Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.249
Filter
3.
Hum Vaccin Immunother ; 20(1): 2350101, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38738709

ABSTRACT

Clinical guidelines have recently advised combination therapy involving immunotherapy (IO) and tyrosine kinase inhibitors (TKI) as the first-line therapy approach for advanced renal cell carcinoma (RCC). Nevertheless, there is currently no available biomarker that can effectively distinguish the progression-free survival (PFS). RNA-sequencing and immunohistochemistry were conducted on our cohort of metastatic RCC patients, namely ZS-MRCC, who received combination therapy consisting of IO and TKI. We further applied RNA-sequencing, immunohistochemistry, and flow cytometry to examine the immune cell infiltration and functionality inside the tumor microenvironment of high-risk localized RCC samples. SPP1 expression was significantly higher in non-responders to IO-TKI therapy. Elevated levels of SPP1 were associated with poor PFS in both the ZS-MRCC cohort (HR = 2.73, p = .018) and validated in the JAVELIN Renal 101 cohort (HR = 1.61, p = .004). By multivariate Cox analysis, SPP1 was identified as a significant independent prognosticator. Furthermore, there existed a negative correlation between elevated levels of SPP1 and the presence of GZMB+CD8+ T cells (Spearman's ρ= -0.48, p < .001). Conversely, SPP1 expression is associated with T cell exhaustion markers. A significant increase in the abundance of Tregs was observed in tumors with high levels of SPP1. Additionally, a machine-learning-based model was constructed to predict the benefit of IO-TKI treatment. High SPP1 is associated with therapeutic resistance and unfavorable PFS in IO-TKI therapy. SPP1 expression have also been observed to be indicative of malfunction and exhaustion in T cells. Increased SPP1 expression has the potential to serve as a potential biomarker for treatment selection of metastatic RCC.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Osteopontin , Protein Kinase Inhibitors , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Male , Female , Immunotherapy/methods , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Osteopontin/metabolism , Osteopontin/genetics , Aged , Tumor Microenvironment/immunology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Treatment Outcome , Adult , CD8-Positive T-Lymphocytes/immunology , Cohort Studies , Combined Modality Therapy
4.
Aging (Albany NY) ; 16(9): 7622-7646, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38728235

ABSTRACT

Renal cell carcinoma (RCC) is one of the most prevalent types of urological cancer. Exosomes are vesicles derived from cells and have been found to promote the development of RCC, but the potential biomarker and molecular mechanism of exosomes on RCC remain ambiguous. Here, we first screened differentially expressed exosome-related genes (ERGs) by analyzing The Cancer Genome Atlas (TCGA) database and exoRBase 2.0 database. We then determined prognosis-related ERGs (PRERGs) by univariate Cox regression analysis. Gene Dependency Score (gDS), target development level, and pathway correlation analysis were utilized to examine the importance of PRERGs. Machine learning and lasso-cox regression were utilized to screen and construct a 5-gene risk model. The risk model showed high predictive accuracy for the prognosis of patients and proved to be an independent prognostic factor in three RCC datasets, including TCGA-KIRC, E-MTAB-1980, and TCGA-KIRP datasets. Patients with high-risk scores showed worse outcomes in different clinical subgroups, revealing that the risk score is robust. In addition, we found that immune-related pathways are highly enriched in the high-risk group. Activities of immune cells were distinct in high-/low-risk groups. In independent immune therapeutic cohorts, high-risk patients show worse immune therapy responses. In summary, we identified several exosome-derived genes that might play essential roles in RCC and constructed a 5-gene risk signature to predict the prognosis of RCC and immune therapy response.


Subject(s)
Carcinoma, Renal Cell , Exosomes , Kidney Neoplasms , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Humans , Exosomes/genetics , Exosomes/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Prognosis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Immunotherapy , Female , Databases, Genetic , Male , Risk Assessment , Risk Factors
5.
World J Urol ; 42(1): 364, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819448

ABSTRACT

OBJECTIVES: Renal cell carcinoma (RCC) is infrequent among young adults. Few studies reported the outcome of RCC in young adults by pathological subtypes. The purpose of this study was to explore the clinicopathological features, survival outcomes and prognostic factors of young adult patients with clear cell (CCRCC) and non-clear cell renal cell carcinoma (NCCRCC). METHODS: This study included young adult patients aged 18-40 years who were diagnosed as renal cell carcinoma (RCC) between 2012 and 2022 at Peking University Third Hospital. All patients underwent either partial nephrectomy or radical nephrectomy, and some received adjuvant therapy. A comparative analysis was performed to investigate the differences in clinicopathological characteristics between the cohort of CCRCC and NCCRCC. Kaplan-Meier survival analysis was utilized to plot survival curves for young adults with RCC. The univariate and multifactorial prognostic analyses were conducted using the log-rank test and COX proportional hazards model. RESULTS: A total of 300 RCC patients aged 18-40 years were performed, of which 201 were diagnosed with CCRCC (67%) and 99 were diagnosed with NCCRCC(33%). The NCCRCC included 29 cases (9.7%) of chromophobe RCC, 28 cases (9.3%) of MiT family translocation RCC, 22 cases (7.3%) of papillary RCC, 11 cases (3.7%) of low malignant potential multifocal cystic RCC, and 6 cases of unclassified RCC (2.0%), 2 cases of mucinous tubule and spindle cell carcinoma (0.7%), and 1 case of FH-deficient RCC (0.3%).The mean age was 33.4 ± 6.1 years old. The overall and progression free 5-year survival rate was 99.1 and 95.3%, respectively. The NCCRCC cohort demonstrated a statistically significant decrease in progression-free survival (PFS) rate when compared to the CCRCC cohort (p < 0.001). There was no statistically significant difference observed in overall survival (OS) (p = 0.069). Pathological stage was a significant independent predictor for OS (p = 0.045). Pathological stage and nuclear grade were both independent predictors for PFS (p = 0.020; p = 0.005). CONCLUSIONS: The clinical and pathological features of young adults diagnosed with CCRCC exhibit notable distinctions from those of NCCRCC patients. The survival outcome was significantly influenced by the pathological stage, while both the nuclear grade and pathological stage had a significant impact on tumor progression. This study offered significant contributions to the understanding of the clinicopathological characteristics and prognostic determinants of renal cell carcinoma (RCC) in young adults.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Adult , Male , Young Adult , Female , Prognosis , Adolescent , Survival Rate , Retrospective Studies , Nephrectomy
6.
Cancer Med ; 13(11): e7308, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38808948

ABSTRACT

BACKGROUND: Exosomes play a crucial role in intercellular communication in clear cell renal cell carcinoma (ccRCC), while the long non-coding RNAs (lncRNAs) are implicated in tumorigenesis and progression. AIMS: The purpose of this study is to construction a exosomes-related lncRNA score and a ceRNA network to predict the response to immunotherapy and potential targeted drug in ccRCC. METHODS: Data of ccRCC patients were obtained from the TCGA database. Pearson correlation analysis was used to identify eExosomes-related lncRNAs (ERLRs) from Top10 exosomes-related genes that have been screened. The entire cohort was randomly divided into a training cohort and a validation cohort in equal scale. LASSO regression and multivariate cox regression was used to construct the ERLRs-based score. Differences in clinicopathological characteristics, immune microenvironment, immune checkpoints, and drug susceptibility between the high- and low-risk groups were also investigated. Finally, the relevant ceRNA network was constructed by machine learning to analyze their potential targets in immunotherapy and drug use of ccRCC patients. RESULTS: A score consisting of 4ERLRs was identified, and patients with higher ERLRs-based score tended to have a worse prognosis than those with lower ERLRs-based score. ROC curves and multivariate Cox regression analysis demonstrated that the score could be considered as a risk factor for prognosis in both training and validation cohorts. Moreover, patients with high scores are predisposed to experience poor overall survival, a larger prevalence of advanced stage (III-IV), a greater tumor mutational burden, a higher infiltration of immunosuppressive cells, and a greater likelihood of responding favorably to immunotherapy. The importance of EMX2OS was determined by mechanical learning, and the ceRNA network was constructed, and EMX2OS may be a potential therapeutic target, possibly exerting its function through the EMX2OS/hsa-miR-31-5p/TLN2 axis. CONCLUSIONS: Based on machine learning, a novel ERLRs-based score was constructed for predicting the survival of ccRCC patients. The ERLRs-based score is a promising potential independent prognostic factor that is closely correlated with the immune microenvironment and clinicopathological characteristics. Meanwhile, we screened out key lncRNAEMX2OS and identified the EMX2OS/hsa-miR-31-5p/TLN2 axis, which may provide new clues for the targeted therapy of ccRCC.


Subject(s)
Carcinoma, Renal Cell , Exosomes , Immunotherapy , Kidney Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/mortality , RNA, Long Noncoding/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Kidney Neoplasms/mortality , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Exosomes/genetics , Immunotherapy/methods , Male , Female , Middle Aged , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks
7.
J Cancer Res Clin Oncol ; 150(5): 278, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801430

ABSTRACT

BACKGROUND: The ramifications of necroptosis on the prognostication of clear cell renal cell carcinoma (ccRCC) remain inadequately expounded. METHODS: A prognostic model delineating the facets of necroptosis in ccRCC was constructed, employing a compendium of algorithms. External validation was effectuated using the E-MTAB-1980 dataset. The exploration of immune infiltration scores was undertaken through the exploitation of multiple algorithms. Single-cell RNA sequencing data were procured from the GSE171306 dataset. Real-time quantitative PCR (RT-qPCR) was engaged to scrutinize the differential expression of SLC25A37 across cancer and paracancer tissues, as well as diverse cell lines. Assessments of proliferative and metastatic alterations in 769-P and 786-O cells were accomplished through Cell Counting Kit-8 (CCK8) and wound healing assays. RESULTS: The necroptosis-related signature (NRS) emerges as a discerning metric, delineating patients' immune attributes, tumor mutation burden, immunotherapy response, and drug susceptibility. Single-cell RNA sequencing analysis unveils the marked enrichment of SLC25A37 in tumor cells. Concurrently, RT-qPCR discloses the overexpression of SLC25A37 in both ccRCC tissues and cell lines. SLC25A37 knockdown mitigates the proliferative and metastatic propensities of 769-P and 786-O cells, as evidenced by CCK8 and wound healing assays. CONCLUSION: The NRS assumes a pivotal role in ascertaining the prognosis, tumor mutation burden, immunotherapy response, drug susceptibility, and immune cell infiltration features of ccRCC patients. SLC25A37 emerges as a putative player in immunosuppressive microenvironments, thereby providing a prospective avenue for the design of innovative immunotherapeutic targets for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Necroptosis , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Humans , Kidney Neoplasms/pathology , Kidney Neoplasms/immunology , Kidney Neoplasms/genetics , Prognosis , Immunotherapy/methods , Cell Line, Tumor , Cell Proliferation , Biomarkers, Tumor/genetics , Tumor Microenvironment/immunology , Gene Expression Regulation, Neoplastic
8.
Cancer Immunol Immunother ; 73(5): 95, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38607586

ABSTRACT

BACKGROUND: Homologous recombination deficiency (HRD), though largely uncharacterized in clear cell renal cell carcinoma (ccRCC), was found associated with RAD51 loss of expression. PBRM1 is the second most common mutated genes in ccRCC. Here, we introduce a HRD function-based PBRM1-RAD51 ccRCC classification endowed with diverse immune checkpoint blockade (ICB) responses. METHODS: Totally 1542 patients from four independent cohorts were enrolled, including our localized Zhongshan hospital (ZSHS) cohort and Zhongshan hospital metastatic RCC (ZSHS-mRCC) cohort, The Cancer Genome Atlas (TCGA) cohort and CheckMate cohort. The genomic profile and immune microenvironment were depicted by genomic, transcriptome data and immunohistochemistry. RESULTS: We observed that PBRM1-loss ccRCC harbored enriched HRD-associated mutational signature 3 and loss of RAD51. Dual-loss of PBRM1 and RAD51 identified patients hyper-sensitive to immunotherapy. This dual-loss subtype was featured by M1 macrophage infiltration. Dual-loss was, albeit homologous recombination defective, with high chromosomal stability. CONCLUSIONS: PBRM1 and RAD51 dual-loss ccRCC indicates superior responses to immunotherapy. Dual-loss ccRCC harbors an immune-desert microenvironment but enriched with M1 macrophages. Dual-loss ccRCC is susceptible to defective homologous recombination but possesses high chromosomal stability.


Subject(s)
Carcinoma, Renal Cell , Carcinoma , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Immunotherapy , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Chromosomal Instability , Tumor Microenvironment , Rad51 Recombinase , DNA-Binding Proteins/genetics , Transcription Factors/genetics
9.
J Mater Chem B ; 12(16): 4039-4052, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38591157

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is a disease with high incidence and poor prognosis. The conventional treatment involves radiotherapy and chemotherapy, but chemotherapeutic agents are often associated with side effects, i.e., cytotoxicity to nontumor cells. Therefore, there is an urgent need for the development of novel therapeutic strategies for ccRCC. We synthesized spherical P/TiO2 nanoparticles (P/TiO2 NPs) by vaporization phosphorization (VP). X-ray photoelectron spectroscopy (XPS) and ultraviolet-visible diffuse reflectance spectroscopy (UV-Vis DRS) analyses confirmed that the anatase TiO2 surface was successfully doped with phosphorus and produced a large number of oxygen vacancies (OV). Serving as a photosensitizer, P/TiO2 NPs not only extended the photoresponse range to the near-infrared II region (NIR II) but also introduced a donor energy level lower than the TiO2 conduction band, narrowing the band gap, which could facilitate the migration of photogenerated charges and trigger the synergistic treatment of photodynamic therapy (PDT) and photothermal therapy (PTT). During NIR irradiation in vitro, the P/TiO2 NPs generated local heat and various oxygen radicals, including 1O2, ˙O2-, H2O2, and ˙OH, which damaged the ccRCC cells. In vivo, administration of the P/TiO2 NPs + NIR reduced the tumor volume by 80%, and had the potential to inhibit tumor metastasis by suppressing intratumor neoangiogenesis. The P/TiO2 NPs showed superior safety and efficacy relative to the conventional chemotherapeutic agent used in ccRCC treatment. This study introduced an innovative paradigm for renal cancer treatment, highlighting the potential of P/TiO2 NPs as safe and effective nanomaterials and presenting a compelling new option for clinical applications in anticancer therapy.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Nanocomposites , Phosphorus , Photochemotherapy , Photothermal Therapy , Titanium , Titanium/chemistry , Titanium/pharmacology , Phosphorus/chemistry , Humans , Animals , Nanocomposites/chemistry , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Mice , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Survival/drug effects , Cell Proliferation/drug effects , Mice, Nude , Mice, Inbred BALB C , Drug Screening Assays, Antitumor , Particle Size , Cell Line, Tumor
10.
JAMA Netw Open ; 7(4): e248747, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38687479

ABSTRACT

Importance: Area-level measures of sociodemographic disadvantage may be associated with racial and ethnic disparities with respect to receipt of treatment for metastatic renal cell carcinoma (mRCC) but have not been investigated previously, to our knowledge. Objective: To assess the association between area-level measures of social vulnerability and racial and ethnic disparities in the treatment of US Medicare beneficiaries with mRCC from 2015 through 2019. Design, Setting, and Participants: This retrospective cohort study included Medicare beneficiaries older than 65 years who were diagnosed with mRCC from January 2015 through December 2019 and were enrolled in fee-for-service Medicare Parts A, B, and D from 1 year before through 1 year after presumed diagnosis or until death. Data were analyzed from November 22, 2022, through January 26, 2024. Exposures: Five different county-level measures of disadvantage and 4 zip code-level measures of vulnerability or deprivation and segregation were used to dichotomize whether an individual resided in the most vulnerable quartile according to each metric. Patient-level factors included age, race and ethnicity, sex, diagnosis year, comorbidities, frailty, Medicare and Medicaid dual enrollment eligibility, and Medicare Part D low-income subsidy (LIS). Main Outcomes and Measures: The main outcomes were receipt and type of systemic therapy (oral anticancer agent or immunotherapy from 2 months before to 1 year after diagnosis of mRCC) as a function of patient and area-level characteristics. Multivariable regression analyses were used to adjust for patient factors, and odds ratios (ORs) from logistic regression and relative risk ratios (RRRs) from multinomial logistic regression are reported. Results: The sample included 15 407 patients (mean [SD] age, 75.6 [6.8] years), of whom 9360 (60.8%) were men; 6931 (45.0%), older than 75 years; 93 (0.6%), American Indian or Alaska Native; 257 (1.7%), Asian or Pacific Islander; 757 (4.9%), Hispanic; 1017 (6.6%), non-Hispanic Black; 12 966 (84.2%), non-Hispanic White; 121 (0.8%), other; and 196 (1.3%), unknown. Overall, 8317 patients (54.0%) received some type of systemic therapy. After adjusting for individual factors, no county or zip code-level measures of social vulnerability, deprivation, or segregation were associated with disparities in treatment. In contrast, patient-level factors, including female sex (OR, 0.78; 95% CI, 0.73-0.84) and LIS (OR, 0.48; 95% CI, 0.36-0.65), were associated with lack of treatment, with particularly limited access to immunotherapy for patients with LIS (RRR, 0.25; 95% CI, 0.14-0.43). Associations between individual-level factors and treatment in multivariable analysis were not mediated by the addition of area-level metrics. Disparities by race and ethnicity were consistently and only observed within the most vulnerable areas, as indicated by the top quartile of each vulnerability deprivation index. Conclusions and Relevance: In this cohort study of older Medicare patients diagnosed with mRCC, individual-level demographics, including race and ethnicity, sex, and income, were associated with receipt of systemic therapy, whereas area-level measures were not. However, individual-level racial and ethnic disparities were largely limited to socially vulnerable areas, suggesting that efforts to improve racial and ethnic disparities may be most effective when targeted to socially vulnerable areas.


Subject(s)
Carcinoma, Renal Cell , Healthcare Disparities , Kidney Neoplasms , Medicare , Humans , Male , Female , Aged , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/ethnology , United States , Retrospective Studies , Medicare/statistics & numerical data , Kidney Neoplasms/therapy , Kidney Neoplasms/ethnology , Healthcare Disparities/statistics & numerical data , Healthcare Disparities/ethnology , Aged, 80 and over , Social Vulnerability , Vulnerable Populations/statistics & numerical data , Socioeconomic Factors
11.
BMC Genomics ; 25(1): 413, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671348

ABSTRACT

BACKGROUND: Disulfidptosis is a novel form of programmed cell death induced by high SLC7A11 expression under glucose starvation conditions, unlike other known forms of cell death. However, the roles of disulfidptosis in cancers have yet to be comprehensively well-studied, particularly in ccRCC. METHODS: The expression profiles and somatic mutation of DGs from the TCGA database were investigated. Two DGs clusters were identified by unsupervised consensus clustering analysis, and a disulfidptosis-related prognostic signature (DR score) was constructed. Furthermore, the predictive capacity of the DR score in prognosis was validated by several clinical cohorts. We also developed a nomogram based on the DR score and clinical features. Then, we investigated the differences in the clinicopathological information, TMB, tumor immune landscapes, and biological characteristics between the high- and low-risk groups. We evaluated whether the DR score is a robust tool for predicting immunotherapy response by the TIDE algorithm, immune checkpoint genes, submap analysis, and CheckMate immunotherapy cohort. RESULTS: We identified two DGs clusters with significant differences in prognosis, tumor immune landscapes, and clinical features. The DR score has been demonstrated as an independent risk factor by several clinical cohorts. The high-risk group patients had a more complicated tumor immune microenvironment and suffered from more tumor immune evasion in immunotherapy. Moreover, patients in the low-risk group had better prognosis and response to immunotherapy, particularly in anti-PD1 and anti-CTLA-4 inhibitors, which were verified in the CheckMate immunotherapy cohort. CONCLUSION: The DR score can accurately predict the prognosis and immunotherapy response and assist clinicians in providing a personalized treatment regime for ccRCC patients.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Prognosis , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Biomarkers, Tumor/genetics , Nomograms , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Mutation , Apoptosis
12.
Biomaterials ; 308: 122550, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38581762

ABSTRACT

Immune checkpoint blockade therapy represented by programmed cell death ligand 1 (PD-L1) inhibitor for advanced renal carcinoma with an objective response rate (ORR) in patients is less than 20%. It is attributed to abundant tumoral vasculature with abnormal structure limiting effector T cell infiltration and drug penetration. We propose a bispecific fibrous glue (BFG) to regulate tumor immune and vascular microenvironments simultaneously. The bispecific precursor glue peptide-1 (pre-GP1) can penetrate tumor tissue deeply and self-assemble into BFG in the presence of neuropilin-1 (NRP-1) and PD-L1. The resultant fibrous glue is capable of normalizing tumoral vasculature as well as restricting immune escape. The pre-GP1 retains a 6-fold higher penetration depth than that of antibody in the multicellular spheroids (MCSs) model. It also shows remarkable tumor growth inhibition (TGI) from 19% to 61% in a murine advanced large tumor model compared to the clinical combination therapy. In addition, in the orthotopic renal tumor preclinical model, the lung metastatic nodules are reduced by 64% compared to the clinically used combination. This pre-GP1 provides a promising strategy to control the progression and metastasis of advanced renal carcinoma.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Kidney Neoplasms/pathology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/therapy , Kidney Neoplasms/immunology , Humans , Mice , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Tumor Microenvironment/drug effects , Mice, Inbred BALB C , Female , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism
13.
Urologie ; 63(5): 439-447, 2024 May.
Article in German | MEDLINE | ID: mdl-38602533

ABSTRACT

BACKGROUND: Renal cell carcinoma is the third most common tumor among urological tumors. In Germany more than 14,000 people are affected every year. The sex ratio is 2/3 men and 1/3 women. OBJECTIVES: The S3 guideline is intended to provide all disciplines dealing with renal cell carcinoma with the current status of diagnostics, therapy and follow-up care of the patients with this tumor. MATERIALS AND METHODS: The first version of the German guideline on renal cell carcinoma was published in 2015. The development was carried out at S3 level, which means that a structured, evidence-based literature search was carried out, recommendations and statements were developed in topic-related working groups and were approved by an interdisciplinary group of officials elected by the different medical societies. The chapters were gradually revised in 2017, 2020 and 2021 to reflect new aspects. This article provides information about the most important innovations of the most recent update from 2023. RESULTS: In the epidemiology subsection, the substance trichlorethene has been added as a risk factor for the development of renal cell carcinoma. While there were no new data on neoadjuvant therapy, the checkpoint inhibitor pembrolizumab was the first substance to demonstrate improved disease-specific and overall survival in the adjuvant situation. The combination nivolumab plus cabozantinib and lenvatinib plus pembrolizumab were included in the chapter on systemic therapy for metastatic clear cell renal cell carcinoma. New are the chapters on non-clear cell renal cell carcinoma and hereditary tumors. CONCLUSIONS: The S3 guideline provides a structured, evidence-based overview of all aspects of renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/diagnosis , Germany , Practice Guidelines as Topic
14.
Curr Opin Urol ; 34(4): 300-306, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38595192

ABSTRACT

PURPOSE OF REVIEW: Oligometastatic renal cell carcinoma (RCC) is a complex entity, potentially leading to a specific clinical management of these patients. Recent and ongoing trials have raised several unresolved questions that could impact clinical routine practice, advocating for the integration of novel treatment options (systemic treatment, cytoreductive surgery, or stereotactic body radiotherapy - SBRT) with varied modalities and objectives. RECENT FINDINGS: Immunotherapy represents a breakthrough in the systemic treatment of mRCC. However, many questions are still unsolved regarding the perfect timing for starting systemic and whether the systemic treatment could improve the activity of metastases-directed strategies. Moreover, the widespread use of adjuvant immunotherapy will challenge the treatment paradigm in the oligorecurrent scenario. Radical surgery of metastases and more recently SBRT - both eventually associated with systemic treatment - actually represent two important approaches to be considered in oligometastatic patients. SUMMARY: Oligometastatic RCC represents a status including a wide spectrum of clinical conditions that requires a tailored treatment approach. The correct management integrates local approaches (either metastasectomy or SRBT) and systemic (immune)-therapy. Several unmet needs have to be investigated, mainly regarding the lack of prospective randomized trials that directly compare modern therapies and different integration strategies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Radiosurgery/methods , Immunotherapy/methods , Neoplasm Metastasis , Metastasectomy/methods , Cytoreduction Surgical Procedures/methods , Nephrectomy/methods , Combined Modality Therapy/methods
15.
Cancer Cell ; 42(5): 732-735, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38579722

ABSTRACT

Saliby et al. show that a machine learning approach can accurately classify clear cell renal cell carcinoma (RCC) into distinct molecular subtypes using transcriptomic data. When applied to tumors biospecimens from the JAVELIN Renal 101 (JR101) trial, a benefit is observed with immune checkpoint inhibitor (ICI)-based therapy across all molecular subtypes.


Subject(s)
Carcinoma, Renal Cell , Immune Checkpoint Inhibitors , Immunotherapy , Kidney Neoplasms , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/drug therapy , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Kidney Neoplasms/drug therapy , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Molecular Targeted Therapy/methods , Treatment Outcome , Machine Learning
16.
Clin Genitourin Cancer ; 22(3): 102081, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641444

ABSTRACT

BACKGROUND AND OBJECTIVE: Considering the rapidly evolving treatment landscape of renal cell carcinoma (RCC), recent descriptions of the RCC population in the UK are lacking, as are real-world data on treatment and patient outcomes. To analyse the demographic and clinical characteristics, treatment patterns, and overall survival of patients with RCC using national data sets in England. PATIENTS AND METHODS: This was a retrospective cohort study of patients diagnosed with RCC (all stages) between 2014-2018 using demographic, clinical, cancer registration, and treatment data. Patients were followed until death or study end (December 31, 2020). Treatments administered in each line were described to understand treatment sequencing. Kaplan-Meier methods were used for time-to-event analyses. Factors associated with discontinuation and survival were identified using Cox proportional hazard models. RESULTS AND LIMITATIONS: Among 32,577 included patients, the median age at diagnosis was 66 years, 63.4% were male, and 6,786 (20.8%) had metastatic RCC at diagnosis. Tyrosine kinase inhibitor (TKI) monotherapy was the most common treatment class across lines. Over three quarters of patients (78.5% [95% CI: 78.0-78.9]) were alive one year after diagnosis (93.2% in the non-metastatic at diagnosis subgroup and 37.1% among patients with metastases at diagnosis). At three years post initial diagnosis, 18.0% patients were alive in the metastatic at diagnosis subgroup. Rapid evolution of the treatment landscape limits the results regarding lines of therapy. CONCLUSION: This large-scale study provides insight on characteristics of patients with RCC, and it highlights the need for better treatment options to improve survival.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/therapy , Male , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Female , Retrospective Studies , Aged , England/epidemiology , Middle Aged , Treatment Outcome , Protein Kinase Inhibitors/therapeutic use , Adult , Aged, 80 and over , Kaplan-Meier Estimate , Survival Rate
17.
J Transl Med ; 22(1): 368, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637886

ABSTRACT

In this study, we investigated CD70 as a promising target for renal cell carcinoma (RCC) therapy and developed a potent chimeric antigen receptor T (CAR-T) cells for potential clinical testing. CD70, found to be highly expressed in RCC tumors, was associated with decreased survival. We generated CAR-T cells expressing VHH sequence of various novel nanobodies from immunized alpaca and a single-chain variable fragment (scFv) derived from human antibody (41D12). In our in vitro experiments, anti-CD70 CAR-T cells effectively eliminated CD70-positive tumor cells while sparing CD70-negative cells. The nanobody-based CAR-T cells demonstrated significantly higher production of cytokines such as IL-2, IFN-γ and TNF-ɑ during co-culture, indicating their potential for enhanced functionality. In xenograft mouse model, these CAR-T cells exhibited remarkable anti-tumor activity, leading to the eradication of RCC tumor cells. Importantly, human T cell expansion after infusion was significantly higher in the VHH groups compared to the scFv CAR-T group. Upon re-challenging mice with RCC tumor cells, the VHH CAR-T treated group remained tumor-free, suggesting a robust and long-lasting anti-tumor response. These findings provide strong support for the potential of nanobody-based CD70 CAR-T cells as a promising therapeutic option for RCC. This warrants further development and consideration for future clinical trials and applications.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Receptors, Chimeric Antigen , Humans , Animals , Mice , Carcinoma, Renal Cell/therapy , T-Lymphocytes , Cell Line, Tumor , Kidney Neoplasms/therapy , Immunotherapy, Adoptive , Xenograft Model Antitumor Assays , CD27 Ligand
18.
World J Urol ; 42(1): 231, 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38613582

ABSTRACT

PURPOSE: To evaluate the impact of the COVID-19 pandemic on renal cell carcinoma (RCC) care in the Netherlands. METHODS: Newly diagnosed RCCs between 2018 and 2021 were selected from the Netherlands Cancer Registry; 2020-2021 was defined as COVID period and 2018-2019 as reference period. Numbers of RCCs were evaluated using 3-week-moving averages, overall and by disease stage and age. Changes in treatment were evaluated with logistic regression analyses. To evaluate possible delays in care, time to start of treatment was assessed. The cumulative number of metastatic RCC (mRCC) over time was assessed to evaluate stage shift. RESULTS: During the 1st COVID wave (weeks 9-22, 2020), the number of new RCC diagnoses decreased with 15%. Numbers restored partially in 2020, but remained 10% lower compared to 2018/2019. The decline was mostly due to a drop in T1a/T1b RCCs and in age > 70 years. 2021 showed similar numbers of new RCC diagnoses compared to 2018/2019 without an increase due to previously missed RCCs. Treatment-related changes during the 1st COVID wave were limited and temporarily; less surgery in T1a RCCs in favor of more active surveillance, and in mRCC targeted therapy was preferred over immunotherapy. Time to start of firstline treatment was not prolonged during the 1st COVID wave. No increase in mRCC was found until the end of 2021. CONCLUSIONS: The COVID-19 pandemic resulted in fewer RCC diagnoses, especially T1a/T1b tumors. Treatment-related changes appeared to be limited, temporarily and in accordance with the adapted guidelines. The diagnostic delay could lead to more advanced RCCs in later years but there are no indications for this yet.


Subject(s)
COVID-19 , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Aged , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/therapy , Delayed Diagnosis , Pandemics , COVID-19/epidemiology , Kidney Neoplasms/epidemiology , Kidney Neoplasms/therapy
19.
Sci Rep ; 14(1): 8603, 2024 04 13.
Article in English | MEDLINE | ID: mdl-38615118

ABSTRACT

Predictive biomarkers of response to immune checkpoint-based therapies (ICI) remain a critically unmet need in the management of advanced renal cell carcinoma (RCC). The complex interplay of the tumour microenvironment (TME) and the circulating immune response has proven to be challenging to decipher. MicroRNAs have gained increasing attention for their role in post-transcriptional gene expression regulation, particularly because they can have immunomodulatory properties. We evaluated the presence of immune-specific extracellular vesicle (EV) microRNAs in the plasma of patients with metastatic RCC (mRCC) prior to initiation of ICI. We found significantly lower levels of microRNA155-3p (miR155) in responders to ICI, when compared to non-responders. This microRNA has unique immunomodulatory properties, thus providing potential biological rationale for our findings. Our results support further work in exploring microRNAs as potential biomarkers of response to immunotherapy.


Subject(s)
Carcinoma, Renal Cell , Circulating MicroRNA , Kidney Neoplasms , MicroRNAs , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Immunotherapy , MicroRNAs/genetics , Biomarkers , Tumor Microenvironment/genetics
20.
BMC Urol ; 24(1): 84, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600527

ABSTRACT

BACKGROUND: Despite the rapid advances in modern medical technology, kidney renal clear cell carcinoma (KIRC) remains a challenging clinical problem in urology. Researchers urgently search for useful markers to break through the therapeutic conundrum due to its high lethality. Therefore, the study explores the value of ADH5 on overall survival (OS) and the immunology of KIRC. METHODS: The gene expression matrix and clinical information on ADH5 in the TCGA database were validated using external databases and qRT-PCR. To confirm the correlation between ADH5 and KIRC prognosis, univariate/multivariate Cox regression analysis was used. We also explored the signaling pathways associated with ADH5 in KIRC and investigated its association with immunity. RESULTS: The mRNA and protein levels showed an apparent downregulation of ADH5 in KIRC. Correlation analysis revealed that ADH5 was directly related to histological grade, clinical stage, and TMN stage (p < 0.05). Univariate and multivariate Cox regression analysis identified ADH5 as an independent factor affecting the prognosis of KIRC. Enrichment analysis looked into five ADH5-related signaling pathways. The results showed no correlation between ADH5 and TMB, TNB, and MSI. From an immunological perspective, ADH5 was found to be associated with the tumor microenvironment, immune cell infiltration, and immune checkpoints. Lower ADH5 expression was associated with greater responsiveness to immunotherapy. Single-cell sequencing revealed that ADH5 is highly expressed in immune cells. CONCLUSION: ADH5 could be a promising prognostic biomarker and a potential therapeutic target for KIRC. Besides, it was found that KIRC patients with low ADH5 expression were more sensitive to immunotherapy.


Subject(s)
Alcohol Dehydrogenase , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/therapy , Kidney , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Prognosis , RNA, Messenger , Tumor Microenvironment , Alcohol Dehydrogenase/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...