ABSTRACT
BACKGROUND: Cuproptosis, as a unique modality of regulated cell death, requires the involvement of ubiquitin-binding enzyme UBE2D2. However, the prognostic and immunotherapeutic values of UBE2D2 in pan-cancer remain largely unknown. METHODS: Using UCSC Xena, TIMER, Clinical Proteomic Tumor Analysis Consortium (CPTAC), and Human Protein Atlas (HPA) databases, we aimed to explore the differential expression pattern of UBE2D2 across multiple cancer types and to evaluate its association with patient prognosis, clinical features, and genetic variations. The association between UBE2D2 and immunotherapy response was assessed by gene set enrichment analysis, tumor microenvironment, immune gene co-expression and drug half maximal inhibitory concentration (IC50) analysis. RESULTS: The mRNA and protein levels of UBE2D2 were markedly elevated in most cancer types, and UBE2D2 exhibited prognostic significance in liver hepatocellular carcinoma (LIHC), kidney chromophobe (KICH), uveal melanomas (UVM), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), and kidney renal papillary cell carcinoma (KIRP). UBE2D2 expression was correlated with clinical features, tumor mutation burden, microsatellite instability, and anti-tumor drug resistance in several tumor types. Gene enrichment analysis showed that UBE2D2 was significantly associated with immune-related pathways. The expression level of UBE2D2 was correlated with immune cell infiltration, including CD4 + T cellsãMacrophages M2ãCD8 + T cells in pan-cancer. PDCD1, CD274 and CTLA4 expression levels were positively correlated with UBE2D2 level in multiple cancers. CONCLUSIONS: We comprehensively investigated the potential value of UBE2D2 as a prognostic and immunotherapeutic predictor for pan-cancer, providing a novel insight for cancer immunotherapy.
Subject(s)
Biomarkers, Tumor , Neoplasms , Tumor Microenvironment , Ubiquitin-Conjugating Enzymes , Humans , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/drug therapy , Immunotherapy , Female , Melanoma/genetics , Melanoma/immunology , Melanoma/drug therapy , Melanoma/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/immunology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , CTLA-4 Antigen/genetics , Uveal Neoplasms , B7-H1 AntigenABSTRACT
Tumor-associated macrophages (TAM) are abundant in several tumor types and usually correlate with poor prognosis. Previously, we demonstrated that anti-inflammatory macrophages (M2) inhibit NK cell effector functions. Here, we explored the impact of TAM on NK cells in the context of clear-cell renal cell carcinoma (ccRCC). Bioinformatics analysis revealed that an exhausted NK cell signature strongly correlated with an M2 signature. Analysis of TAM from human ccRCC samples confirmed that they exhibited an M2-skewed phenotype and inhibited IFN-γ production by NK cells. Moreover, human M0 macrophages cultured with conditioned media from ccRCC cell lines generated macrophages with an M2-skewed phenotype (TAM-like), which alike TAM, displayed suppressive activity on NK cells. Moreover, TAM depletion in the mouse Renca ccRCC model resulted in delayed tumor growth and reduced volume, accompanied by an increased frequency of IFN-γ-producing tumor-infiltrating NK cells that displayed heightened expression of T-bet and NKG2D and reduced expression of the exhaustion-associated co-inhibitory molecules PD-1 and TIM-3. Therefore, in ccRCC, the tumor microenvironment polarizes TAM toward an immunosuppressive profile that promotes tumor-infiltrating NK cell dysfunction, contributing to tumor progression. In addition, immunotherapy strategies targeting TAM may result in NK cell reinvigoration, thereby counteracting tumor progression.
Subject(s)
Carcinoma, Renal Cell , Interferon-gamma , Kidney Neoplasms , Killer Cells, Natural , Tumor-Associated Macrophages , Killer Cells, Natural/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Humans , Animals , Mice , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Disease Progression , Cell Line, Tumor , Tumor Microenvironment/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/immunology , Programmed Cell Death 1 Receptor/metabolismABSTRACT
The complete regression of clear cell renal cell carcinoma (ccRCC) obtained pre-clinically with anti-carbonic anhydrase IX (CAIX) G36 chimeric antigen receptor (CAR) T cells in doses equivalent to â 108 CAR T cells/kg renewed the potential of this target to treat ccRCC and other tumors in hypoxia. The immune checkpoint blockade (ICB) brought durable clinical responses in advanced ccRCC and other tumors. Here, we tested CD8α/4-1BB compared to CD28-based anti-CAIX CAR peripheral blood mononuclear cells (PBMCs) releasing anti-programmed cell death ligand-1 (PD-L1) IgG4 for human ccRCC treatment in vitro and in an orthotopic NSG mice model in vivo. Using a â 107 CAR PBMCs cells/kg dose, anti-CAIX CD28 CAR T cells releasing anti-PD-L1 IgG highly decrease both tumor volume and weight in vivo, avoiding the occurrence of metastasis. This antitumoral superiority of CD28-based CAR PBMCs cells compared to 4-1BB occurred under ICB via PD-L1. Furthermore, the T cell exhaustion status in peripheral CD4 T cells, additionally to CD8, was critical for CAR T cells efficiency. The lack of hepatotoxicity and nephrotoxicity upon the administration of a 107 CAR PMBCs cells/kg dose is the basis for carrying out clinical trials using anti-CAIX CD28 CAR PBMCs cells releasing anti-PD-L1 antibodies or anti-CAIX 4-1BB CAR T cells, offering exciting new prospects for the treatment of refractory ccRCC and hypoxic tumors.
Subject(s)
B7-H1 Antigen , Carbonic Anhydrase IX , Carcinoma, Renal Cell , Kidney Neoplasms , Receptors, Chimeric Antigen , Animals , Antibodies/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD28 Antigens , Carbonic Anhydrase IX/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Immune Checkpoint Inhibitors , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Leukocytes, Mononuclear/pathology , Mice , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunologyABSTRACT
Natural Killer (NK) cells play a key role in cancer immunosurveillance. However, NK cells from cancer patients display an altered phenotype and impaired effector functions. In addition, evidence of a regulatory role for NK cells is emerging in diverse models of viral infection, transplantation, and autoimmunity. Here, we analyzed clear cell renal cell carcinoma (ccRCC) datasets from The Cancer Genome Atlas (TCGA) and observed that a higher expression of NK cell signature genes is associated with reduced survival. Analysis of fresh tumor samples from ccRCC patients unraveled the presence of a high frequency of tumor-infiltrating PD-L1+ NK cells, suggesting that these NK cells might exhibit immunoregulatory functions. In vitro, PD-L1 expression was induced on NK cells from healthy donors (HD) upon direct tumor cell recognition through NKG2D and was further up-regulated by monocyte-derived IL-18. Moreover, in vitro generated PD-L1hi NK cells displayed an activated phenotype and enhanced effector functions compared to PD-L1- NK cells, but simultaneously, they directly inhibited CD8+ T cell proliferation in a PD-L1-dependent manner. Our results suggest that tumors might drive the development of PD-L1-expressing NK cells that acquire immunoregulatory functions in humans. Hence, rational manipulation of these regulatory cells emerges as a possibility that may lead to improved anti-tumor immunity in cancer patients.
Subject(s)
B7-H1 Antigen/biosynthesis , CD8-Positive T-Lymphocytes/cytology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Killer Cells, Natural/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Datasets as Topic , Disease-Free Survival , Gene Expression , Humans , Interferon-gamma/pharmacology , Interleukin-18/pharmacology , K562 Cells , Kaplan-Meier Estimate , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Monitoring, Immunologic , Monocytes/metabolism , Recombinant Proteins/pharmacology , Up-RegulationABSTRACT
Our study investigated the underlying mechanism for the 14q24 renal cell carcinoma (RCC) susceptibility risk locus identified by a genome-wide association study (GWAS). The sentinel single-nucleotide polymorphism (SNP), rs4903064, at 14q24 confers an allele-specific effect on expression of the double PHD fingers 3 (DPF3) of the BAF SWI/SNF complex as assessed by massively parallel reporter assay, confirmatory luciferase assays, and eQTL analyses. Overexpression of DPF3 in renal cell lines increases growth rates and alters chromatin accessibility and gene expression, leading to inhibition of apoptosis and activation of oncogenic pathways. siRNA interference of multiple DPF3-deregulated genes reduces growth. Our results indicate that germline variation in DPF3, a component of the BAF complex, part of the SWI/SNF complexes, can lead to reduced apoptosis and activation of the STAT3 pathway, both critical in RCC carcinogenesis. In addition, we show that altered DPF3 expression in the 14q24 RCC locus could influence the effectiveness of immunotherapy treatment for RCC by regulating tumor cytokine secretion and immune cell activation.
Subject(s)
Carcinoma, Renal Cell/genetics , Chromosomes, Human, Pair 14 , DNA-Binding Proteins/genetics , Genetic Loci , Kidney Neoplasms/genetics , STAT3 Transcription Factor/genetics , Transcription Factors/genetics , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Chromatin/chemistry , Chromatin/immunology , Chromatin Assembly and Disassembly/immunology , Cytokines/genetics , Cytokines/immunology , DNA-Binding Proteins/immunology , Gene Expression Regulation , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Polymorphism, Single Nucleotide , STAT3 Transcription Factor/immunology , T-Lymphocytes, Cytotoxic , Transcription Factors/immunologySubject(s)
Humans , Aged , Aged, 80 and over , Age Factors , Kidney Neoplasms/immunology , Adaptive Immunity , Immunity, InnateABSTRACT
BACKGROUND: Recent studies have observed an association between immune-related adverse events (irAE) and favorable clinical outcomes in the setting of cancer treatment with immune checkpoint inhibitors (ICI). However, results have been variable and inconclusive. Therefore, we have conducted a pan-cancer meta-analysis evaluating the relationship between irAEs and clinical outcomes. MATERIALS AND METHODS: The search included studies published in PubMed, Embase, and Web of Science from conception to 12.28.2019 as well as abstracts published in the ASCO and ESMO meetings from 2015 to 2019. Studies were included if ICI was used in advanced or metastatic cancer settings and excluded if data contained only combination therapy regimens or contained anti-CTLA-4. Raw data for overall response rate (ORR), hazard ratios (HR), number of patients (n), and p values for overall survival (OS) and progression-free survival (PFS) were extracted. Pooled sensitivity (SN), specificity (SP), positive (PPV) and negative predictive values (NPV), and odds ratios (ORs) were calculated using the 2 × 2 table and logit transformed proportions; and summary receiver operating curve (sROC) was generated using the bivariate approach for ORR. Pooled HRs were calculated using the means weighted by inverse of the variance for OS and PFS. Heterogeneity was assumed and random effects model was used throughout the analyses. RESULTS: Final analysis included 32 studies, among which ORR data were available in 15 studies, OS in 17, and PFS in 16. 17 studies evaluated non-small cell lung cancer (NSCLC), two studies melanoma, one study gastric cancer, three studies renal cell carcinoma (RCC), seven studies various cancer types, two studies urothelial carcinoma, and one study head and neck cancer (HNSCC). With respect to ORR, pooled SN, SP, PPV and NPV, and OR were 0.522 [0.423-0.619], 0.810 [0.771-0.844], 0.516 [0.413-0.618], 0.819 [0.764-0.864], and 4.59 [3.24-6.50], respectively. The area under the curve (AUC) derived from the sROC was 0.773. HR for OS and PFS were 0.47 [95% CI 0.37-0.60] and 0.46 [95% CI 0.37-0.56], respectively. Between-study publication bias was present for ORR, OS, and PFS; however, results remained significant after trim-fill analysis. CONCLUSION: irAEs predict OR, OS, and PFS across different types of cancer and may represent useful biomarkers in the clinical setting.
Subject(s)
Immune Checkpoint Inhibitors/adverse effects , Neoplasms/therapy , Area Under Curve , B7-H1 Antigen/antagonists & inhibitors , Bias , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/therapy , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/therapy , Humans , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/immunology , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Leukocyte L1 Antigen Complex/immunology , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/therapy , Melanoma/immunology , Melanoma/mortality , Melanoma/therapy , Neoplasms/immunology , Neoplasms/mortality , Progression-Free Survival , Sensitivity and Specificity , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Stomach Neoplasms/therapy , Treatment OutcomeABSTRACT
A 56-year-old white man with a 74 pack-year smoking history presented with macroscopic hematuria and a significant weight loss of 45 pounds in 6 months. His clinical laboratory tests indicated iron defi ciency anemia and a computed tomography (CT) scan showed a left kidney tumor, mediastinal lymph nodes, and multiple lung metastases. A percutaneous CT-guided kidney biopsy revealed grade 3 clear cell renal carcinoma based on World Health Organization/International Society of Urologic Pathology classifi cation. The patient started first line systemic treatment for intermediate-risk metastatic renal cell carcinoma (mRCC) with combination immunotherapy with nivolumab plus ipilimumab.1 After 10 days of the first cycle, he presented with a pruritic maculopapular rash covering 20% of his body surface.
Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Anti-Inflammatory Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Humans , Immunotherapy/methods , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/pathology , Nivolumab/administration & dosage , Nivolumab/adverse effects , Prednisone/administration & dosage , Treatment OutcomeABSTRACT
BACKGROUND: Clear cell renal cell carcinoma (ccRCC), the most aggressive renal cancer, is characterized by early lymph node metastases and bad prognosis. Most therapies targeting advanced or metastatic ccRCC are based, as first-line treatment, on the administration of the vascular endothelial growth factor (VEGF) neutralizing antibody termed Bevacizumab. Despite proven benefits, the expected results were not obtained for the majority of patients. The possibility that an intricate interplay between angiogenesis and immune-checkpoints might exist lead us to evaluate tumor angiogenesis, by means of VEGF expression together with the immune checkpoint HLA-G/ILT4. METHODS: Tumor specimens were obtained from patients from two separate cohorts: One from "Evita Pueblo" Hospital from Berazategui, (Buenos Aires, Argentina) and the second includes patients surgically operated at the Urology Department of Saint-Louis Hospital (Paris, France) with a confirmed ccRCC diagnosis. Immunohistochemistry was performed with specific antibodies directed against HLA-G, VEGF-A, VEGF-C, D240, CD34, ILT4 and Ca-IX. In addition, gene expression levels were measured in a cell line derived from a ccRCC patient by semi-quantitative RT-PCR. RESULTS: Our results show that the highly vascularized tumors of ccRCC patients express high levels of VEGF and the immune-checkpoint HLA-G. In addition, ILT4, one of the HLA-G receptors, was detected on macrophages surrounding tumor cells, suggesting the generation of an immune-tolerant microenvironment that might favor tumorigenesis. Notably, RT-qPCR analysis provided the first evidence on the transcriptional relationship between HLA-G/ILT4 and the VEGF family. Namely, in the presence of HLA-G or ILT4, the levels of VEGF-A are diminished whereas those of VEGF-C are increased. CONCLUSIONS: In an effort to find new therapeutic molecules and fight against metastasis dissemination associated with the poor survival rates of ccRCC patients, these findings provide the rationale for co-targeting angiogenesis and the immune checkpoint HLA-G.
Subject(s)
Carcinoma, Renal Cell/genetics , HLA-G Antigens/metabolism , Kidney Neoplasms/genetics , Membrane Glycoproteins/metabolism , Neovascularization, Pathologic/genetics , Receptors, Immunologic/metabolism , Vascular Endothelial Growth Factor A/genetics , Adult , Aged , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Kidney/blood supply , Kidney/pathology , Kidney/surgery , Kidney Neoplasms/immunology , Kidney Neoplasms/mortality , Kidney Neoplasms/therapy , Male , Membrane Glycoproteins/antagonists & inhibitors , Middle Aged , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Nephrectomy , Receptors, Immunologic/antagonists & inhibitors , Retrospective Studies , Survival Rate , Vascular Endothelial Growth Factor A/antagonists & inhibitorsABSTRACT
Metastatic renal cell carcinoma (mRCC) encompasses a heterogeneous group of neoplasms with distinct clinical behavior and prognoses. As a result of the increasing number of therapeutic options in the metastatic setting, it is crucial to improve prognostic stratification ability. We aimed to evaluate the prognostic value of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and combination platelet count and neutrophil lymphocyte ratio (COP-NLR) in patients with mRCC. We evaluated a cohort of mRCC patients treated with first-line pazopanib or sunitinib. Levels of NLR, PLR and COP-NLR were measured prior to systemic treatment and evaluated as prognostic predictors. Primary endpoint was overall survival (OS). Data from 276 patients were included, of which 54.7% received first-line pazopanib and 45.3%, sunitinib. Memorial Sloan-Kettering Cancer Center risk classification was intermediate and poor in 50% and 42.6% of patients, respectively. High NLR (> 3.5) was associated with inferior OS (median 9.6 vs 17.8 months, P < 0.001). A high PLR (> 200) was associated with inferior OS (median 10.3 vs 17 months, P = 0.002). The median OS in the COP-NLR 1, 2 and 3 groups were 19.0 months (95% CI 15.3-26.0), 13.1 months (95% CI 9.8-17.0) and 7.4 months (95% CI 3.6-11.9), respectively (P < 0.001). In the multivariate analysis, high NLR and high COP-NLR were associated with inferior OS. Both high NLR and high COP-NLR were associated with poorer OS in our cohort of patients with mRCC treated with first-line pazopanib or sunitinib.
Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/drug therapy , Child , Female , Humans , Indazoles/therapeutic use , Inflammation/blood , Kidney Neoplasms/drug therapy , Lymphocyte Count , Male , Middle Aged , Neutrophils , Platelet Count , Prognosis , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Sunitinib/therapeutic use , Young AdultABSTRACT
Combination treatments with immuno-oncology (IO) agents and IO agents plus a vascular endothelial growth factor receptor tyrosine kinase inhibitor (VEGFR-TKI) have been approved for first-line treatment of patients with metastatic renal cell carcinoma (mRCC). No direct comparisons have been performed among these treatment options. We performed a systematic review and network meta-analysis to compare and rank the available regimens for first-line treatment in terms of survival benefit and efficacy. In accordance with the Preferred Reporting Items for Systematic Review statement, a systematic search of reported studies was performed in MEDLINE, the Cochrane Central Register of Controlled Trials, and EMBASE up to May 31, 2019. Network meta-analysis models were adjusted using the Bayesian method. Four randomized clinical trials, with a total of 3758 patients, met the inclusion criteria. Considering systemic therapy, 1880 patients had received sunitinib and 550, 432, 442, and 454 patients had received ipilimumab plus nivolumab (ipi + nivo), pembrolizumab plus axitinib (pembro + axi), avelumab plus axitinib (avelu + axi), and atezolizumab plus bevacizumab (atezo + bev). No difference was found in overall survival between ipi + nivo and pembro + axi for the intention to treat population (hazard ratio [HR], 1.34; 95% credible interval [CrI], 0.92-1.97). No difference was found in progression-free survival among the treatments. The overall response rate (ORR) was superior with pembro + axi and avelu + axi compared with the ORR with the other treatments (atezo + bev vs. pembro + axi: HR, 0.66; 95% CrI, 0.52-0.84; ipi + nivo vs. pembro + axi: HR, 0.73; 95% CrI, 0.59-0.90; atezo + bev vs. avelu + axi: HR, 0.55; 95% CrI, 0.43-0.71; avelu + axi vs. ipi + nivo: HR, 1.66; 95% CrI, 1.31-2.12), with no differences across them (HR, 1.21; 95% CrI, 0.95-1.53). In the present indirect comparison, for an intention to treat population, we found no survival differences between pembro + axi and ipi + nivo. All treatments showed better progression-free survival compared with sunitinib that was similar among them. The combination of an IO agent (pembrolizumab or avelumab) and axitinib seemed to be the most effective therapy for the ORR.
Subject(s)
Carcinoma, Renal Cell/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Prognosis , Survival RateABSTRACT
BACKGROUND (OR PURPOSE): Nivolumab has been shown to be effective for the treatment of second-line mRCC. The present study has investigated the effectiveness and safety of nivolumab in real-world Eastern Spanish patients with advanced mRCC at TKI progression. PATIENTS AND METHODS: A retrospective review of mRCC patients treated with nivolumab as a second-line treatment was performed. Analyzed variables included age, sex, ECOG (quality of life scale designed by the Eastern Cooperative Oncology Group), histology, nephrectomy, location of metastases, number of metastasis locations, previous treatments, analytical data from the standard blood count and biochemistry, and response to treatment. RESULTS: 98 patients from 18 sites in Spain were retrospectively reviewed. The majority of patients were male (75%), had ECOG 0-1 (90.6%), had no brain metastasis (91.4%), had undergone one prior systemic regimen (94.3%), and were current/former smokers (97.1%). Fourteen patients (13.1%) had non-clear cell histology, seven (7.1%) had poor-IMDC prognostic group characteristics, 13 patients (13.1%) had liver metastasis and 35 (35.7%) had bone lesions. All patients received prior systemic therapy (63.3% sunitinib, 34.7% pazopanib). During the study, a median of eight doses of nivolumab was given (range 2-62) and 11 patients received more than 12 doses. Eleven patients (11.2%) received nivolumab as a third or fourth line of treatment. Median duration of therapy was 3.6 months (range 0.5-29.3). Confirmed response rate was 25%. Median progression free survival was 7.8 months (range 1.2-12.1). Median overall survival was 16.3 months (range 1.7-29.3). After discontinuation of treatment, 27.58% of the patients received subsequent systemic cancer therapy. Side effects were mostly grade 1-2 (7.2% had hypothyroidism and 6.2% liver toxicity, 4% had nephritis and 2% hypophysitis). Two cases of grade 3-4 adverse events (2%) were reported. CONCLUSION: Benefit/risk profile of nivolumab in Eastern-Spanish real-world population with mRCC after tyrosine-kinase inhibitors was consistent with prior real-life studies reported as well as pivotal study.
Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Nivolumab/therapeutic use , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Humans , Kidney Neoplasms/epidemiology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Retrospective Studies , Spain/epidemiology , Survival RateABSTRACT
PURPOSE: Considering the recent publication of the results of several clinical trials for metastatic clear cell renal cell carcinoma (mRCC), we performed a systematic review and meta-analysis of randomized studies comparing standard first-line VEGFR-targeted therapy to immune checkpoint inhibitors-based combinations for mRCC patients. METHODS: 3960 patients from 5 randomized clinical trials where available for evaluation. RESULT: In the all-comers population, immunotherapy-based combinations were able to decrease the risk of death over the standard of care by 26% (HR 0.74; 95% CI 0.60-0.92; p = 0.006), to decrease the risk of progression by 21% (HR 0.79; 95% CI 0.72-0.86; p < 0.00001), and to increase the relative risk of response by 40% (HR 1.40; 95% CI 1.11-1.77; p = 0.006). For poor/intermediate-risk patients, the risk of death is decreased by 41% and the risk of progression by 27%. CONCLUSIONS: The benefit of immunotherapy-based combinations in mRCC patients is independent from the IMDC risk group, but it is stronger for poor/intermediate-risk patients.
Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/therapy , Kidney Neoplasms/therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Randomized Controlled Trials as Topic , Survival Rate , Treatment OutcomeABSTRACT
Patients with non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC) have shown benefit from anti-PD-1 therapies. However, not all patients experience tumor shrinkage, durable responses or prolonged survival, demonstrating the need to find response markers. In blood samples from NSCLC and RCC patients obtained before and after anti-PD-1 treatment, we studied leukocytes by complete blood cell count, lymphocyte subsets using flow cytometry and plasma concentration of nine soluble mediators, in order to find predictive biomarkers of response and to study changes produced after anti-PD-1 therapy. In baseline samples, discriminant analysis revealed a combination of four variables that helped differentiate stable disease-response (SD-R) from progressive disease (PD) patients: augmented frequency of central memory CD4+ T cells and leukocyte count was associated with response while increased percentage of PD-L1+ natural killer cells and naïve CD4+ T cells was associated with lack of response. After therapy, differential changes between responders and non-responders were found in leukocytes, T cells and TIM-3+ T cells. Patients with progressive disease showed an increase in the frequency of TIM-3 expressing CD4+ and CD8+ T cells, whereas SD-R patients showed a decrease in these subsets. Our findings indicate that a combination of immune variables from peripheral blood (PB) could be useful to distinguish response groups in NSCLC and RCC patients treated with anti-PD-1 therapy. Frequency of TIM-3+ T cells showed differential changes after treatment in PD vs SD-R patients, suggesting that it may be an interesting marker for monitoring progression during therapy.
Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Aged , C-Reactive Protein/analysis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Renal Cell/immunology , Female , Hepatitis A Virus Cellular Receptor 2/blood , Humans , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Male , Middle AgedABSTRACT
PURPOSE: Although overexpression of the eukaryotic translation initiation factor 4E (eIF4E) is detected in patients with renal cell carcinoma (RCC) and associated with poor prognosis, the possible roles of eIF4E in RCC have not been revealed. METHODS: The effects of eIF4E inhibition on cell growth, migration, survival, chemo-/immunotherapy and eIF4E pathways via pharmacological inhibitor and genetic siRNA knockdown were analyzed in RCC cells. RESULTS: In this work, we demonstrate that eIF4E is critically involved in multiple biological functions of RCC. We firstly inhibited eIF4E activity by ribavirin in two cell lines (Caki-1 and ACHN) representing RCC metastasis models. We demonstrated that ribavirin inhibited proliferation and migration and induced apoptosis in RCC in a dose-dependent manner. We further confirmed that the inhibitory effects of ribavirin were attributed to its ability in inhibiting eIF4E-regulated protein translation and activity. eIF4E inhibition using siRNA knockdown mimicked ribavirin's effector in RCC cells. Importantly, eIF4E inhibition by both ribavirin and siRNA knockdown significantly sensitized RCC response to chemo- and immunotherapeutic agents in vitro as well as in vivo. CONCLUSIONS: Our findings clearly demonstrate the roles of eIF4E in RCC growth, survival, metastasis and resistance. Ribavirin is an antiviral drug, and its clinical efficacy is currently being investigated in the treatment of various cancers. Our findings support and provide a preclinical evidence for clinical trial for the combination of ribavirin with chemo-/immunotherapy in RCC.
Subject(s)
Carcinoma, Renal Cell/therapy , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Immunotherapy , Kidney Neoplasms/therapy , RNA, Small Interfering/genetics , Ribavirin/pharmacology , Animals , Antimetabolites/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Cell Proliferation/drug effects , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor AssaysABSTRACT
The immune system regulates angiogenesis in cancer by way of both pro- and antiangiogenic activities. A bidirectional link between angiogenesis and the immune system has been clearly demonstrated. Most antiangiogenic molecules do not inhibit only VEGF signaling pathways but also other pathways which may affect immune system. Understanding of the role of these pathways in the regulation of immunosuppressive mechanisms by way of specific inhibitors is growing. Renal cell carcinoma (RCC) is an immunogenic tumor in which angiogenesis and immunosuppression work hand in hand, and its growth is associated with impaired antitumor immunity. Given the antitumor activity of selected TKIs in metastatic RCC (mRCC), it seems relevant to assess their effect on the immune system. The confirmation that TKIs improve cell cytokine response in mRCC provides a basis for the rational combination and sequential treatment of TKIs and immunotherapy.
Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Immunotherapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , HumansABSTRACT
With the enormous and growing interest in the clinical application of immunotherapy, we are currently facing the need to accurately monitor the immune function of cancer patients. Here, we describe changes in the immune status of a patient with metastatic type-2-papillary renal cell carcinoma, before and after surgery and subsequent immunotherapy with a dendritic cell-tumor cell hybrid vaccine. Through the accurate assessment of monocyte-derived dendritic cells (Mo-DCs) function, we show that Mo-DCs were freed from tumor-induced maturation blockage by tumor resection surgery, while Mo-DCs-tumor induced suppression and anergy were only interrupted by the vaccination treatment. Our data suggest that the evaluation of Mo-DCs' function may provide a powerful and precise tool to monitor immune restoration in cancer patients.
Subject(s)
Cancer Vaccines/immunology , Carcinoma, Renal Cell/therapy , Dendritic Cells/physiology , Immunotherapy/methods , Kidney Neoplasms/therapy , Monitoring, Immunologic , T-Lymphocytes, Regulatory/immunology , Adult , Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/transplantation , Humans , Immunosuppression Therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Male , Tumor Escape/drug effects , VaccinationABSTRACT
This study aimed to explore the clinical value of the CD4(+) T cell ATP levels in patients with renal cell carcinoma through the application of the ImmuKnow(TM)-Cylex(®) assay. We recruited 104 patients with renal cancer who had undergone surgery at Fuzhou General Hospital from March 2009 to June 2012, and were subsequently treated by dendritic cell and cytokine-induced killer cell bio-therapy or interferon-α therapy. The changes in CD4(+) T cell ATP levels were detected at the perioperative period and at 10 days, 1 month, 3 months, and 1 year after the surgery using the ImmuKnow assay. In addition, the differences in ATP levels in different therapy groups were compared and the prognosis conditions were analyzed. Our results demonstrated that no significant difference in the ATP levels occurred at different time points; furthermore, there were no obviously different ATP levels between the different therapy groups, and the ATP levels were found to have no clinical significance for the assessment of renal cancer prognosis. Overall, this study suggested that CD4(+) T cell ATP levels as detected by the ImmuKnow assay have no obvious clinical value in patients with renal cancer.
Subject(s)
Carcinoma, Renal Cell/immunology , Immunoassay/methods , Kidney Neoplasms/immunology , Adenosine Triphosphate/metabolism , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes , Carcinoma, Renal Cell/drug therapy , Cytokine-Induced Killer Cells/immunology , Female , Humans , Interferon-alpha/therapeutic use , Kaplan-Meier Estimate , Male , Middle Aged , PrognosisABSTRACT
AbstractObjective: to analyze the process of tissue repair in patients with venous ulcers using inelastic compression therapy (the Unna Boot), in comparison with the use of the elastic bandage.Method: a controlled randomized clinical trial in which the patients (n=18) were allocated to two groups, those who used the Unna Boot (group B) and those who used the elastic bandage (group A). The study's follow-up period was 13 weeks.Results: a significant reduction took place, at the level of 5%, in the area, in square centimeters, of the ulcers of group B (p<0.0001) throughout the treatment, and there was a tendency of group A for reduction in the area of the ulcer, in centimeters squared (p=0.06), only after the fifth week.Conclusion: the treatment with the Unna Boot presented better results in venous ulcers with areas over 10cm², and the elastic bandage with Petrolatum(r) gauze in venous ulcers below 10cm². Brazilian Clinical Trials Register: Trial (req: 195) and WHO UTN U1111-1122-5489.
ResumoObjetivo:analisar o processo de reparo tecidual de pacientes com úlcera venosa em uso da terapia compressiva inelástica (Bota de Unna), em comparação ao uso da bandagem elástica.Método:ensaio clínico controlado randomizado em que os pacientes (n=18) foram alocados em dois grupos, os que utilizavam a Bota de Unna (grupo B) e os que utilizavam a atadura elástica (grupo A). O tempo de seguimento da pesquisa foi de treze semanas.Resultados:ocorreu redução significativa, no nível de 5%, na área, em centímetros quadrados, das úlceras do grupo B (p<0,0001) ao longo de todo o tratamento, e tendência do grupo A à redução, na área da úlcera, em centímetros quadrados (p=0,06), apenas após a quinta semana.Conclusão:o tratamento com a Bota de Unna apresentou melhor resultado em úlceras venosas com áreas superiores a 10cm², e a atadura elástica com a gaze Petrolatum(r)em úlceras venosas inferiores a 10cm². Registro Brasileiro de Ensaios Clínicos: Trial (req: 195) e WHO UTN U1111-1122-5489.
ResumenObjetivo:analizar el proceso de reparación del tejido de pacientes con úlcera venosa que usan la terapia compresiva inelástica (Bota de Unna), en comparación con el uso del vendaje elástico.Método:ensayo clínico controlado aleatorio en que los pacientes (n=18) fueron designados en dos grupos, los que utilizaban la Bota de Unna (grupo B) y los que utilizaban el vendaje elástico (grupo A). El tiempo de duración de la investigación fue de trece semanas.Resultados:se constató reducción significativa, al nivel de 5%, en el área, en centímetros cuadrados, de las úlceras del grupo B (p<0,0001) a lo largo de todo el tratamiento; y tendencia del grupo A a la reducción, en el área de la úlcera, en centímetros cuadrados (p=0,06), solamente después de la quinta semana.Conclusión:el tratamiento con la Bota de Unna presentó mejor resultado en úlceras venosas con áreas superiores a 10cm², y el vendaje elástico con la gasa Petrolatum(r)en úlceras venosas inferiores a 10cm². Registro Brasileño de Ensayos Clínicos: Trial (req: 195) y WHO UTN U1111-1122-5489.
Subject(s)
Animals , Female , Mice , Graft vs Host Disease , Kidney Neoplasms , Lymphocyte Transfusion , Stem Cell Transplantation , Allografts , Cell Line, Tumor , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasm MetastasisABSTRACT
We investigated the expression and clinical value of the soluble major histocompatibility complex class I-related chain A (sMICA) molecule in the serum of patients with renal tumors. Sixty patients diagnosed with renal tumors were enrolled in the experimental group, whereas 20 healthy volunteers served as the control group. The sMICA levels were measured via enzyme-linked immunosorbent assay, and the results were analyzed in combination with data from pathol-ogy examination. The experimental group had a statistically significant higher sMICA level (P < 0.05) than the control group. The sMICA level was higher in patients with malignant tumors than in those with be-nign tumors. We also observed a positive relationship among different tumor-node-metastasis (TNM) pathological stages with more advanced diseases exhibiting higher sMICA levels. As a tumor-associated antigen, MICA has a close relationship with renal tumorigenesis and immune es-cape. Our results indicated that sMICA levels were related to tumor pathol-ogy, TNM stage, and metastasis. Therefore, sMICA might be a potential marker for tumor characteristics, prognosis, and recurrence prediction.