cancer treatment. The combination of CIRT and immunotherapy showed robust effectiveness in preclinical studies of various tumors, thus holds promise for overcoming radiation resistance of RCC and enhancing therapeutic outcomes. Here, we provide a comprehensive review on the biophysical effects of CIRT, the efficacy of combination treatment and the underlying mechanisms involved in, as well as its therapeutic potential specifically within RCC.


Subject(s)
Carcinoma, Renal Cell , Heavy Ion Radiotherapy , Immune Checkpoint Inhibitors , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/radiotherapy , Carcinoma, Renal Cell/immunology , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/therapy , Kidney Neoplasms/radiotherapy , Kidney Neoplasms/immunology , Combined Modality Therapy , Animals , Immunotherapy/methods
8.
Theranostics ; 14(9): 3693-3707, 2024.
Article in English | MEDLINE | ID: mdl-38948062

ABSTRACT

Background: Immune checkpoint inhibitors (ICI) are routinely used in advanced clear cell renal cell carcinoma (ccRCC). However, a substantial group of patients does not respond to ICI therapy. Radiation is a promising approach to increase ICI response rates since it can generate anti-tumor immunity. Targeted radionuclide therapy (TRT) is a systemic radiation treatment, ideally suited for precision irradiation of metastasized cancer. Therefore, the aim of this study is to explore the potential of combined TRT, targeting carbonic anhydrase IX (CAIX) which is overexpressed in ccRCC, using [177Lu]Lu-DOTA-hG250, and ICI for the treatment of ccRCC. Methods: In this study, we evaluated the therapeutic and immunological action of [177Lu]Lu-DOTA-hG250 combined with aPD-1/a-CTLA-4 ICI. First, the biodistribution of [177Lu]Lu-DOTA-hG250 was investigated in BALB/cAnNRj mice bearing Renca-CAIX or CT26-CAIX tumors. Renca-CAIX and CT26-CAIX tumors are characterized by poor versus extensive T-cell infiltration and homogeneous versus heterogeneous PD-L1 expression, respectively. Tumor-absorbed radiation doses were estimated through dosimetry. Subsequently, [177Lu]Lu-DOTA-hG250 TRT efficacy with and without ICI was evaluated by monitoring tumor growth and survival. Therapy-induced changes in the tumor microenvironment were studied by collection of tumor tissue before and 5 or 8 days after treatment and analyzed by immunohistochemistry, flow cytometry, and RNA profiling. Results: Biodistribution studies showed high tumor uptake of [177Lu]Lu-DOTA-hG250 in both tumor models. Dose escalation therapy studies in Renca-CAIX tumor-bearing mice demonstrated dose-dependent anti-tumor efficacy of [177Lu]Lu-DOTA-hG250 and remarkable therapeutic synergy including complete remissions when a presumed subtherapeutic TRT dose (4 MBq, which had no significant efficacy as monotherapy) was combined with aPD-1+aCTLA-4. Similar results were obtained in the CT26-CAIX model for 4 MBq [177Lu]Lu-DOTA-hG250 + a-PD1. Ex vivo analyses of treated tumors revealed DNA damage, T-cell infiltration, and modulated immune signaling pathways in the TME after combination treatment. Conclusions: Subtherapeutic [177Lu]Lu-DOTA-hG250 combined with ICI showed superior therapeutic outcome and significantly altered the TME. Our results underline the importance of investigating this combination treatment for patients with advanced ccRCC in a clinical setting. Further investigations should focus on how the combination therapy should be optimally applied in the future.


Subject(s)
Carbonic Anhydrase IX , Carcinoma, Renal Cell , Immune Checkpoint Inhibitors , Kidney Neoplasms , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/pathology , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/radiotherapy , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase IX/antagonists & inhibitors , Humans , Cell Line, Tumor , Radioisotopes/therapeutic use , Radioisotopes/pharmacology , Radioisotopes/administration & dosage , Lutetium/therapeutic use , Female , Antigens, Neoplasm/metabolism , Tissue Distribution , Tumor Microenvironment/drug effects , Tumor Protein, Translationally-Controlled 1 , Xenograft Model Antitumor Assays , Combined Modality Therapy/methods , Mice, Inbred BALB C , Antibodies, Monoclonal
9.
Clin Exp Med ; 24(1): 161, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023752

ABSTRACT

Mitochondrial dysfunction and necrotic apoptosis, pivotal in therapeutic strategies for multiple diseases, lack comprehensive understanding in the context of renal clear cell carcinoma (ccRCC). This study explores their potential as valuable tools for ccRCC prediction, prevention, and personalized medical care. Transcriptomic and clinical datasets were acquired from the Cancer Genome Atlas (TCGA) repository. Mitochondrial and necrosis-associated gene sets were sourced from MitoCarta3.0 and the KEGG Pathway databases, respectively. Six necrosis-related mitochondrial genes (nc-MTGs) with prognostic significance were analyzed and screened, and a prognostic model was constructed. The accuracy of the model was verified using external data (E-MTAB-1980). TISCH was used to explore nc-MTGs at the cellular level. Finally, the expression level of BH3 interacting domain death agonist (BID) in ccRCC cell line was detected by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR), and the effect of BID down-regulation on tumor cell migration was verified by transwell assays and wound-healing experiments. We established and validated a prognostic model for clear cell renal cell carcinoma (ccRCC) utilizing six necrosis-related mitochondrial genes (nc-MTGs), affirming its efficacy in evaluating tumor progression. RT-PCR results showed that BID expression was up-regulated in ccRCC tissues compared with controls and exhibited oncogenic effects. In vitro cell function experiments showed that BID may be an important factor affecting the migration of ccRCC. Our study is the first to elucidate the biological functions and prognostic significance of mitochondrial molecules related to necroptosis, providing a new way to evaluate mitochondrial therapeutics in patients with ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Necrosis , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Prognosis , Immunotherapy , Cell Line, Tumor , Genes, Mitochondrial , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Mitochondria/genetics , Transcriptome , Male
10.
World J Urol ; 42(1): 451, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066794

ABSTRACT

PURPOSE: To evaluate the incidence of postoperative complications after cytoreductive nephrectomy (CN) following first-line treatment for metastatic renal cell carcinoma (mRCC), and to compare it with postoperative complications of upfront CN. METHODS: For this population-based retrospective study, the PearlDiver Mariner database (PearlDiver Technologies, Colorado Springs, CO), a database of insurance billing records was analyzed. Using relevant ICD-9/10 and CPT codes, patients diagnosed with mRCC between 2011 and 2021, who received first-line systemic molecular therapy (SMT), either tyrosine kinase inhibitors (TKI) or immune-checkpoint inhibitors (ICI), were identified. The selected population was stratified into two cohorts according to the timing of CN (deferred: after SMT vs. upfront: before SMT). Propensity-score matching (PSM) was performed as per baseline patients' characteristics to control for potential confounders between the two cohorts. The primary outcome was to compare 30-day postoperative complications rate between patients undergoing upfront vs. deferred CN. RESULTS: After PSM, 162 patients who received upfront CN were compared with 162 patients who underwent deferred CN. The overall rate of 30-day postoperative complications was statistically significantly higher in patients undergoing deferred CN (33.9%), compared to patients treated with upfront CN (21%, p < 0.01). In addition, the rate of both medical (26.5% vs. 14.2%, p < 0.01) and surgical (14.8 vs. 7.4%, p = 0.03) complication rate was statistically significantly higher in deferred vs. upfront CN. Multivariable logistic regression analysis revealed that none of the treatment regimens significantly predicted the occurrence of postoperative complications. CONCLUSION: Patients undergoing deferred CN experience a higher rates of overall, medical, and surgical 30-day postoperative complications compared to those receiving upfront surgery. Findings from this study should be interpreted within the limitations of this type of analysis.


Subject(s)
Carcinoma, Renal Cell , Cytoreduction Surgical Procedures , Immunotherapy , Kidney Neoplasms , Nephrectomy , Postoperative Complications , Propensity Score , Humans , Kidney Neoplasms/surgery , Kidney Neoplasms/drug therapy , Nephrectomy/methods , Male , Female , Carcinoma, Renal Cell/surgery , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/therapy , Middle Aged , Retrospective Studies , Postoperative Complications/epidemiology , Aged , Molecular Targeted Therapy , Incidence
11.
Medicine (Baltimore) ; 103(30): e38991, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058879

ABSTRACT

Immune checkpoint inhibitor (ICI) combinations, as well as ICIs combined with tyrosine kinase inhibitors, have considerable potential for renal cell carcinoma (RCC) treatment. Newer targeted medications, gut microbiome, nanomedicines, and cyclin-dependent kinase (CDK) inhibitors demonstrate significant potential in preventing side effects and resistance associated with RCC treatment. Most patients, including those demonstrating long-term treatment effects, eventually demonstrate cancer progression. Nevertheless, recent studies have further revealed RCC pathogenesis and many acquired drug resistance mechanisms, which together have led to the identification of promising therapeutic targets. In addition to having roles in metabolism, immunogenicity, and the immune response to tumors, CDK4 and CDK6 regulate the cell cycle. Targeting CDK4 and CDK6, either separately or in combination with already approved treatments, may improve therapeutic outcomes in patients with kidney cancer. Other novel drugs, including pegylated interleukin 10, colony-stimulating factor 1 receptor inhibitors, CD40 agonists, and C-X-C receptor 4 inhibitors affect the tumor microenvironment and cancer cell metabolism. Moreover, a triple ICI combination has been noted to be efficacious. In general, compared with sunitinib as a single-drug treatment, newer ICI combinations improve overall survival in patients with RCC. Future research on the prevention of adverse events and medication resistance related to newer therapies may aid in ensuring effective treatment outcomes among patients with RCC. This article aims to summarize innovative immunotherapy drug combinations for RCC treatment and the mechanisms of action, drug resistance, and treatment of adverse events associated with these combinations.


Subject(s)
Carcinoma, Renal Cell , Immune Checkpoint Inhibitors , Immunotherapy , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/therapy , Immunotherapy/methods , Immunotherapy/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects
12.
Front Immunol ; 15: 1361010, 2024.
Article in English | MEDLINE | ID: mdl-39034992

ABSTRACT

Background: Immune-checkpoint inhibitors (ICIs) have significantly improved metastatic renal cell carcinoma (mRCC) prognosis, although their efficacy in patients with bone metastases (BMs) remains poorly understood. We investigated the prognostic role of natremia in pretreated RCC patients with BMs receiving immunotherapy. Materials and methods: This retrospective multicenter study included RCC patients with BMs receiving nivolumab as second-line therapy or beyond. Inclusion criteria involved baseline sodium levels (pre-ICI) and sodium levels after 4 weeks of nivolumab initiation (post-ICI). The population was divided into two groups based on the median value, and response rates, progression-free survival (PFS), and overall survival (OS) were assessed. Results: Among 120 eligible patients, those with pre-treatment sodium levels ≥140 mEq/L showed longer OS (18.7 vs. 12.0 months, p=0.04). Pre-treatment sodium levels ≥140 mEq/L were associated with better OS compared to levels <140 mE/L (18.7 vs. 12.0, p=0.04). Post-treatment sodium levels ≥140 mEq/L were associated with improved PFS (9.6 vs. 3.2 months) and OS (25.1 vs. 8.8 months) (p=0.05 and p<0.01, respectively). Patients with consistent sodium levels ≥140 mEq/L at both time points exhibited the best outcomes compared to those with lower values (PFS 11.5 vs. 3.3 months and OS 42.2 vs. 9.0 months, respectively, p<0.01). Disease control rate was significantly higher in the latter group (p<0.01). Multivariate analysis confirmed the prognostic significance of sodium levels. Conclusion: Elevated sodium levels (≥140 mEq/L) pre- and post-ICI treatment correlate with better survival outcomes in mRCC patients with BMs. This finding suggests sodium level assessment as a potential prognostic factor in these patients and warrants further investigation, particularly in combination immunotherapy settings.


Subject(s)
Bone Neoplasms , Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Sodium , Humans , Male , Female , Middle Aged , Bone Neoplasms/secondary , Bone Neoplasms/mortality , Bone Neoplasms/therapy , Aged , Retrospective Studies , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/mortality , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Sodium/blood , Immunotherapy/methods , Nivolumab/therapeutic use , Prognosis , Immune Checkpoint Inhibitors/therapeutic use , Adult , Treatment Outcome , Aged, 80 and over
13.
Front Immunol ; 15: 1352632, 2024.
Article in English | MEDLINE | ID: mdl-39035007

ABSTRACT

Introduction: This study investigates the role of Fibroblast Activation Protein (FAP)-positive cancer-associated fibroblasts (FAP+CAF) in shaping the tumor immune microenvironment, focusing on its association with immune cell functionality and cytokine expression patterns. Methods: Utilizing immunohistochemistry, we observed elevated FAP+CAF density in metastatic versus primary renal cell carcinoma (RCC) tumors, with higher FAP+CAF correlating with increased T cell infiltration in RCC, a unique phenomenon illustrating the complex interplay between tumor progression, FAP+CAF density, and immune response. Results: Analysis of immune cell subsets in FAP+CAF-rich stromal areas further revealed significant correlations between FAP+ stroma and various T cell types, particularly in RCC and non-small cell lung cancer (NSCLC). This was complemented by transcriptomic analyses, expanding the range of stromal and immune cell subsets interrogated, as well as to additional tumor types. This enabled evaluating the association of these subsets with tumor infiltration, tumor vascularization and other components of the tumor microenvironment. Our comprehensive study also encompassed cytokine, angiogenesis, and inflammation gene signatures across different cancer types, revealing heterogeneous cellular composition, cytokine expressions and angiogenic profiles. Through cytokine pathway profiling, we explored the relationship between FAP+CAF density and immune cell states, uncovering potential immunosuppressive circuits that limit anti-tumor activity in tumor-resident immune cells. Conclusions: These findings underscore the complexity of tumor biology and the necessity for personalized therapeutic and patient enrichment approaches. The insights gathered from FAP+CAF prevalence, immune infiltration, and gene signatures provide valuable perspectives on tumor microenvironments, aiding in future research and clinical strategy development.


Subject(s)
Cancer-Associated Fibroblasts , Immunotherapy , Serine Endopeptidases , Tumor Microenvironment , Tumor Microenvironment/immunology , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Immunotherapy/methods , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Cytokines/metabolism , Endopeptidases , Membrane Proteins/metabolism , Membrane Proteins/genetics , Gelatinases/metabolism , Gelatinases/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lung Neoplasms/metabolism
14.
Lancet ; 404(10451): 476-491, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39033764

ABSTRACT

The landscape of the management of renal cell carcinoma has evolved substantially in the last decade, leading to improved survival in localised and advanced disease. We review the epidemiology, pathology, and diagnosis of renal cell carcinoma and discuss the evidence for current management strategies from localised to metastatic disease. Developments in adjuvant therapies are discussed, including use of pembrolizumab-the first therapy to achieve overall survival benefit in the adjuvant setting. The treatment of advanced disease, including landmark trials that have established immune checkpoint inhibition as a standard of care, are also reviewed. We also discuss the current controversies that exist surrounding the management of metastatic renal cell carcinoma, including the use of risk assessment models for disease stratification and treatment selection for frontline therapy. Management of non-clear cell renal cell carcinoma subtypes is also reviewed. Future directions of research, including a discussion of ongoing clinical trials and the need for reliable biomarkers to guide treatment in kidney cancer, are also highlighted.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/epidemiology , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Chemotherapy, Adjuvant
15.
Mol Cancer ; 23(1): 146, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014460

ABSTRACT

The advent of PD1/PD-L1 inhibitors has significantly transformed the therapeutic landscape for clear cell renal cell carcinoma (ccRCC). This review provides an in-depth analysis of the biological functions and regulatory mechanisms of PD1 and PD-L1 in ccRCC, emphasizing their role in tumor immune evasion. We comprehensively evaluate the clinical efficacy and safety profiles of PD1/PD-L1 inhibitors, such as Nivolumab and Pembrolizumab, through a critical examination of recent clinical trial data. Furthermore, we discuss the challenges posed by resistance mechanisms to these therapies and potential strategies to overcome them. We also explores the synergistic potential of combination therapies, integrating PD1/PD-L1 inhibitors with other immunotherapies, targeted therapies, and conventional modalities such as chemotherapy and radiotherapy. In addition, we examine emerging predictive biomarkers for response to PD1/PD-L1 blockade and biomarkers indicative of resistance, providing a foundation for personalized therapeutic approaches. Finally, we outline future research directions, highlighting the need for novel therapeutic strategies, deeper mechanistic insights, and the development of individualized treatment regimens. Our work summarizes the latest knowledge and progress in this field, aiming to provide a valuable reference for improving clinical efficacy and guiding future research on the application of PD1/PD-L1 inhibitors in ccRCC.


Subject(s)
B7-H1 Antigen , Carcinoma, Renal Cell , Immune Checkpoint Inhibitors , Kidney Neoplasms , Programmed Cell Death 1 Receptor , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Biomarkers, Tumor , Treatment Outcome , Animals , Drug Resistance, Neoplasm , Molecular Targeted Therapy , Immunotherapy/methods
16.
J Bras Nefrol ; 46(3): e20240013, 2024.
Article in English, Portuguese | MEDLINE | ID: mdl-38991206

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by the development of hamartomas in the central nervous system, heart, skin, lungs, and kidneys and other manifestations including seizures, cortical tubers, radial migration lines, autism and cognitive disability. The disease is associated with pathogenic variants in the TSC1 or TSC2 genes, resulting in the hyperactivation of the mTOR pathway, a key regulator of cell growth and metabolism. Consequently, the hyperactivation of the mTOR pathway leads to abnormal tissue proliferation and the development of solid tumors. Kidney involvement in TSC is characterized by the development of cystic lesions, renal cell carcinoma and renal angiomyolipomas, which may progress and cause pain, bleeding, and loss of kidney function. Over the past years, there has been a notable shift in the therapeutic approach to TSC, particularly in addressing renal manifestations. mTOR inhibitors have emerged as the primary therapeutic option, whereas surgical interventions like nephrectomy and embolization being reserved primarily for complications unresponsive to clinical treatment, such as severe renal hemorrhage. This review focuses on the main clinical characteristics of TSC, the mechanisms underlying kidney involvement, the recent advances in therapy for kidney lesions, and the future perspectives.


Subject(s)
Tuberous Sclerosis , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics , Tuberous Sclerosis/therapy , Humans , Kidney Neoplasms/therapy , Kidney Neoplasms/etiology , MTOR Inhibitors/therapeutic use , TOR Serine-Threonine Kinases , Angiomyolipoma/etiology , Angiomyolipoma/therapy , Nephrology , Tuberous Sclerosis Complex 1 Protein/genetics , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/genetics
17.
Clin Exp Med ; 24(1): 167, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052149

ABSTRACT

Renal cell carcinoma (RCC) is characterized by a variety of subtypes, each defined by unique genetic and morphological features. This study utilizes single-cell RNA sequencing to explore the molecular heterogeneity of RCC. A highly proliferative cell subset, termed as "Prol," was discovered within RCC tumors, and its increased presence was linked to poorer patient outcomes. An artificial intelligence network, encompassing traditional regression, machine learning, and deep learning algorithms, was employed to develop a Prol signature capable of predicting prognosis. The signature demonstrated superior performance in predicting RCC prognosis compared to other signatures and exhibited pan-cancer prognostic capabilities. RCC patients with high Prol signature scores exhibited resistance to targeted therapies and immunotherapies. Furthermore, the key gene CEP55 from the Prol signature was validated by both proteinomics and quantitative real time polymerase chain reaction. Our findings may provide new insights into the molecular and cellular mechanisms of RCC and facilitate the development of novel biomarkers and therapeutic targets.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Single-Cell Analysis , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Prognosis , Cell Proliferation , Female , Male , Sequence Analysis, RNA , Biomarkers, Tumor/genetics , Middle Aged , Gene Expression Profiling
18.
Aging (Albany NY) ; 16(13): 11072-11089, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970774

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is a common kidney cancer with subtle early symptoms, high recurrence rates, and low sensitivity to traditional treatments like radiotherapy and chemotherapy. Identifying novel therapeutic targets is critical. The expression level of adenylate kinases 7 (AK7) in ccRCC was examined by the TCGAportal and UALCAN databases. The effect of AK7 on proliferation, invasion and migration of ccRCC cell lines was evaluated by cell assay. The correlation between AK7 expression and prognosis, as well as its direct relationship with immunotherapy efficacy, was analyzed using CANCERTOOL and Kaplan-Meier plotter data. Moreover, the TISIDB database was used to study the relationship between AK7 and immune markers. The effect of overexpressed AK7 combined with PD1 monoclonal antibody on ccRCC was evaluated in animal experiments. The results showed that low level of AK7 expression was observed in ccRCC tissues. The expression of AK7 can regulate the proliferation, invasion, and migration of human ccRCC cell lines. The level of AK7 expression was associated with OS of ccRCC patients. This was potentially due to the negative connection between AK7 expression and CD8+ T cell depletion, indicating that immunotherapy might be less effective in individuals with low AK7 expression. Conversely, augmenting AK7 demonstrated an enhanced effectiveness of anti-PD1 therapy. The findings of our research strongly indicated that AK7 could serve as both a prognostic indicator and therapeutic target for patients with ccRCC. Moreover, the overexpression of AK7 combined with anti-PD1 held promising potential as a therapeutic approach for treating ccRCC.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Programmed Cell Death 1 Receptor , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Animals , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Immunotherapy/methods , Mice , Cell Proliferation/drug effects , Cell Movement , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Disease Progression , Prognosis , Female , Male , Gene Expression Regulation, Neoplastic , Xenograft Model Antitumor Assays
19.
Nat Commun ; 15(1): 5935, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009593

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common form of kidney cancer, but a comprehensive description of its genomic landscape is lacking. We report the whole genome sequencing of 778 ccRCC patients enrolled in the 100,000 Genomes Project, providing for a detailed description of the somatic mutational landscape of ccRCC. We identify candidate driver genes, which as well as emphasising the major role of epigenetic regulation in ccRCC highlight additional biological pathways extending opportunities for therapeutic interventions. Genomic characterisation identified patients with divergent clinical outcome; higher number of structural copy number alterations associated with poorer prognosis, whereas VHL mutations were independently associated with a better prognosis. The observations that higher T-cell infiltration is associated with better overall survival and that genetically predicted immune evasion is not common supports the rationale for immunotherapy. These findings should inform personalised surveillance and treatment strategies for ccRCC patients.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Mutation , Von Hippel-Lindau Tumor Suppressor Protein , Whole Genome Sequencing , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Prognosis , Male , Female , DNA Copy Number Variations , Middle Aged , Epigenesis, Genetic , Aged , Gene Expression Regulation, Neoplastic , Immunotherapy/methods
20.
SELECTION OF CITATIONS
SEARCH DETAIL