Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 666
Filter
1.
J Pathol Clin Res ; 10(4): e12388, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38923836

ABSTRACT

Tumor structure is heterogeneous and complex, and it is difficult to obtain complete characteristics by two-dimensional analysis. The aim of this study was to visualize and characterize volumetric vascular information of clear cell renal cell carcinoma (ccRCC) tumors using whole tissue phenotyping and three-dimensional light-sheet microscopy. Here, we used the diagnosing immunolabeled paraffin-embedded cleared organs pipeline for tissue clearing, immunolabeling, and three-dimensional imaging. The spatial distributions of CD34, which targets blood vessels, and LYVE-1, which targets lymphatic vessels, were examined by calculating three-dimensional density, vessel length, vessel radius, and density curves, such as skewness, kurtosis, and variance of the expression. We then examined those associations with ccRCC outcomes and genetic alteration state. Formalin-fixed paraffin-embedded tumor samples from 46 ccRCC patients were included in the study. Receiver operating characteristic curve analyses revealed the associations between blood vessel and lymphatic vessel distributions and pathological factors such as a high nuclear grade, large tumor size, and the presence of venous invasion. Furthermore, three-dimensional imaging parameters stratified ccRCC patients regarding survival outcomes. An analysis of genomic alterations based on volumetric vascular information parameters revealed that PI3K-mTOR pathway mutations related to the blood vessel radius were significantly different. Collectively, we have shown that the spatial elucidation of volumetric vasculature information could be prognostic and may serve as a new biomarker for genomic alterations. High-end tissue clearing techniques and volumetric immunohistochemistry enable three-dimensional analysis of tumors, leading to a better understanding of the microvascular structure in the tumor space.


Subject(s)
Carcinoma, Renal Cell , Imaging, Three-Dimensional , Kidney Neoplasms , Microvessels , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/blood supply , Kidney Neoplasms/diagnostic imaging , Female , Male , Microvessels/pathology , Middle Aged , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Adult , Prognosis
2.
Oncogene ; 43(18): 1386-1396, 2024 May.
Article in English | MEDLINE | ID: mdl-38467852

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) presents a unique profile characterized by high levels of angiogenesis and robust vascularization. Understanding the underlying mechanisms driving this heterogeneity is essential for developing effective therapeutic strategies. This study revealed that ubiquitin B (UBB) is downregulated in ccRCC, which adversely affects the survival of ccRCC patients. UBB exerts regulatory control over vascular endothelial growth factor A (VEGFA) by directly interacting with specificity protein 1 (SP1), consequently exerting significant influence on angiogenic processes. Subsequently, we validated that DNA methyltransferase 3 alpha (DNMT3A) is located in the promoter of UBB to epigenetically inhibit UBB transcription. Additionally, we found that an unharmonious UBB/VEGFA ratio mediates pazopanib resistance in ccRCC. These findings underscore the critical involvement of UBB in antiangiogenic therapy and unveil a novel therapeutic strategy for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Down-Regulation , Gene Expression Regulation, Neoplastic , Kidney Neoplasms , Neovascularization, Pathologic , Sp1 Transcription Factor , Vascular Endothelial Growth Factor A , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Humans , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Kidney Neoplasms/drug therapy , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Line, Tumor , Animals , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Indazoles/pharmacology , Indazoles/therapeutic use , DNA Methyltransferase 3A/metabolism , Sulfonamides/pharmacology , Mice , Ubiquitin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , Drug Resistance, Neoplasm/genetics , Promoter Regions, Genetic , Female , Male , Angiogenesis
3.
Jpn J Radiol ; 42(6): 648-655, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38388790

ABSTRACT

PURPOSE: To retrospectively evaluate the depiction rate of feeding arteries in biopsy-proven clear cell renal cell carcinoma (CCRCC) on four-dimensional computed tomography angiography (4D-CTA) images. MATERIALS AND METHODS: This study included 22 patients with 22 CCRCC and 30 feeding arteries treated with transcatheter renal artery embolization. The depiction rate of the feeding arteries on preprocedural 4D-CTA was evaluated. Images were acquired by 320-row multi-detector computed tomography (CT) 15‒36 s after starting to inject a contrast agent (600 mg/kg iodine) intravenously into patients at 2.1 s intervals (11 phases). Two board-certified radiologists retrospectively assessed the feeder depiction rate in all 11 phases with reference to the procedural images as the gold standard. Discrepancies were resolved by consultation with a third radiologist. RESULTS: Among the feeders, 11 (36.7%) were segmental or lobar, and 19 (63.3%) were interlobar or arcuate arteries. The feeder depiction rate was the highest (25 [83.3%] of 30) in the 5th phase (delay, 23.4 s) where the gap in contrast enhancement between the renal artery and cortex was the largest. This was followed by the 6th (23 [76.7%] of 30), 4th (22 [73.3%] of 30]), and 7th (21 [70.0%] of 30) phases. The overall rate of depicting feeding arteries in the 11 phases of 4D-CTA was 28 (93.3%) of 30. CONCLUSIONS: The depiction rate of CCRCC feeding arteries including lobar or smaller artery branches by 4D-CTA was favorable. The feeding arteries were optimally visualized during the phase with the largest contrast gap between the renal artery and cortex.


Subject(s)
Carcinoma, Renal Cell , Computed Tomography Angiography , Contrast Media , Four-Dimensional Computed Tomography , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/blood supply , Female , Male , Retrospective Studies , Middle Aged , Aged , Computed Tomography Angiography/methods , Aged, 80 and over , Four-Dimensional Computed Tomography/methods , Adult , Renal Artery/diagnostic imaging , Kidney/diagnostic imaging , Kidney/blood supply , Embolization, Therapeutic/methods
4.
Diagn Interv Imaging ; 103(11): 510-515, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35934617

ABSTRACT

PURPOSE: The purpose of this study was to report the technical feasibility and outcomes of percutaneous image-guided cryoablation with temporary balloon occlusion of the renal artery for the treatment of central renal tumors. MATERIALS AND METHODS: All consecutive patients with central renal tumors treated with cryoablation and temporary renal artery occlusion from January 2017 to October 2021 were retrospectively included. Patient demographics, tumor's characteristics, procedural data, technical success, primary and secondary clinical efficacy, complications (according to Cardiovascular and Interventional Radiology Society of Europe [CIRSE] classification) and follow-up were investigated. RESULTS: A total of 14 patients (8 men, 6 women; mean age 72.4 years ± 21.4 [SD] years; age range: 42-93 years) with 14 central renal tumors (median size, 32 mm; IQR: 23.5, 39.5 mm; range: 13-50 mm) were treated with percutaneous image-guided cryoablation and temporary balloon occlusion of the renal artery. Technical success was 13/14 (93%), with 1/14 (7%) failure of vascular access. A median of 4 cryoprobes (IQR: 3, 4.75) were inserted and protective hydrodissection was performed in 11/14 (79%) patients. Median time to perform cryoprobes insertion, hydrodissection and vascular access was 26.5 min (IQR: 18, 35 min), 10 min (IQR: 10, 17 min) and 30 min (IQR: 20, 45 min) respectively. Median duration of the whole intervention was 150 min (IQR: 129, 180 min; range: 100-270 min). Median hospital stay was 2.5 days (IQR: 2, 4 days; range: 2-14 days). Major complications occurred in 3/14 (21%) patients. Primary efficacy rate was 93% (13/14 patients). Median oncological follow-up was 25 months (IQR: 11, 33 months; range: 6-39 months). One patient experienced renal tumor recurrence at 14-months of follow-up, which was successfully treated with repeat cryoablation. CONCLUSION: Percutaneous image-guided cryoablation of renal tumors with temporary balloon occlusion of the renal artery is technically feasible, with a high technical success rate and paths the way for percutaneous treatment of central renal tumors.


Subject(s)
Balloon Occlusion , Carcinoma, Renal Cell , Cryosurgery , Kidney Neoplasms , Renal Artery , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Balloon Occlusion/methods , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cryosurgery/methods , Kidney Neoplasms/blood supply , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Retrospective Studies , Tomography, X-Ray Computed , Treatment Outcome , Surgery, Computer-Assisted/methods , Feasibility Studies
5.
Mol Cancer ; 21(1): 111, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538475

ABSTRACT

BACKGROUND: Sunitinib resistance can be classified into primary and secondary resistance. While accumulating research has indicated several underlying factors contributing to sunitinib resistance, the precise mechanisms in renal cell carcinoma are still unclear. METHODS: RNA sequencing and m6A sequencing were used to screen for functional genes involved in sunitinib resistance. In vitro and in vivo experiments were carried out and patient samples and clinical information were obtained for clinical analysis. RESULTS: We identified a tumor necrosis factor receptor-associated factor, TRAF1, that was significantly increased in sunitinib-resistant cells, resistant cell-derived xenograft (CDX-R) models and clinical patients with sunitinib resistance. Silencing TRAF1 increased sunitinib-induced apoptotic and antiangiogenic effects. Mechanistically, the upregulated level of TRAF1 in sunitinib-resistant cells was derived from increased TRAF1 RNA stability, which was caused by an increased level of N6-methyladenosine (m6A) in a METTL14-dependent manner. Moreover, in vivo adeno-associated virus 9 (AAV9) -mediated transduction of TRAF1 suppressed the sunitinib-induced apoptotic and antiangiogenic effects in the CDX models, whereas knockdown of TRAF1 effectively resensitized the sunitinib-resistant CDXs to sunitinib treatment. CONCLUSIONS: Overexpression of TRAF1 promotes sunitinib resistance by modulating apoptotic and angiogenic pathways in a METTL14-dependent manner. Targeting TRAF1 and its pathways may be a novel pharmaceutical intervention for sunitinib-treated patients.


Subject(s)
Adenosine , Carcinoma, Renal Cell , Kidney Neoplasms , Methyltransferases , Sunitinib , TNF Receptor-Associated Factor 1 , Adenosine/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Methyltransferases/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Sunitinib/pharmacology , TNF Receptor-Associated Factor 1/genetics , TNF Receptor-Associated Factor 1/metabolism
6.
Anticancer Res ; 42(3): 1313-1325, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220222

ABSTRACT

BACKGROUND/AIM: Chloride intracellular channel protein 1 (CLIC1) is known as a promoter of cancer progression, metastasis, and angiogenesis. Thus, CLIC1 could be a future therapeutic target. This study aimed to evaluate the effect of anti-CLIC1 antibodies on tumour cells and vessels of human renal cell carcinoma (RCC) in rabbit cornea and chick embryo chorioallantoic membrane (CAM) models. MATERIALS AND METHODS: Human cc-RCC xenografts on rabbit cornea and CAM surface were performed. Anti-CLIC1 antibodies were applied for 5 consecutive days on both tumor models. We comparatively evaluated treated and untreated tumors by combining ultrasonography with microscopic techniques. RESULTS: RCC implants rapidly recruited blood vessels and had an exponential growth rate on both tumor models. Anti-CLIC1 antibodies suppressed tumor growth by inducing tumor cell necrosis. Tumor vessels regressed rapidly but not completely during anti-CLIC1 antibodies based therapy. CONCLUSION: Anti-CLIC1 antibodies induced tumor necrosis and tumor vasculature regression in human cc-RCC xenografts in both in vivo experimental models.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Chloride Channels/antagonists & inhibitors , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Neovascularization, Pathologic , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Chick Embryo , Chloride Channels/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Molecular Targeted Therapy , Necrosis , Rabbits , Signal Transduction , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Urol Oncol ; 40(1): 13.e1-13.e8, 2022 01.
Article in English | MEDLINE | ID: mdl-34535355

ABSTRACT

PURPOSE: To evaluate the role of dynamic contrast-enhanced CT (DCE-CT) as an independent non-invasive biomarker in predicting long term outcome in patients with metastatic renal cell carcinoma (mRCC) on antiangiogenic treatment. MATERIAL AND METHODS: Eighty two mRCC patients were prospectively enrolled from 09/2011 to 04/2015, out of which 71 were included in the final data analysis; the population was observed until 12/2020 to obtain complete overall survival data. DCE-CT imaging was performed at baseline and 10 to 12 weeks after start of treatment with targeted therapy. DCE-CT included a dynamic acquisition after injection of 50 ml of nonionic contrast agent at 6 ml/s using a 4D spiral mode (10 cm z-axis coverage, acquisition time 43 sec, 100 kVp (abdomen), 80 kVp (chest), 80-100 mAs) on a dual source scanner (Definition FLASH, Siemens). Blood flow (BF) was calculated for target tumor volumes using a deconvolution model. Progression free survival (PFS) and overall survival (OS) were analyzed using Kaplan-Meier statistics (SPSS version 24). RESULTS: Patients were treated with either sunitinib, pazopanib, sorafenib, tivozanib, axitinib, or cabozantinib. A cut-off value of 50% blood flow reduction at follow-up allowed for identification of patients with favorable long-term outcome: Median OS in n = 42 patients with an average blood flow reduction of >50% (mean, 79%) was 34 (range, 14-54) months, while n = 21 patients with an average reduction of less than 50% (mean, 28%) showed a median OS of 12 (range, 6-18) months, and n = 8 patients with an increase in blood flow survived for a median of 7 (range, 3-11) months. CONCLUSION: Blood flow in metastases measured with DCE-CT at first follow-up is a strong predictor of overall survival in mRCC patients on antiangiogenic treatment.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Contrast Media , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Regional Blood Flow , Tomography, X-Ray Computed/methods , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Prospective Studies , Time Factors , Treatment Outcome
8.
Urology ; 159: 120-126, 2022 01.
Article in English | MEDLINE | ID: mdl-34537195

ABSTRACT

OBJECTIVE: To evaluate the safety, efficacy, and early oncologic outcomes of pathologic T3a (pT3a) renal cell carcinoma with venous involvement treated with robotic partial nephrectomy (RPN), given that experience and outcomes in this group is limited. METHODS: A retrospective chart review of patients undergoing RPN from September 2009 to July 2020 was performed. Outcomes were captured from patients with pT3a disease with vein involvement. Clinical characteristics were analyzed using SPSS (IBM, Armonk, NY). Local recurrence-free survival and metastasis-free survival at 2 years were calculated from Kaplan-Meier survival curves. RESULTS: For 45 included patients, mean operative and warm ischemia times were 199.6 ± 47.3 minutes and 30.5 ± 10.5 minutes, with mean estimated blood loss of 324.9 ± 209.5 cc. Rates of transfusion, embolization, re-admission, and re-operation at 30 days were 8.9% (4/45), 2.2% (1/45), 11.1% (5/45), and 6.7% (3/45; cystoscopic stent placement), respectively. All tumors were malignant on pathology, with clear cell renal cell carcinoma being the most common (91.0%, n = 41). The positive margin rate was 6.7% (n = 3). Local recurrence occurred in 4.4% (n = 2) at a mean time of 5.2 ± 2.3 months. Four patients (8.9%) progressed to metastatic disease at a mean of 22.2 ± 23.0 months. At 2 years, local recurrence-free survival was 95.4% and metastasis-free survival was 95.3%. CONCLUSION: We present the largest known series of patients RPN for pT3a renal masses with venous tumor involvement. We found it both feasible and safe in the appropriate hands. Short term oncologic outcomes for these patients appear more favorable than historic literature suggested.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Nephrectomy , Postoperative Complications , Robotic Surgical Procedures , Venous Thrombosis , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/surgery , Female , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/blood supply , Kidney Neoplasms/complications , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Male , Margins of Excision , Middle Aged , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/etiology , Neoplasm Staging , Nephrectomy/adverse effects , Nephrectomy/methods , Outcome and Process Assessment, Health Care , Postoperative Complications/diagnosis , Postoperative Complications/epidemiology , Postoperative Complications/prevention & control , Retrospective Studies , Risk Adjustment/methods , Robotic Surgical Procedures/adverse effects , Robotic Surgical Procedures/methods , Venous Thrombosis/diagnosis , Venous Thrombosis/epidemiology , Venous Thrombosis/etiology
9.
FASEB J ; 36(1): e22080, 2022 01.
Article in English | MEDLINE | ID: mdl-34882832

ABSTRACT

Angiogenesis is required in embryonic development and tissue repair in the adult. Vascular endothelial growth factor (VEGF) initiates angiogenesis, and VEGF or its receptor is targeted therapeutically to block pathological angiogenesis. Additional pro-angiogenic cues, such as CXCL12 acting via the CXCR4 receptor, co-operate with VEGF/VEGFR2 to cue vascular patterning. We studied the role of FGD5, an endothelial Rho GTP/GDP exchange factor (RhoGEF), to regulate CXCR4-dependent signals in the endothelial cell (EC). Patient-derived renal cell carcinomas produce a complex milieu of growth factors that stimulated sprouting angiogenesis and endothelial tip cell differentiation ex vivo that was blocked by EC FGD5 loss. In a simplified model, CXCL12 augmented sprouting and tip gene expression under conditions where VEGF was limiting. CXCL12-stimulated tip cell differentiation was dependent on PI3 kinase (PI3K)-ß activity. Knockdown of EC FGD5 abolished CXCR4 signaling to PI3K-ß and Akt. Further, inhibition of Rac1, a Rho GTPase required for PI3K-ß activity, recapitulated the signaling defects of FGD5 deficiency, suggesting that FGD5 may regulate PI3K-ß activity through Rac1. Overexpression of a RhoGEF deficient, Dbl domain-deleted FGD5 mutant reduced CXCL12-stimulated Akt phosphorylation and failed to rescue PI3K signaling in native FGD5-deficient EC, indicating that FGD5 RhoGEF activity is required for FDG5 function. Endothelial expression of mutant PI3K-ß with an inactivated Rho binding domain confirmed that CXCL12-stimulated PI3K activity in EC requires Rac1-GTP co-regulation. Together, this data identify the role of FGD5 to generate Rac1-GTP to regulate pro-angiogenic CXCR4-dependent PI3K-ß signaling in EC. Inhibition of FGD5 activity may complement current angiogenesis inhibitor drugs.


Subject(s)
Carcinoma, Renal Cell , Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Kidney Neoplasms , Neoplasm Proteins/metabolism , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Guanine Nucleotide Exchange Factors/genetics , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/genetics
10.
BMC Cancer ; 21(1): 1238, 2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34794412

ABSTRACT

BACKGROUND: Window-of-opportunity trials, evaluating the engagement of drugs with their biological target in the time period between diagnosis and standard-of-care treatment, can help prioritise promising new systemic treatments for later-phase clinical trials. Renal cell carcinoma (RCC), the 7th commonest solid cancer in the UK, exhibits targets for multiple new systemic anti-cancer agents including DNA damage response inhibitors, agents targeting vascular pathways and immune checkpoint inhibitors. Here we present the trial protocol for the WIndow-of-opportunity clinical trial platform for evaluation of novel treatment strategies in REnal cell cancer (WIRE). METHODS: WIRE is a Phase II, multi-arm, multi-centre, non-randomised, proof-of-mechanism (single and combination investigational medicinal product [IMP]), platform trial using a Bayesian adaptive design. The Bayesian adaptive design leverages outcome information from initial participants during pre-specified interim analyses to determine and minimise the number of participants required to demonstrate efficacy or futility. Patients with biopsy-proven, surgically resectable, cT1b+, cN0-1, cM0-1 clear cell RCC and no contraindications to the IMPs are eligible to participate. Participants undergo diagnostic staging CT and renal mass biopsy followed by treatment in one of the treatment arms for at least 14 days. Initially, the trial includes five treatment arms with cediranib, cediranib + olaparib, olaparib, durvalumab and durvalumab + olaparib. Participants undergo a multiparametric MRI before and after treatment. Vascularised and de-vascularised tissue is collected at surgery. A ≥ 30% increase in CD8+ T-cells on immunohistochemistry between the screening and nephrectomy is the primary endpoint for durvalumab-containing arms. Meanwhile, a reduction in tumour vascular permeability measured by Ktrans on dynamic contrast-enhanced MRI by ≥30% is the primary endpoint for other arms. Secondary outcomes include adverse events and tumour size change. Exploratory outcomes include biomarkers of drug mechanism and treatment effects in blood, urine, tissue and imaging. DISCUSSION: WIRE is the first trial using a window-of-opportunity design to demonstrate pharmacological activity of novel single and combination treatments in RCC in the pre-surgical space. It will provide rationale for prioritising promising treatments for later phase trials and support the development of new biomarkers of treatment effect with its extensive translational agenda. TRIAL REGISTRATION: ClinicalTrials.gov: NCT03741426 / EudraCT: 2018-003056-21 .


Subject(s)
Antineoplastic Agents/therapeutic use , Bayes Theorem , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Capillary Permeability/drug effects , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/pathology , Humans , Kidney/pathology , Kidney Neoplasms/blood supply , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating , Magnetic Resonance Imaging , Medical Futility , Nephrectomy , Non-Randomized Controlled Trials as Topic , Phthalazines/therapeutic use , Piperazines/therapeutic use , Proof of Concept Study , Quinazolines/therapeutic use , Treatment Outcome , Tumor Burden
11.
Virchows Arch ; 479(6): 1187-1196, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34611731

ABSTRACT

Renal cell carcinomas (RCC) are well-vascularized tumors. Although clear cell RCC (CCRCC) show a characteristic vascular network, some cases show overlapping features with other RCC. We aimed to evaluate vascular architectural patterns, microvessel density (MVD), and endothelial cell density (ECD) in CCRCC compared to clear cell papillary RCC (ccpRCC). Thirty-four RCC (17 CCRCC and 17 ccpRCC) were included in the study. CD34 was used to evaluate vascular architectural patterns by microscopic estimation in all cases. CD34, ERG, and Bioquant Osteo 2019 Imaging Analysis Software were used to evaluate MVD and ECD in 17 CCRCC and 15 ccpRCC. Mean MVD was 526.63 in CCRCC vs. 426.18 in ccpRCC (p = 0.16); mean ECD was 937.50 in CCRCC vs. 1060.21 in ccpRCC (p = 0.25). CD34 highlighted four distinct vascular architectural patterns: pseudoacinar, Golgi-like, lacunae, and scattered. Lacunae and pseudoacinar was the most frequent combination in CCRCC; lacunae and Golgi-like was the predominant combination among ccpRCC. Pseudoacinar was most extensive in CCRCC and least in ccpRCC; Golgi-like was predominant in ccpRCC and uncommon in CCRCC. The extent of pseudoacinar and Golgi-like vascular architectural patterns was significantly different between CCRCC and ccpRCC (p < 0.05). Pathologists acquainted with these different vascular architectural patterns may utilize them as an additional tool in the distinction of CCRCC from ccpRCC.


Subject(s)
Blood Vessels/pathology , Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/blood supply , Neovascularization, Pathologic , Adult , Aged , Aged, 80 and over , Antigens, CD34/analysis , Biomarkers, Tumor/analysis , Biopsy , Blood Vessels/chemistry , Diagnosis, Differential , Endothelial Cells/chemistry , Endothelial Cells/pathology , Female , Humans , Immunohistochemistry , Male , Microvascular Density , Middle Aged , Neoplasm Grading , Predictive Value of Tests , Retrospective Studies
12.
APMIS ; 129(9): 535-547, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33948984

ABSTRACT

The increasing demands for personalized targeted therapy directed against renal cell carcinoma have driven a search for predictive markers. Novel therapies targeting HIF-1α in renal cell carcinoma have been developed, and HIF-1α has been suggested as a novel predictive marker of response to therapy. The surgical resection of a kidney tumor induces tissue ischemia, and HIF-1α is an oxygen-sensitive transcription factor, which is known to be upregulated during hypoxia. This study investigated the impact of intra-surgical and post-surgical ischemia on protein expression levels of HIF-1α and three related biomarkers (VEGF, GLUT-1, and CAIX) in 20 patients with renal cell carcinoma with immunohistochemistry and Western blotting. Surgical ischemia did not have a significant impact on protein expression levels of any of the investigated markers. Long-post-surgical ischemia resulted in reduced expression levels of HIF-1α, probably due to autolysis. Our results suggest that HIF-1α is a stable protein, with expression levels not affected by intra-surgical ischemia, and hence, HIF-1α is suited for marker analysis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/blood supply , Kidney Neoplasms/metabolism , Aged , Aged, 80 and over , Carbonic Anhydrase IX/metabolism , Carcinoma, Renal Cell/surgery , Female , Glucose Transporter Type 1/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Kidney Neoplasms/surgery , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Nephrectomy , Prognosis , Prospective Studies , Tissue Array Analysis , Tumor Hypoxia/genetics , Tumor Hypoxia/physiology , Vascular Endothelial Growth Factor A/metabolism
13.
Cell Commun Signal ; 19(1): 39, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33761933

ABSTRACT

BACKGROUND: Tumor angiogenesis, an essential process for cancer proliferation and metastasis, has a critical role in prognostic of kidney renal clear cell carcinoma (KIRC), as well as a target in guiding treatment with antiangiogenic agents. However, tumor angiogenesis subtypes and potential epigenetic regulation mechanisms in KIRC patient remains poorly characterized. System evaluation of angiogenesis subtypes in KIRC patient might help to reveal the mechanisms of KIRC and develop more target treatments for patients. METHOD: Ten independent tumor angiogenesis signatures were obtained from molecular signatures database (MSigDB) and gene set variation analysis was performed to calculate the angiogenesis score in silico using the Cancer Genome Atlas (TCGA) KIRC dataset. Tumor angiogenesis subtypes in 539 TCGA-KIRC patients were identified using consensus clustering analysis. The potential regulation mechanisms was studied using gene mutation, copy number variation, and differential methylation analysis (DMA). The master transcription factors (MTF) that cause the difference in tumor angiogenesis signals were completed by transcription factor enrichment analysis. RESULTS: The angiogenesis score of a prognosis related angiogenesis signature including 189 genes was significantly correlated with immune score, stroma score, hypoxia score, and vascular endothelial growth factor (VEGF) signal score in 539 TCGA KIRC patients. MMRN2, CLEC14A, ACVRL1, EFNB2, and TEK in candidate gene set showed highest correlation coefficient with angiogenesis score in TCGA-KIRC patients. In addition, all of them were associated with overall survival in both TCGA-KIRC and E-MTAB-1980 KIRC data. Clustering analysis based on 183 genes in angiogenesis signature identified two prognosis related angiogenesis subtypes in TCGA KIRC patients. Two clusters also showed different angiogenesis score, immune score, stroma score, hypoxia score, VEGF signal score, and microenvironment score. DMA identified 59,654 differential methylation sites between two clusters and part of these sites were correlated with tumor angiogenesis genes including CDH13, COL4A3, and RHOB. In addition, RFX2, SOX13, and THRA were identified as top three MTF in regulating angiogenesis signature in KIRC patients. CONCLUSION: Our study indicate that evaluation the angiogenesis subtypes of KIRC based on angiogenesis signature with 183 genes and potential epigenetic mechanisms may help to develop more target treatments for KIRC patients. Video Abstract.


Subject(s)
Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Genomics , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Neovascularization, Pathologic/genetics , Cohort Studies , DNA Copy Number Variations/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Humans , Mutation/genetics , Prognosis , Transcription Factors/metabolism , Tumor Microenvironment/genetics
14.
Ultrasound Med Biol ; 47(4): 1089-1098, 2021 04.
Article in English | MEDLINE | ID: mdl-33468358

ABSTRACT

Singular value decomposition-based clutter filters can robustly reject tissue clutter, allowing for detection of slow blood flow in imaging microvasculature. However, to identify microvessels, high ultrasound frequency must be used to increase the spatial resolution at the expense of shorter depth of penetration. Deconvolution using Tikhonov regularization is an imaging processing method widely used to improve spatial resolution. The ringing artifact of Tikhonov regularization, though, can produce image artifacts such as non-existent microvessels, which degrade image quality. Therefore, a deconvolution method using total variation is proposed in this study to improve spatial resolution and mitigate the ringing artifact. Performance of the proposed method was evaluated using chicken embryo brain, ex ovo chicken embryo chorioallantoic membrane and tumor data. Results revealed that the reconstructed power Doppler (PD) images are substantially improved in spatial resolution compared with original PD images: the full width half-maximum (FWHM) of the cross-sectional profile of a microvessel was improved from 132 to 83 µm. Two neighboring microvessels that were 154 µm apart were better separated using the proposed method than conventional PD imaging. Additionally, 223 FWHMs measured from the cross-sectional profiles of 223 vessels were used to determine the improvement in FWHM with the proposed method statistically. The mean ± standard deviation of the FWHM without and with the proposed method was 233.19 ± 85.08 and 172.31 ± 75.11 µm, respectively; the maximum FWHM without and with the proposed method was 693.01 and 668.69 µm; and the minimum FWHM without and with the proposed method was 73.92 and 45.74 µm. There were statistically significant differences between FWHMs with and without the proposed method according to the rank-sum test, p < 0.0001. The contrast-to-noise ratio improved from 1.06 to 4.03 dB with use of the proposed method. We also compared the proposed method with Tikhonov regularization using ex ovo chicken embryo chorioallantoic membrane data. We found that the proposed method outperformed Tikhonov regularization as false microvessels appeared using the Tikhonov regularization but not with the proposed method. These results indicate that the proposed method is capable of providing more robust PD images with higher spatial resolution and higher contrast-to-noise ratio.


Subject(s)
Brain/blood supply , Carcinoma, Renal Cell/blood supply , Chorioallantoic Membrane/blood supply , Image Processing, Computer-Assisted/methods , Microvessels/diagnostic imaging , Ultrasonography, Doppler/methods , Animals , Artifacts , Chick Embryo , Neoplasm Transplantation , Signal Processing, Computer-Assisted , Signal-To-Noise Ratio
15.
Cells ; 11(1)2021 12 22.
Article in English | MEDLINE | ID: mdl-35011579

ABSTRACT

The modulation of subpopulations of pro-angiogenic monocytes (VEGFR-1+CD14 and Tie2+CD14) was analyzed in an ancillary study from the prospective PazopanIb versus Sunitinib patient preferenCE Study (PISCES) (NCT01064310), where metastatic renal cell carcinoma (mRCC) patients were treated with two anti-angiogenic drugs, either sunitinib or pazopanib. Blood samples from 86 patients were collected prospectively at baseline (T1), and at 10 weeks (T2) and 20 weeks (T3) after starting anti-angiogenic therapy. Various subpopulations of myeloid cells (monocytes, VEGFR-1+CD14 and Tie2+CD14 cells) decreased during treatment. When patients were divided into two subgroups with a decrease (defined as a >20% reduction from baseline value) (group 1) or not (group 2) at T3 for VEGFR-1+CD14 cells, group 1 patients presented a median PFS and OS of 24 months and 37 months, respectively, compared with a median PFS of 9 months (p = 0.032) and a median OS of 16 months (p = 0.033) in group 2 patients. The reduction in Tie2+CD14 at T3 predicted a benefit in OS at 18 months after therapy (p = 0.04). In conclusion, in this prospective clinical trial, a significant decrease in subpopulations of pro-angiogenic monocytes was associated with clinical response to anti-angiogenic drugs in patients with mRCC.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Monocytes/pathology , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antigens, CD/metabolism , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/pathology , Disease Models, Animal , Humans , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Mice, Inbred BALB C , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoplasm Metastasis , Sunitinib/pharmacology , Sunitinib/therapeutic use , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-1/metabolism
16.
Cancer Immunol Immunother ; 70(6): 1619-1634, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33237349

ABSTRACT

The microRNA let-7d has been reported to be a tumor suppressor in renal cell carcinoma (RCC). Tumor-associated macrophages (TAM) are M2-polarized macrophages that can enhance tumor growth and angiogenesis in many human cancers. However, the role of let-7d in TAM-associated RCC progression remains elusive. First, we observed a strongly inverse correlation between let-7d expression and microvessel density in RCC tissues. Furthermore, the proliferation, migration, and tube formation of HUVECs were significantly inhibited by conditioned medium from a coculture system of the phorbol myristate acetate pretreated human THP-1 macrophages and let-7d-overexpressing RCC cells. Moreover, the proportion of M2 macrophages was significantly lower in the group that was cocultured with let-7d-overexpressing RCC cells. Subcutaneous xenografts formed by the injection of let-7d-overexpressing RCC cells together with THP-1 cells resulted in a significant decrease in the M2 macrophage ratio and microvessel density compared with those formed by the injection of control RCC cells with THP-1 cells. In silico and experimental analysis revealed interleukin-10 (IL-10) and IL-13 as let-7d target genes. Importantly, the addition of IL-10 and IL-13 counteracted the inhibitory effects of the conditioned medium from the coculture system with let-7d-overexpressing RCC cells in vitro. Additionally, overexpression of IL-10 and IL-13 reversed the effects of let-7d on macrophage M2 polarization and tumor angiogenesis in vivo. Finally, the expression of IL-10 and IL-13 were inversely correlated with the expression of let-7d in RCC clinical specimens. These results suggest that let-7d may inhibit intratumoral macrophage M2 polarization and subsequent tumor angiogenesis by targeting IL-10 and IL-13.


Subject(s)
Carcinoma, Renal Cell/prevention & control , Interleukin-10/antagonists & inhibitors , Interleukin-13/antagonists & inhibitors , Kidney Neoplasms/prevention & control , Macrophage Activation/immunology , MicroRNAs/genetics , Neovascularization, Pathologic/therapy , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology , Prognosis , THP-1 Cells/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
BMC Bioinformatics ; 21(1): 558, 2020 Dec 04.
Article in English | MEDLINE | ID: mdl-33276732

ABSTRACT

BACKGROUND: High resolution 2D whole slide imaging provides rich information about the tissue structure. This information can be a lot richer if these 2D images can be stacked into a 3D tissue volume. A 3D analysis, however, requires accurate reconstruction of the tissue volume from the 2D image stack. This task is not trivial due to the distortions such as tissue tearing, folding and missing at each slide. Performing registration for the whole tissue slices may be adversely affected by distorted tissue regions. Consequently, regional registration is found to be more effective. In this paper, we propose a new approach to an accurate and robust registration of regions of interest for whole slide images. We introduce the idea of multi-scale attention for registration. RESULTS: Using mean similarity index as the metric, the proposed algorithm (mean ± SD [Formula: see text]) followed by a fine registration algorithm ([Formula: see text]) outperformed the state-of-the-art linear whole tissue registration algorithm ([Formula: see text]) and the regional version of this algorithm ([Formula: see text]). The proposed algorithm also outperforms the state-of-the-art nonlinear registration algorithm (original: [Formula: see text], regional: [Formula: see text]) for whole slide images and a recently proposed patch-based registration algorithm (patch size 256: [Formula: see text] , patch size 512: [Formula: see text]) for medical images. CONCLUSION: Using multi-scale attention mechanism leads to a more robust and accurate solution to the problem of regional registration of whole slide images corrupted in some parts by major histological artifacts in the imaged tissue.


Subject(s)
Algorithms , Artifacts , Blood Vessels/pathology , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Blood Vessels/diagnostic imaging , Carcinoma, Renal Cell/blood supply , Humans , Immunohistochemistry/methods , Microscopy
18.
Oncol Rep ; 44(5): 1961-1970, 2020 11.
Article in English | MEDLINE | ID: mdl-32901879

ABSTRACT

Renal cell carcinoma (RCC) is one of the most common malignant cancers in the adult urinary system worldwide. Tumor angiogenesis is a critical process during cancer progression, as it modulates carcinogenesis and metastasis. In recent years, microRNA­218 (miR­218) has been confirmed to play a crucial role in tumor suppression. However, the role of miR­218 in RCC angiogenesis remains unclear. In the present study, it was found that the expression of miR­218 was decreased in RCC tumor tissues and cell lines as detected by real­time PCR analysis. Tube formation assays and migration assays also confirmed that miR­218 inhibited the interaction between RCC cells and vascular endothelial cells by suppressing proangiogenic factor vascular endothelial growth factor A (VEGFA) in RCC cells. miR­218 also repressed the subcutaneous tumorigenesis of RCC cells in nude mice, and the corneal angiogenesis in rabbit eyes. The underlying molecular mechanism was elucidated; miR­218 targets GRB2­associated binding protein 2 (GAB2), thereby inhibiting the PI3K/AKT/mTOR/VEGFA pathway. These results provide new insights into the mechanism of RCC carcinogenesis and progression, suggesting that miRNA­218 may be a therapeutic target for the treatment of RCC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Renal Cell/blood supply , Kidney Neoplasms/blood supply , MicroRNAs/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Cell Proliferation , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Male , Mice , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Rabbits , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
19.
Oncogene ; 39(41): 6480-6492, 2020 10.
Article in English | MEDLINE | ID: mdl-32879446

ABSTRACT

Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. However, only a fraction of patients respond, and most ultimately develop resistance to current angiogenesis inhibitor therapies. Activity of alternative pro-angiogenic growth factors, acting via RTK or G-protein coupled receptors (GPCR), may mediate VEGF inhibitor resistance. The phosphoinositide 3-kinase (PI3K)ß isoform is uniquely coupled to both RTK and GPCRs. We investigated the role of endothelial cell (EC) PI3Kß in tumor angiogenesis. Pro-angiogenic GPCR ligands were expressed by patient-derived renal cell carcinomas (PD-RCC), and selective inactivation of PI3Kß reduced PD-RCC-stimulated EC spheroid sprouting. EC-specific PI3Kß knockout (ΕC-ßKO) in mice potentiated the sunitinib-induced reduction in subcutaneous growth of LLC1 and B16F10, and lung metastasis of B16F10 tumors. Compared to single-agent sunitinib treatment, tumors in sunitinib-treated ΕC-ßKO mice showed a marked decrease in microvessel density, and reduced new vessel formation. The fraction of perfused mature tumor microvessels was increased in ΕC-ßKO mice suggesting immature microvessels were most sensitive to combined sunitinib and PI3Kß inactivation. Taken together, EC PI3Kß inactivation with sunitinib inhibition reduces microvessel turnover and decreases heterogeneity of the tumor microenvironment, hence PI3Kß inhibition may be a useful adjuvant antiangiogenesis therapy with sunitinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Renal Cell/pathology , Class I Phosphatidylinositol 3-Kinases/metabolism , Kidney Neoplasms/pathology , Neovascularization, Pathologic/pathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice, Knockout , Microvessels/drug effects , Microvessels/pathology , Morpholines/pharmacology , Morpholines/therapeutic use , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Sunitinib/pharmacology , Sunitinib/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Eur J Histochem ; 64(3)2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32613819

ABSTRACT

A simple dark field microscopy technique was used for visualization of blood vessels in normal human renal tissues and carcinoma. Phase contrast condenser ring apt for high power objectives was combined with a 10x objective in order to create a dark field illumination of the specimens examined. The endothelial lining of the vessels had been stained by using CD31 monoclonal antibodies combined with conventional peroxidase immunohistochemistry. The final DAB addition used for this technique induced an intense light scatter in the dark field microscope. This scattered light originating from the endothelial lining made the walls of the bright vessels easily detectable from the dark background.


Subject(s)
Endothelium, Vascular/diagnostic imaging , Platelet Endothelial Cell Adhesion Molecule-1/immunology , 3,3'-Diaminobenzidine/chemistry , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Renal Cell/blood supply , Chromogenic Compounds/chemistry , Humans , Hydrogen Peroxide/chemistry , Immunohistochemistry , Kidney/blood supply , Kidney Neoplasms/blood supply , Microscopy/methods , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...