1
|
Abah MO, Ogenyi DO, Zhilenkova AV, Essogmo FE, Ngaha Tchawe YS, Uchendu IK, Pascal AM, Nikitina NM, Rusanov AS, Sanikovich VD, Pirogova YN, Boroda A, Moiseeva AV, Sekacheva MI. Innovative Therapies Targeting Drug-Resistant Biomarkers in Metastatic Clear Cell Renal Cell Carcinoma (ccRCC). Int J Mol Sci 2024; 26:265. [PMID: 39796121 PMCID: PMC11720203 DOI: 10.3390/ijms26010265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 01/13/2025] Open
Abstract
A thorough study of Clear Cell Renal Cell Carcinoma (ccRCC) shows that combining tyrosine kinase inhibitors (TKI) with immune checkpoint inhibitors (ICI) shows promising results in addressing the tumor-promoting influences of abnormal immunological and molecular biomarkers in metastatic Clear Cell Renal Cell Carcinoma (ccRCC). These abnormal biomarkers enhance drug resistance, support tumor growth, and trigger cancer-related genes. Ongoing clinical trials are testing new treatment options that appear more effective than earlier ones. However, more research is needed to confirm their long-term safety use and potential side effects. This study highlights vital molecular and immunological biomarkers associated with drug resistance in Clear Cell Renal Cell Carcinoma (ccRCC). Furthermore, this study identifies a number of promising drug candidates and biomarkers that serve as significant contributors to the enhancement of the overall survival of ccRCC patients. Consequently, this article offers pertinent insights on both recently completed and ongoing clinical trials, recommending further toxicity study for the prolonged use of this treatment strategy for patients with metastatic ccRCC, while equipping researchers with invaluable information for the progression of current treatment strategies.
Collapse
Affiliation(s)
- Moses Owoicho Abah
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
- Department of Cancer Bioinformatics and Molecular Biology, Royal Society of Clinical and Academic Researchers (ROSCAR) International, Abuja 900104, Nigeria
| | - Deborah Oganya Ogenyi
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Angelina V. Zhilenkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Freddy Elad Essogmo
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Yvan Sinclair Ngaha Tchawe
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Ikenna Kingsley Uchendu
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
- Medical Laboratory Science Department, Faculty of Health Science and Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 410001, Nigeria
| | - Akaye Madu Pascal
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Natalia M. Nikitina
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Alexander S. Rusanov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Varvara D. Sanikovich
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Yuliya N. Pirogova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Alexander Boroda
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Aleksandra V. Moiseeva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Marina I. Sekacheva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| |
Collapse
|
2
|
Bodard S, Delavaud C, Dariane C, Boudhabhay I, Bensenouci NEI, Timsit MO, Correas JM, Verkarre V, Hélénon O. Low-grade oncocytic tumor of the kidney: imaging features of a novel tumor entity. Abdom Radiol (NY) 2024; 49:4307-4323. [PMID: 39068611 DOI: 10.1007/s00261-024-04487-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 07/30/2024]
Abstract
PURPOSES Low-grade oncocytic tumor (LOT) is a rare renal tumor that has emerged from the spectrum of eosinophilic/oncocytic renal tumors and poses a diagnostic challenge due to its similarity to chromophobe renal cell carcinoma (CHRCC) and renal oncocytoma (RO). The imaging features of this novel tumor entity have not yet been clearly described. The purpose of this study was to describe the imaging features of LOT with radiologic-pathologic correlation. METHODS We conducted a retrospective observational study involving two expert centers. We identified 12 pathologically proven LOT with preoperative imaging available, including at least computed tomography (CT) or magnetic resonance imaging (MRI), from the past 12 years. Three experienced radiologists performed the imaging analysis independently. RESULTS All tumors presented well-defined borders. Nine of the 12 LOT exhibited an early peripheral enhancement with complete or almost complete centripetal fill-in on nephrographic or delayed phases without any particular shape. Three showed a homogeneous contrast enhancement. Macroscopic fat and calcifications were not observed in any of the tumors. CONCLUSION Early peripheral enhancement with complete or almost complete centripetal fill-in on nephrographic or delayed phases without any particular shape suggests a LOT diagnosis. Further analyses involving larger studies are needed to fully confirm these imaging characteristics. To date, a percutaneous biopsy should be performed before considering management.
Collapse
Affiliation(s)
- Sylvain Bodard
- Adult Department of Radiology, Service d'Imagerie Adulte, AP-HP-Centre, Hôpital Necker Enfants Malades, Université de Paris Cité, 149 Rue de Sèvres, 75015, Paris, France.
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, Paris, France.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Christophe Delavaud
- Adult Department of Radiology, Service d'Imagerie Adulte, AP-HP-Centre, Hôpital Necker Enfants Malades, Université de Paris Cité, 149 Rue de Sèvres, 75015, Paris, France
| | - Charles Dariane
- Service d'Urologie, AP-HP-Centre, Hôpital Européen Georges Pompidou, Université de Paris Cité, 75015, Paris, France
| | - Idris Boudhabhay
- Service de Transplantation Rénale, AP-HP-Centre, Hôpital Necker Enfants Malades, Université de Paris Cité, 75015, Paris, France
| | - Nour El Imane Bensenouci
- Service d'Anatomie Pathologie, AP-HP-Centre, Hôpital Européen Georges Pompidou, Université de Paris Cité, 75015, Paris, France
| | - Marc-Olivier Timsit
- Service d'Urologie, AP-HP-Centre, Hôpital Européen Georges Pompidou, Université de Paris Cité, 75015, Paris, France
| | - Jean-Michel Correas
- Adult Department of Radiology, Service d'Imagerie Adulte, AP-HP-Centre, Hôpital Necker Enfants Malades, Université de Paris Cité, 149 Rue de Sèvres, 75015, Paris, France
- Laboratoire d'Imagerie Biomédicale, Sorbonne Université, CNRS, INSERM, Paris, France
| | - Virginie Verkarre
- Service d'Anatomie Pathologie, AP-HP-Centre, Hôpital Européen Georges Pompidou, Université de Paris Cité, 75015, Paris, France
- Equipe INSERM UMR 970 "Genetic and Metabolism of Rare Tumors" Equipe Labélisée Ligue Contre Le Cancer, PARCC, SIRIC CARPEM, Université de Paris-Cité, Paris, France
| | - Olivier Hélénon
- Adult Department of Radiology, Service d'Imagerie Adulte, AP-HP-Centre, Hôpital Necker Enfants Malades, Université de Paris Cité, 149 Rue de Sèvres, 75015, Paris, France
| |
Collapse
|
3
|
Quinn AE, Bell SD, Marrah AJ, Wakefield MR, Fang Y. The Current State of the Diagnoses and Treatments for Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2024; 16:4034. [PMID: 39682220 DOI: 10.3390/cancers16234034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Clear cell renal cell carcinoma is the most common form of kidney cancer, accounting for 75% of malignant kidney tumors, and is generally associated with poor patient outcomes. With risk factors including smoking, obesity, and hypertension, all of which have a high prevalence in the United States and Europe, as well as genetic factors including tuberous sclerosis complex and Von Hippel-Lindau syndrome, there is an increasing need to expand our present understanding. The current clear cell renal cell carcinoma knowledge is outdated, with obsolete diagnostic criteria and moderately invasive surgical treatments still prevailing, partially ascribed to its resistance to chemotherapy and radiation therapy. The standard of treatment relies on surgical intervention, including radical nephrectomy and partial nephrectomy, while more recent treatments target neoplastic growth pathways and immune regulation checkpoints.
Collapse
Affiliation(s)
- Anthony E Quinn
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Scott D Bell
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
| | - Austin J Marrah
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Mark R Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
4
|
Ruan X, Lai C, Li L, Wang B, Lu X, Zhang D, Fang J, Lai M, Yan F. Integrative analysis of single-cell and bulk multi-omics data to reveal subtype-specific characteristics and therapeutic strategies in clear cell renal cell carcinoma patients. J Cancer 2024; 15:6420-6433. [PMID: 39513109 PMCID: PMC11540511 DOI: 10.7150/jca.101451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/28/2024] [Indexed: 11/15/2024] Open
Abstract
Background: Kidney renal clear cell carcinoma (KIRC) is the most prevalent subtype of malignant renal cell carcinoma and is well known as a common genitourinary cancer. Stratifying tumors based on heterogeneity is essential for better treatment options. Methods: In this study, consensus clusters were constructed based on gene expression, DNA methylation, and gene mutation data, which were combined with multiple clustering algorithms. After identifying two heterogeneous subtypes, we analyzed the molecular characteristics, immunotherapy response, and drug sensitivity differences of each subtype. And we further integrated bulk data and single-cell RNA sequencing (scRNA-Seq) data to infer the immune cell composition and malignant tumor cell proportion of subtype-related cell subpopulations. Results: Among the two identified consensus subtypes (CS1 and CS2), CS1 was enriched in more inflammation-related and oncogenic pathways than CS2. Simultaneously, CS1 showed a worse prognosis and we found more copy number variations and BAP1 mutations in CS1. Although CS1 had a high immune infiltration score, it exhibited high expression of suppressive immune features. Based on the prediction of immunotherapy and drug sensitivity, we inferred that CS1 may respond poorly to immunotherapy and be less sensitive to targeted drugs. The analysis of bulk data integrated with single-cell data further reflected the high expression of inhibitory immune features in CS1 and the high proportion of malignant tumor cells. And CS2 contained a large number of plasmacytoid B cells, presenting an activated immune microenvironment. Finally, the robustness of our subtypes was successfully validated in four external datasets. Conclusion: In summary, we conducted a comprehensive analysis of multi-omics data with 10 clustering algorithms to reveal the molecular characteristics of KIRC patients and validated the relevant conclusions by single-cell analysis and external data. Our findings discovered new KIRC subtypes and may further guide personalized and precision treatments.
Collapse
Affiliation(s)
- Xinjia Ruan
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Chong Lai
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310012, P.R. China
| | - Leqi Li
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Bei Wang
- School of Mathematical Sciences, Jiangsu Second Normal University, Nanjing 210013, P.R. China
| | - Xiaofan Lu
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Jingya Fang
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Fangrong Yan
- State Key Laboratory of Natural Medicines, Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
5
|
Tian J, Xu T, Fan L, Sun H. Value of Contrast-Enhanced Ultrasound in the Diagnosis of Clear Cell Renal Cell Carcinoma: A Meta-Analysis. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024. [PMID: 39400336 DOI: 10.1002/jcu.23861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
This meta-analysis aims to evaluate the accuracy of contrast-enhanced ultrasound (CEUS) in diagnosing clear cell renal cell carcinoma (ccRCC). Literature published in five databases was searched. Twelve studies were included. The pooled results showed sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio of CEUS were 79% (95% CI: 75%-83%), 84% (95% CI: 80%-88%), 4.97 (95% CI: 3.86-6.39), 0.24 (95% CI: 0.20-0.30), and 20.32 (95% CI: 13.84-29.84), respectively. The area under the summary receiver operating characteristic curve was 0.86. In conclusion, CEUS demonstrates high sensitivity and specificity in diagnosing ccRCC, showing good diagnostic accuracy.
Collapse
Affiliation(s)
- Jiangke Tian
- Ultrasound Department, Beijing United Family Hospital, Beijing, China
| | - Tao Xu
- Ultrasound Department, Beijing United Family Hospital, Beijing, China
| | - Lijun Fan
- Ultrasound Department, Beijing United Family Hospital, Beijing, China
| | - Haiou Sun
- Ultrasound Department, Beijing United Family Hospital, Beijing, China
| |
Collapse
|
6
|
Yanagi M, Kiriyama T, Akatsuka J, Endo Y, Toyama Y, Kimura G, Nishimura T, Kondo Y. Role of collateral vessels on contrast-enhanced computed tomography in predicting metastatic potential for small renal cell carcinoma. Discov Oncol 2024; 15:523. [PMID: 39365374 PMCID: PMC11452607 DOI: 10.1007/s12672-024-01409-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND The presence of collateral vessels (CVs) on contrast-enhanced computed tomography is a poor prognostic factor in renal cell carcinoma (RCC), but its value in small RCC (sRCC; < 4 cm) remains unknown. In this study, we investigated whether presence of CVs is a predictor of high potential for metastasis in sRCC. METHODS We retrospectively reviewed clinical and imaging data of patients with pathologically confirmed sRCC evaluated at our institution between 2011 and 2021. All sRCCs were pathologically diagnosed by biopsy, metastasectomy, partial nephrectomy, or radical nephrectomy. CVs were defined as blood vessels of any diameter connecting the tumor with the surrounding perirenal tissues on contrast-enhanced computed tomography. The rate of metastasis-free survival (MFS), defined as the time from pathological diagnosis to confirmed metastasis, was compared among patients without CVs, those with one CV, and those with two or more CVs. RESULTS Of 141 patients, 4 (2.8%) had metastatic sRCC at initial diagnosis. In the 137 patients with nonmetastatic sRCC, the diagnosis was pathologically confirmed following radical surgery. The median follow-up period from pathological diagnosis was 73.9 months, and the overall 5-year MFS was 93.5%. The 5-year MFS was significantly poorer in patients with two or more CVs than in those with one CV (63.8% vs. 96.3%; p = 0.0003) and those without CVs (63.8% vs. 100%; p < 0.0001). CONCLUSIONS sRCCs with two or more CVs might have high potential for metastasis. Conversely, sRCCs without CVs might not be aggressive and be suitable for active surveillance.
Collapse
Affiliation(s)
- Masato Yanagi
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan.
| | - Tomonari Kiriyama
- Department of Radiology, Nippon Medical School Hospital, Tokyo, Japan
| | - Jun Akatsuka
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| | - Yuki Endo
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| | - Yuka Toyama
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| | - Go Kimura
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| | - Taiji Nishimura
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| | - Yukihiro Kondo
- Department of Urology, Nippon Medical School Hospital, Tokyo, Japan
| |
Collapse
|
7
|
Rahman NU, Neeli H, Gupta A, Sharma J, Minj P. Venous Tropism in Renal Cell Carcinoma: A Rare Imaging Presentation. Cureus 2024; 16:e70927. [PMID: 39502995 PMCID: PMC11537773 DOI: 10.7759/cureus.70927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
Renal cell carcinoma (RCC) is one of the most prevalent malignancies in adults. Accurate staging and assessment of tumor extent are critical for effective management. RCC often demonstrates a tendency for venous invasion, commonly extending into the renal vein and inferior vena cava. However, RCC extending into the gonadal veins is an uncommon manifestation. In this report, we present the imaging features of a 62-year-old woman presented with left flank pain. The ultrasound examination revealed a large hyperechoic mass in the left renal fossa, replacing almost the entire left kidney with the contiguous extension of the tumor into the left renal vein and ovarian vein. Color Doppler showed the presence of flow within the thrombus, confirming the presence of a malignant thrombus. Contrast-enhanced CT examination revealed a large heterogeneously enhancing mass in the left kidney with the contiguous extension of tumor thrombus in the left renal vein, IVC, and into the left ovarian vein. This report highlights how imaging techniques can be instrumental in detecting and characterizing rare but significant tumor extensions, which can have crucial prognostic implications for overall patient management.
Collapse
Affiliation(s)
- Nadeem Ur Rahman
- Radiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Hemanth Neeli
- Radiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Ayush Gupta
- Radiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Jitendra Sharma
- Radiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Pratik Minj
- Pathology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| |
Collapse
|
8
|
Scalco E, Pozzi S, Rizzo G, Lanzarone E. Uncertainty quantification in multi-class segmentation: Comparison between Bayesian and non-Bayesian approaches in a clinical perspective. Med Phys 2024; 51:6090-6102. [PMID: 38808956 DOI: 10.1002/mp.17189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/17/2024] [Accepted: 05/12/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Automatic segmentation techniques based on Convolutional Neural Networks (CNNs) are widely adopted to automatically identify any structure of interest from a medical image, as they are not time consuming and not subject to high intra- and inter-operator variability. However, the adoption of these approaches in clinical practice is slowed down by some factors, such as the difficulty in providing an accurate quantification of their uncertainty. PURPOSE This work aims to evaluate the uncertainty quantification provided by two Bayesian and two non-Bayesian approaches for a multi-class segmentation problem, and to compare the risk propensity among these approaches, considering CT images of patients affected by renal cancer (RC). METHODS Four uncertainty quantification approaches were implemented in this work, based on a benchmark CNN currently employed in medical image segmentation: two Bayesian CNNs with different regularizations (Dropout and DropConnect), named BDR and BDC, an ensemble method (Ens) and a test-time augmentation (TTA) method. They were compared in terms of segmentation accuracy, using the Dice score, uncertainty quantification, using the ratio of correct-certain pixels (RCC) and incorrect-uncertain pixels (RIU), and with respect to inter-observer variability in manual segmentation. They were trained with the Kidney and Kidney Tumor Segmentation Challenge launched in 2021 (Kits21), for which multi-class segmentations of kidney, RC, and cyst on 300 CT volumes are available. Moreover, they were tested considering this and other two public renal CT datasets. RESULTS Accuracy results achieved large differences across the structures of interest for all approaches, with an average Dice score of 0.92, 0.58, and 0.21 for kidney, tumor, and cyst, respectively. In terms of uncertainties, TTA provided the highest uncertainty, followed by Ens and BDC, whereas BDR provided the lowest, and minimized the number of incorrect certain pixels worse than the other approaches. Again, large differences were seen across the three structures in terms of RCC and RIU. These metrics were associated with different risk propensity, as BDR was the most risk-taking approach, able to provide higher accuracy in its prediction, but failing to assign uncertainty on incorrect segmentation in every case. The other three approaches were more conservative, providing large uncertainty regions, with the drawback of giving alert also on correct areas. Finally, the analysis of the inter-observer segmentation variability showed a significant variation among the four approaches on the external dataset, with BDR reporting the lowest agreement (Dice = 0.82), and TTA obtaining the highest score (Dice = 0.94). CONCLUSIONS Our outcomes highlight the importance of quantifying the segmentation uncertainty and that decision-makers can choose the approach most in line with the risk propensity degree required by the application and their policy.
Collapse
Affiliation(s)
- Elisa Scalco
- Institute of Biomedical Technologies (ITB), National Research Council (CNR), Segrate, Milan, Italy
| | - Silvia Pozzi
- Department of Management, Information and Production Engineering, University of Bergamo, Bergamo, Italy
| | - Giovanna Rizzo
- Institute Of Intelligent Industrial Technologies and Systems (STIIMA), National Research Council (CNR), Milan, Italy
| | - Ettore Lanzarone
- Department of Management, Information and Production Engineering, University of Bergamo, Bergamo, Italy
| |
Collapse
|
9
|
Ladurner M, Lindner AK, Rehder P, Tulchiner G. The influence of sex hormones on renal cell carcinoma. Ther Adv Med Oncol 2024; 16:17588359241269664. [PMID: 39175990 PMCID: PMC11339752 DOI: 10.1177/17588359241269664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/25/2024] [Indexed: 08/24/2024] Open
Abstract
Kidney cancer is a common malignancy that constitutes around 5% of all cancer cases. Males are twice as likely to acquire renal cell carcinoma (RCC) compared to females and experience a higher rate of mortality. These disparities indicate that sex hormone (SH)-dependent pathways may have an impact on the aetiology and pathophysiology of RCC. Examination of SH involvement in conventional signalling pathways, as well as genetics and genomics, especially the involvement of ribonucleic acid, reveal further insights into sex-related differences. An understanding of SHs and their influence on kidney cancer is essential to offer patients individualized medicine that would better meet their needs in terms of prevention, diagnosis and treatment. This review presents the understanding of sex-related differences in the clinical manifestation of kidney cancer patients and the underlying biological processes.
Collapse
Affiliation(s)
- Michael Ladurner
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Peter Rehder
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gennadi Tulchiner
- Department of Urology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| |
Collapse
|
10
|
Jung YY, Baek SH, Um JY, Ahn KS. Fangchinoline targets human renal cell carcinoma cells through modulation of apoptotic and non‑apoptotic cell deaths. Pathol Res Pract 2024; 260:155445. [PMID: 38996614 DOI: 10.1016/j.prp.2024.155445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024]
Abstract
The process of apoptosis is one of the essential processes involved in maintenance of homeostasis in the human body. It can aid to remove misfolded proteins or cellular organelles. This sequence is especially necessary in cancer cells. However, specifically targeting already apoptotic pathways can induce drug resistance in cancer cells and hence drugs can induce cell death by alternative mechanism. We investigated whether fangchinoline (FCN) can target renal carcinoma cells by inducing multiple cell death mechanisms. Both paraptosis, autophagy, and apoptosis were induced by FCN through stimulation of diverse molecular signaling pathways. FCN induced ROS production with GSH/GSSG imbalance, and ER stress. In addition, formation of autophagosome and autophagy related markers were stimulated by FCN. Moreover, FCN induced cell cycle arrest and PARP cleavage. Except for blocking protein synthesis, these three cell death pathways were found to be complementarily working together with each other. FCN also exhibited synergistic effects with paclitaxel in inducing programmed cell death in RCC cells. Our data indicates that FCN could induce apoptotic cell death and non-apoptotic cell death pathways and can be con-tribute to development of novel cancer prevention or therapy.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, the Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, the Republic of Korea.
| |
Collapse
|
11
|
Coppola A, Tessitore L, Fontana F, Piacentino F, Recaldini C, Minenna M, Capogrosso P, Minici R, Laganà D, Ierardi AM, Carrafiello G, D’Angelo F, Carcano G, Cacioppa LM, Dehò F, Venturini M. Dual-Energy Computed Tomography in Urological Diseases: A Narrative Review. J Clin Med 2024; 13:4069. [PMID: 39064110 PMCID: PMC11277677 DOI: 10.3390/jcm13144069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Dual-Energy computed tomography (DECT) with its various advanced techniques, including Virtual Non-Contrast (VNC), effective atomic number (Z-eff) calculation, Z-maps, Iodine Density Index (IDI), and so on, holds great promise in the diagnosis and management of urogenital tumours. In this narrative review, we analyze the current status of knowledge of this technology to provide better lesion characterization, improve the staging accuracy, and give more precise treatment response assessments in relation to urological tumours.
Collapse
Affiliation(s)
- Andrea Coppola
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Luigi Tessitore
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Federico Fontana
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Filippo Piacentino
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Chiara Recaldini
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Manuela Minenna
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| | - Paolo Capogrosso
- Urology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
| | - Roberto Minici
- Radiology Unit, Dulbecco University Hospital, 88100 Catanzaro, Italy
| | - Domenico Laganà
- Radiology Unit, Dulbecco University Hospital, 88100 Catanzaro, Italy
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Anna Maria Ierardi
- Radiology Unit, IRCCS Ca Granda Ospedale Maggiore Policlinico, Via Sforza 35, 20122 Milan, Italy
| | - Gianpaolo Carrafiello
- Radiology Unit, IRCCS Ca Granda Ospedale Maggiore Policlinico, Via Sforza 35, 20122 Milan, Italy
| | - Fabio D’Angelo
- Department of Medicine and Surgery, Insubria University, 21100 Varese, Italy
- Orthopedic Surgery Unit, ASST Sette Laghi, 21100 Varese, Italy
| | - Giulio Carcano
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
- Emergency and Transplant Surgery Department, ASST Sette Laghi, 21100 Varese, Italy
| | - Laura Maria Cacioppa
- Department of Clinical, Special and Dental Sciences, University Politecnica delle Marche, 60126 Ancona, Italy
- Division of Interventional Radiology, Department of Radiological Sciences, University Hospital “Azienda Ospedaliera Universitaria delle Marche”, 60126 Ancona, Italy
| | - Federico Dehò
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
- Urology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
| | - Massimo Venturini
- Diagnostic and Interventional Radiology Unit, Circolo Hospital, ASST Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Technological Innovation, Insubria University, 21100 Varese, Italy
| |
Collapse
|
12
|
Rachamala HK, Madamsetty VS, Angom RS, Nakka NM, Dutta SK, Wang E, Mukhopadhyay D, Pal K. Targeting mTOR and survivin concurrently potentiates radiation therapy in renal cell carcinoma by suppressing DNA damage repair and amplifying mitotic catastrophe. J Exp Clin Cancer Res 2024; 43:159. [PMID: 38840237 PMCID: PMC11155143 DOI: 10.1186/s13046-024-03079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) was historically considered to be less responsive to radiation therapy (RT) compared to other cancer indications. However, advancements in precision high-dose radiation delivery through single-fraction and multi-fraction stereotactic ablative radiotherapy (SABR) have led to better outcomes and reduced treatment-related toxicities, sparking renewed interest in using RT to treat RCC. Moreover, numerous studies have revealed that certain therapeutic agents including chemotherapies can increase the sensitivity of tumors to RT, leading to a growing interest in combining these treatments. Here, we developed a rational combination of two radiosensitizers in a tumor-targeted liposomal formulation for augmenting RT in RCC. The objective of this study is to assess the efficacy of a tumor-targeted liposomal formulation combining the mTOR inhibitor everolimus (E) with the survivin inhibitor YM155 (Y) in enhancing the sensitivity of RCC tumors to radiation. EXPERIMENTAL DESIGN We slightly modified our previously published tumor-targeted liposomal formulation to develop a rational combination of E and Y in a single liposomal formulation (EY-L) and assessed its efficacy in RCC cell lines in vitro and in RCC tumors in vivo. We further investigated how well EY-L sensitizes RCC cell lines and tumors toward radiation and explored the underlying mechanism of radiosensitization. RESULTS EY-L outperformed the corresponding single drug-loaded formulations E-L and Y-L in terms of containing primary tumor growth and improving survival in an immunocompetent syngeneic mouse model of RCC. EY-L also exhibited significantly higher sensitization of RCC cells towards radiation in vitro than E-L and Y-L. Additionally, EY-L sensitized RCC tumors towards radiation therapy in xenograft and murine RCC models. EY-L mediated induction of mitotic catastrophe via downregulation of multiple cell cycle checkpoints and DNA damage repair pathways could be responsible for the augmentation of radiation therapy. CONCLUSION Taken together, our study demonstrated the efficacy of a strategic combination therapy in sensitizing RCC to radiation therapy via inhibition of DNA damage repair and a substantial increase in mitotic catastrophe. This combination therapy may find its use in the augmentation of radiation therapy during the treatment of RCC patients.
Collapse
Affiliation(s)
- Hari K Rachamala
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Vijay S Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
- PolyARNA Therapeutics, One Kendal Square, Cambridge, MA, 01329, USA
| | - Ramcharan S Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Naga M Nakka
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA.
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic Florida, 4500 San Pablo Road S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
13
|
Liu K, Russo M, Ellis JS, Capua JD, Wu D, Smolinski-Zhao S, Kalva S, Arellano RS, Irani Z, Uppot R, Linderman SW, Gupta R, Aizenberg J, Srinivasan S, Som A. Transient, Image-Guided Gel-Dissection for Percutaneous Thermal Ablation. Adv Healthc Mater 2024:e2400272. [PMID: 38678431 DOI: 10.1002/adhm.202400272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/02/2024] [Indexed: 04/30/2024]
Abstract
Image-guided tumor ablative therapies are mainstay cancer treatment options but often require intra-procedural protective tissue displacement to reduce the risk of collateral damage to neighboring organs. Standard of care strategies, such as hydrodissection (fluidic injection), are limited by rapid diffusion of fluid and poor retention time, risking injury to adjacent organs, increasing cancer recurrence rates from incomplete tumor ablations, and limiting patient qualification. Herein, a "gel-dissection" technique is developed, leveraging injectable hydrogels for longer-lasting, shapeable, and transient tissue separation to empower clinicans with improved ablation operation windows and greater control. A rheological model is designed to understand and tune gel-dissection parameters. In swine models, gel-dissection achieves 24 times longer-lasting tissue separation dynamics compared to saline, with 40% less injected volume. Gel-dissection achieves anti-dependent dissection between free-floating organs in the peritoneal cavity and clinically significant thermal protection, with the potential to expand minimally invasive therapeutic techniques, especially across locoregional therapies including radiation, cryoablation, endoscopy, and surgery.
Collapse
Affiliation(s)
- Kathy Liu
- Materials Science & Mechanical Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02138, USA
| | - Mario Russo
- Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Joshua S Ellis
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - John Di Capua
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Dufan Wu
- Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
- Department of Radiology, Division of Neuroradiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Sara Smolinski-Zhao
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Sanjeeva Kalva
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ronald S Arellano
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Zubin Irani
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Raul Uppot
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Stephen W Linderman
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | - Rajiv Gupta
- Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
- Department of Radiology, Division of Neuroradiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Joanna Aizenberg
- Materials Science & Mechanical Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Shriya Srinivasan
- Materials Science & Mechanical Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02138, USA
| | - Avik Som
- Department of Radiology, Division of Interventional Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Radiology, Division of Neuroradiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| |
Collapse
|
14
|
Al-Zubi M, Al-Shami K, Sawalha L, Alguzo HM, Al Demour S, Al-Mnayyis AM, Alazab R, Al-Rawashdah SF, Alzoubi LT, Al-khawaldeh SR. Can We Predict the Grade of Clear Cell Renal Cell Carcinoma from Houns-Field Unit of Renal Lesion on Computerized Tomography Scan, a Retrospective Cross-Sectional Study. Int J Gen Med 2024; 17:1571-1577. [PMID: 38680191 PMCID: PMC11055518 DOI: 10.2147/ijgm.s452754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/16/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction Renal cell carcinoma (RCC) is a type of urological malignancy that affects approximately 2% of the global population. Imaging modalities, especially computed tomography (CT) scanning, play a critical role in diagnosing RCC. In this study, we investigated whether there is a relationship between tumour grade of clear cell RCC and HU values of renal lesions on CT scan performed before operation. Materials and Methods We conducted a retrospective analysis of 123 patients who underwent radical or partial (open or laparoscopic) nephrectomy for clear cell RCC between January 2017 and January 2021. Post-operation histopathological grades were recorded according to World Health Organization (WHO)/International Society of Urological Pathology (ISUP) 2016 grading system and divided into low grade (includes grade 1 and 2) and high grade (grade 3 and 4), and their links to age, sex, smoking habits, tumour size, and HUs of renal lesions were evaluated. Results The mean age of the patients studied was 63.02 years old. About 56.9% of the patients were low grade (grade 1 or grade 2), while 43.1% were high grade (grade 3 or 4). The mean tumour size was 6.31 cm. There were no significant differences in tumour grade according to age, sex, or smoking habits. We found a significant relation between tumour grade and HU in the pre-contrast and nephrogenic phases, with p values of 0.001 and 0.037, respectively. On the other hand, there was no significant relation linking the tumour grade to the difference in HU between these phases, where there was a p value of 0.641. Conclusion HU in the pre-contrast and nephrogenic phases in addition to tumour size on CT scan have a significant relation to clear cell RCC grade.
Collapse
Affiliation(s)
- Mohammad Al-Zubi
- Department of Surgery, Division of Urology, Yarmouk University MEdical SChool, Irbid, 21110, Jodan
| | - Khayry Al-Shami
- Department of Clinical Medical Sciences, Yarmouk University Medical school, Irbid, Jordan
| | - Leen Sawalha
- Department of Clinical Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Heyam Mahmoud Alguzo
- Department of Clinical Medical Sciences, Yarmouk University Medical school, Irbid, Jordan
| | - Saddam Al Demour
- Department of Special Surgery, Division of Urology, the University of Jordan medical School, Amman, 11972, Jordan
| | | | - Rami Alazab
- Department of Surgery & Urology, Jordan University of Science & Technology, Irbid, 21110, Jordan
| | - Samer Fathi Al-Rawashdah
- Department of Special Surgery, School of Medicine, Mutah University medical School, Karak, 61710, Jordan
| | | | | |
Collapse
|
15
|
Xu Z, Wu Y, Zhao G, Jin B, Jiang P. A novel DNA methylation signature revealed GDF6 and RCC1 as potential prognostic biomarkers correlated with cell proliferation in clear cell renal cell carcinoma. Mol Biol Rep 2023; 51:16. [PMID: 38087057 DOI: 10.1007/s11033-023-09003-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) accounts for the majority (80%-90%) of renal cell carcinoma (RCC) patients at the time of diagnosis, and approximately 15% of ccRCC patients will develop distant metastasis or recurrence during their lifetime. Increasing number of studies have revealed that the aberrant DNA methylations is closely correlated with the tumorigenesis in ccRCC. RESULTS In this study, we utilized a LASSO (least absolute shrinkage and selection operator) model to identify a combination of 13 probes-based DNA methylation signature that associated with the progression-free survival (PFS) of ccRCC patients. First, differentially methylated regions (CpGs) related to PFS and phenotypes were identified. Next, prognostic DNA methylation probes were selected from the differentially methylated probes (DMPs) and calculated risk scores to stratify patients with ccRCC. The performance of this signature was validated in an independent testing set using various analyses, including Kaplan-Meier analysis for PFS and receiver operating characteristic (ROC) curve analysis. Based on our 13-DNA methylation probes signature, ccRCC patients were successfully stratified into high- and low-risk groups. Combining DNA methylation signature with clinical variables such as T stage, M stage and tumor grade could further improve the accuracy of prediction. Moreover, we highlight two molecular biomarkers (RCC1 and GDF6) corresponding to our probes. Invitro experiments showed that knockdown of RCC1 or GDF6 in ccRCC cell lines reduced cell proliferation, which indicated that both biomarkers are associated with tumorigenesis. CONCLUSIONS The 13-probes-based DNA methylation signature has the potential to serve as an independent tool for survival outcome improvement and treatment strategy selection for ccRCC patients. In addition, our findings suggest that RCC1 and GDF6 may serve as promising markers for ccRCC.
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Zhejiang Engineering Research Center for Bladder Tumor Innovation Diagnosis and Treatment, Hangzhou, 31003, Zhejiang, China
| | - Yunfei Wu
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Zhejiang Engineering Research Center for Bladder Tumor Innovation Diagnosis and Treatment, Hangzhou, 31003, Zhejiang, China
| | - Guanan Zhao
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Department of Urology, Lishui People's Hospital, Lishui, 323050, Zhejiang, China
| | - Baiye Jin
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
- Zhejiang Engineering Research Center for Bladder Tumor Innovation Diagnosis and Treatment, Hangzhou, 31003, Zhejiang, China
| | - Peng Jiang
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
- Zhejiang Engineering Research Center for Bladder Tumor Innovation Diagnosis and Treatment, Hangzhou, 31003, Zhejiang, China.
| |
Collapse
|
16
|
Nalbant MO, Inci E. Assessment of Imaging Findings of Renal Carcinoma Subtypes with 3.0T MRI. Niger J Clin Pract 2023; 26:1750-1757. [PMID: 38044783 DOI: 10.4103/njcp.njcp_373_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/06/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND The prevalence of renal masses has escalated as a result of the augmented utilization of cross-sectional imaging techniques. The approach to managing renal masses may exhibit variability contingent upon the subtype of renal cell carcinoma (RCC). AIM This research aimed to distinguish between clear cell and papillary RCCs, utilizing dynamic contrast magnetic resonance imaging (MRI) and diffusion-weighted imaging (DWI). MATERIALS AND METHODS The study assessed the MR images of 112 patients with RCC. Two radiologists independently analyzed tumor size, vascular involvement, signal characteristics in T1- and T2-weighted sequences, the presence of hemosiderin, both microscopic and macroscopic fat content, enhancement patterns, and apparent diffusion coefficient (ADC) values derived from b-values of 1000 s/mm². RESULTS Seventy patients had clear cell RCC, and 42 had papillary. In the clear cell RCC, microscopic fat content was significantly higher than the papillary RCC (P < 0.001). However, in papillary RCC, hemosiderin content was substantially greater (P = 0.001). On T2-weighted MR images, clear cell RCCs were usually hyperintense, while papillary RCCs were hypointense (P < 0.001). Even though the rapid enhancement pattern was observed in clear cell RCCs, the progressive enhancement pattern was more prevalent in papillary RCCs (P < 0.001). CONCLUSION Hyperintensity on T2-weighted images, microscopic fat content, and rapid enhancement pattern may be indicative of clear cell RCC, whereas hypointensity on T2-weighted images, hemosiderin content, and a progressive contrast pattern may be diagnostic for papillary RCC.
Collapse
Affiliation(s)
- M O Nalbant
- Department of Radiology, University of Health Sciences, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | | |
Collapse
|
17
|
Mao X, Wang G, Wang Z, Duan C, Wu X, Xu H. Theranostic Lipid Nanoparticles for Renal Cell Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2306246. [PMID: 37747365 DOI: 10.1002/adma.202306246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/07/2023] [Indexed: 09/26/2023]
Abstract
Renal cell carcinoma (RCC) is a common urological malignancy and represents a leading threat to healthcare. Recent years have seen a series of progresses in the early diagnosis and management of RCC. Theranostic lipid nanoparticles (LNPs) are increasingly becoming one of the focuses in this field, because of their suitability for tumor targeting and multimodal therapy. LNPs can be precisely fabricated with desirable chemical compositions and biomedical properties, which closely match the physiological characteristics and clinical needs of RCC. Herein, a comprehensive review of theranostic LNPs is presented, emphasizing the generic tool nature of LNPs in developing advanced micro-nano biomaterials. It begins with a brief overview of the compositions and formation mechanism of LNPs, followed with an introduction to kidney-targeting approaches, such as passive, active, and stimulus responsive targeting. With examples provided, a series of modification strategies for enhancing the tumor targeting and functionality of LNPs are discussed. Thereafter, research advances on applications of these LNPs for RCC including bioimaging, liquid biopsy, drug delivery, physical therapy, and gene therapy are summarized and discussed from an interdisciplinary perspective. The final part highlights the milestone achievements of translation medicine, current challenges as well as future development directions of LNPs for the diagnosis and treatment of RCC.
Collapse
Affiliation(s)
- Xiongmin Mao
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Guanyi Wang
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zijian Wang
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chen Duan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoliang Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hua Xu
- Department of Urology, Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
18
|
Tang J, Yao F, Yao Z, Xing XL. Characterization of tumor microenvironment and sensitive chemotherapy drugs based on cuproptosis-related signatures in renal cell carcinoma. Aging (Albany NY) 2023; 15:9695-9717. [PMID: 37728407 PMCID: PMC10564438 DOI: 10.18632/aging.205043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
Cuproptosis is a novel type of copper-induced cell death and is considered as a new therapeutic target for many cancers. Distant metastases occur in about 40% of patients with advanced renal cell carcinoma (RCC), with a poor 5-year prognosis of about 10%. Through a series of comprehensive analyses, four differentially expressed cuproptosis-related lncRNAs (DECRLs) were identified as candidate biomarkers for RCC. The risk model constructed by using these four DECRLs can better predict the prognosis of patients with RCC, which is determined by the receiver operating characteristic (Time dependent area under curve value at 1-year, 3-year, 5-year, and 10-year were 0.82, 0.80, 0.76, and 0.73 respectively). There were significant differences in immune status between high-risk and low-risk RCC patients. The differentially expressed gene enrichment terms between high- and low-risk patients was also dominated by immune-related terms. The risk score was also correlated with immunotherapy as measured by the tumor immune dysfunction and exclusion (TIDE) score. In addition, we also found that the sensitivity of many chemotherapy drugs varies widely between high- and low-risk patients. The sensitivity of the three chemotherapy drugs (AZD4547, Vincristine, and WEHI-539) varied among high- and low-risk patients, and was significantly negatively correlated with risk values, suggesting that they could be used as clinical treatment drugs for RCC. Our study not only obtained four potential biomarkers, but also provided guidance for immunotherapy and chemotherapy treatment of RCC, as well as new research strategies for the screening of other cancer biomarkers and sensitive drugs.
Collapse
Affiliation(s)
- Jiefu Tang
- The First Affiliated Hospital of Hunan University of Medicine, School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua 418000, Hunan, P.R. China
| | - Fan Yao
- The First Affiliated Hospital of Hunan University of Medicine, School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua 418000, Hunan, P.R. China
| | - Zhiyong Yao
- The First Affiliated Hospital of Hunan University of Medicine, School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua 418000, Hunan, P.R. China
| | - Xiao-Liang Xing
- The First Affiliated Hospital of Hunan University of Medicine, School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua 418000, Hunan, P.R. China
| |
Collapse
|
19
|
Zhao Y, Lin X, Zeng W, Qin X, Miao B, Gao S, Liu J, Li Z. Berberine inhibits the progression of renal cell carcinoma cells by regulating reactive oxygen species generation and inducing DNA damage. Mol Biol Rep 2023:10.1007/s11033-023-08381-w. [PMID: 37217616 DOI: 10.1007/s11033-023-08381-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/09/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Berberine is a natural isoquinoline alkaloid that has been shown to have antitumor properties in a growing number of studies. However, its role in renal cell carcinoma remains unclear. This study investigates berberine's effect and mechanism in renal cell carcinoma. METHODS The methyl-tetrazolium, colony formation, and lactate dehydrogenase assay were used to detect proliferation and cytotoxicity, respectively. Flow cytometry, caspase-Glo 3/7 assay, and adenosine triphosphate assay were used to detect apoptosis and the adenosine triphosphate levels. Wound healing and transwell assay were used to examine the migration ability of renal cell carcinoma cells. Besides, the level of reactive oxygen species (ROS) was explored using a DCFH-DA-based kit. Additionally, western blot and Immunofluorescence assay was taken to determine the levels of relative proteins. RESULTS In vitro, our findings indicated that the proliferation and migration of renal cell carcinoma cells treated with berberine in various concentrations were inhibited, while the level of ROS and apoptosis rate were increased. Furthermore, The results of western blot showed that the expression of Bax, Bad, Bak, Cyto c, Clv-Caspase 3, Clv-Caspase 9, E-cadherin, TIMP-1and γH2AX were up-regulated, while Bcl-2, N-cadherin, Vimentin, Snail, Rad51 and PCNA were down-regulated after treating with berberine with various concentration. CONCLUSION The result of this study revealed that berberine inhibits renal cell carcinoma progression via regulating ROS generation and inducing DNA break.
Collapse
Affiliation(s)
- Yuwan Zhao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Xinghua Lin
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Wenfeng Zeng
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Xingzhang Qin
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Bailiang Miao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Sheng Gao
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China
| | - Jianjun Liu
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
| | - Zhuo Li
- Department of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, 57 Renmin Street South, 524001, Zhanjiang, Guangdong, China.
| |
Collapse
|
20
|
Kumar S, Virarkar M, Vulasala SSR, Daoud T, Ozdemir S, Wieseler C, Vincety-Latorre F, Gopireddy DR, Bhosale P, Lall C. Magnetic Resonance Imaging Virtual Biopsy of Common Solid Renal Masses-A Pictorial Review. J Comput Assist Tomogr 2023; 47:186-198. [PMID: 36790908 DOI: 10.1097/rct.0000000000001424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
ABSTRACT The expanded application of radiologic imaging resulted in an increased incidence of renal masses in the recent decade. Clinically, it is difficult to determine the malignant potential of the renal masses, thus resulting in complex management. Image-guided biopsies are the ongoing standard of care to identify molecular variance but are limited by tumor accessibility and heterogeneity. With the evolving importance of individualized cancer therapies, radiomics has displayed promising results in the identification of tumoral mutation status on routine imaging. This article discusses how magnetic resonance imaging features can guide a radiologist toward identifying renal mass characteristics.
Collapse
Affiliation(s)
- Sindhu Kumar
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Mayur Virarkar
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Sai Swarupa R Vulasala
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Taher Daoud
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Savas Ozdemir
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Carissa Wieseler
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | | | - Dheeraj R Gopireddy
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| | - Priya Bhosale
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chandana Lall
- From the Department of Radiology, University of Florida College of Medicine, Jacksonville, FL
| |
Collapse
|
21
|
Ji J, Xu Y, Xie M, He X, Ren D, Qiu T, Liu W, Chen Z, Shi W, Zhang Z, Wang X, Wang W, Ma J, Qian Q, Jing A, Ma X, Qin J, Ding Y, Geng T, Yang J, Sun Z, Liu W, Liu S, Liu B. VHL-HIF-2α axis-induced SEMA6A upregulation stabilized β-catenin to drive clear cell renal cell carcinoma progression. Cell Death Dis 2023; 14:83. [PMID: 36739418 PMCID: PMC9899268 DOI: 10.1038/s41419-023-05588-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 02/06/2023]
Abstract
SEMA6A is a multifunctional transmembrane semaphorin protein that participates in various cellular processes, including axon guidance, cell migration, and cancer progression. However, the role of SEMA6A in clear cell renal cell carcinoma (ccRCC) is unclear. Based on high-throughput sequencing data, here we report that SEMA6A is a novel target gene of the VHL-HIF-2α axis and overexpressed in ccRCC. Chromatin immunoprecipitation and reporter assays revealed that HIF-2α directly activated SEMA6A transcription in hypoxic ccRCC cells. Wnt/β-catenin pathway activation is correlated with the expression of SEMA6A in ccRCC; the latter physically interacted with SEC62 and promoted ccRCC progression through SEC62-dependent β-catenin stabilization and activation. Depletion of SEMA6A impaired HIF-2α-induced Wnt/β-catenin pathway activation and led to defective ccRCC cell proliferation both in vitro and in vivo. SEMA6A overexpression promoted the malignant phenotypes of ccRCC, which was reversed by SEC62 depletion. Collectively, this study revealed a potential role for VHL-HIF-2α-SEMA6A-SEC62 axis in the activation of Wnt/β-catenin pathway. Thus, SEMA6A may act as a potential therapeutic target, especially in VHL-deficient ccRCC.
Collapse
Affiliation(s)
- Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuxin Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Mengru Xie
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xingbei He
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Dexu Ren
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Teng Qiu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wenwen Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Zefeng Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wen Shi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Zhen Zhang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xiujun Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Weiling Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jinming Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Qilan Qian
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Aixin Jing
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xinhui Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jingting Qin
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yuanyuan Ding
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Ting Geng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Jiayan Yang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Zhichao Sun
- Department of Pathology, The Second People's Hospital of Lianyungang, 41 Hailian East Road, Haizhou, Lianyungang, 222006, Jiangsu, PR China
| | - Wei Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Shunfang Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 430030, PR China.
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
22
|
Das CJ, Aggarwal A, Singh P, Nayak B, Yadav T, Lal A, Gorsi U, Batra A, Shamim SA, Duara BK, Arulraj K, Kaushal S, Seth A. Imaging Recommendations for Diagnosis, Staging, and Management of Renal Tumors. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1759718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
AbstractRenal cell carcinomas accounts for 2% of all the cancers globally. Most of the renal tumors are detected incidentally. Ultrasound remains the main screening modality to evaluate the renal masses. A multi -phase contrast enhanced computer tomography is must for characterizing the renal lesions. Imaging plays an important role in staging, treatment planning and follow up of renal cancers. In this review , we discuss the imaging guidelines for the management of renal tumors.
Collapse
Affiliation(s)
- Chandan J Das
- Department of Radiodiagnosis and Interventional Radiology, AIIMS, New Delhi, India
| | - Ankita Aggarwal
- Department of Radiodiagnosis, VMMC and SJH, New Delhi, India
| | | | - B Nayak
- Department of Urology, AIIMS, New Delhi, India
| | - Taruna Yadav
- Department of Radiodiagnosis, Jodhpur, Rajasthan, India
| | - Anupam Lal
- Department of Radiodiagnosis, PGI, Chandigarh, India
| | - Ujjwal Gorsi
- Department of Radiodiagnosis, PGI, Chandigarh, India
| | - Atul Batra
- Department of Medical Oncology, AIIMS, IRCH, New Delhi, India
| | | | | | | | | | - Amlesh Seth
- Department of Urology, AIIMS, New Delhi, India
| |
Collapse
|
23
|
Rizzo A, Racca M, Dall’Armellina S, Rescigno P, Banna GL, Albano D, Dondi F, Bertagna F, Annunziata S, Treglia G. The Emerging Role of PET/CT with PSMA-Targeting Radiopharmaceuticals in Clear Cell Renal Cancer: An Updated Systematic Review. Cancers (Basel) 2023; 15:355. [PMID: 36672305 PMCID: PMC9857064 DOI: 10.3390/cancers15020355] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recent articles proposed the employment of positron emission tomography/computed tomography (PET/CT) with prostate-specific membrane antigen (PSMA)-targeting radiopharmaceuticals in clear cell renal cell carcinoma (ccRCC). METHODS The authors performed a comprehensive literature search of studies on the performance of PET/CT with PSMA-targeting radiopharmaceuticals in ccRCC. Original articles concerning this imaging examination were included in newly diagnosed ccRCC patients and ccRCC patients with disease recurrence. RESULTS A total of sixteen papers concerning the diagnostic performance of PSMA-targeted PET/CT in ccRCC (331 patients) were included in this systematic review. The included articles demonstrated an excellent detection rate of PSMA-targeting PET/CT in ccRCC. CONCLUSIONS PSMA-targeted PET/CT seems promising in detecting ccRCC lesions as well as in discriminating the presence of aggressive phenotypes. Prospective multicentric studies are warranted to strengthen the role of PSMA-targeting PET/CT in ccRCC.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy
| | - Sara Dall’Armellina
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy
| | - Pasquale Rescigno
- Department of Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | | | - Domenico Albano
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Bertagna
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
24
|
Pietersen PI, Lynggård Bo Madsen J, Asmussen J, Lund L, Nielsen TK, Pedersen M, Engvad B, Graumann O. Multiparametric magnetic resonance imaging for characterizing renal tumors: A validation study of the algorithm presented by Cornelis et al. J Clin Imaging Sci 2023; 13:7. [PMID: 36908585 PMCID: PMC9992978 DOI: 10.25259/jcis_124_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Objectives In the last decade, the incidence of renal cell carcinoma (RCC) has been rising, with the greatest increase observed for solid tumors. Magnetic resonance imaging (MRI) protocols and algorithms have recently been available for classifying RCC subtypes and benign subtypes. The objective of this study was to prospectively validate the MRI algorithm presented by Cornelis et al. for RCC classification. Material and Methods Over a 7-month period, 38 patients with 44 renal tumors were prospectively included in the study and received an MRI examination in addition to the conventional investigation program. The MRI sequences were: T2-weighted, dual chemical shift MRI, diffusion-weighted imaging (DWI), and dynamic contrast-enhanced T1-weighted in wash-in and wash-out phases. The images were evaluated according to the algorithm by two experienced, blinded radiologists, and the histopathological diagnosis served as the gold standard. Results Of 44 tumors in 38 patients, only 8 tumors (18.2%) received the same MRI diagnosis according to the algorithm as the histopathological diagnosis. MRI diagnosed 16 angiomyolipoma, 14 clear cell RCC (ccRCC), 12 chromophobe RCC (chRCC), and two papillary RCC (pRCC), while histopathological examination diagnosed 24 ccRCC, four pRCC, one chRCC, and one mixed tumor of both pRCC and chRCC. Malignant tumors were statistically significantly larger than the benign (3.16 ± 1.34 cm vs. 2.00 ± 1.04 cm, P = 0.006). Conclusion This prospective study could not reproduce Cornelis et al.'s results and does not support differentiating renal masses using multiparametric MRI without percutaneous biopsy in the future. The MRI algorithm showed few promising results to categorize renal tumors, indicating histopathology for clinical decisions and follow-up regimes of renal masses are still required.
Collapse
Affiliation(s)
| | - Janni Lynggård Bo Madsen
- Research and Innovation Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jon Asmussen
- Department of Radiology, Odense University Hospital, Odense, Denmark
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark
| | | | - Michael Pedersen
- Department of Clinical Medicine - Comparative Medicine Lab, Aarhus University Hospital, Aarhus, Denmark
| | - Birte Engvad
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Ole Graumann
- Department of Radiology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
25
|
Haghighi N, Doosti A, Kiani J. Evaluation of Apoptosis, Cell Proliferation and Cell Cycle Progression by Inactivation of the NEAT1 Long Noncoding RNA in a Renal Carcinoma Cell Line Using CRISPR/Cas9. IRANIAN JOURNAL OF BIOTECHNOLOGY 2023; 21:e3180. [PMID: 36811109 PMCID: PMC9938936 DOI: 10.30498/ijb.2022.310632.3180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 03/06/2022] [Indexed: 02/24/2023]
Abstract
Background Long noncoding RNAs (lncRNAs) play an important role in cellular mechanisms including transcription, translation, and apoptosis. NEAT1 is one of the essential types of lncRNAs in humans that can bind to active genes and modify their transcription. NEAT1 upregulation in various forms of cancer such as kidney cancer has been reported. Kidney cancer accounts for approximately 3% of all cancers worldwide and occurs almost twice as often in men as in women. Objectives This study has been performed to knockout the NEAT1 gene using the CRISPR/Cas9 technique in the Renal Cell Carcinoma ACHN cell line and to evaluate its effects on cancer progression and apoptosis. Material and Methods Two specific (single guide RNA (sgRNA) sequences for the NEAT1 gene were designed by CHOPCHOP software. These sequences were then cloned into plasmid pSpcas9, and recombinant vectors PX459-sgRNA1 and PX459-sgRNA2 were generated. ACHN cells were transfected using recombinant vectors carrying sgRNA1 and sgRNA2. The expression level of apoptosis-related genes was assessed by real-time PCR. Annexin, MTT and cell scratch tests were performed to evaluate the survival, proliferation, and migration of the knocked out cells, respectively. Results The results have shown successful knockout of the NEAT1 gene in the cells of the treatment group. Expressions of P53, BAK, BAX and FAS genes in the cells of the treatment group (NEAT1 knockout) showed significant increases in expression compared to the cells of the control group (P <0.01). Additionally, decreased expression of BCL2 and survivin genes was observed in knockout cells compared to the control group (p <0.05). In addition, in the cells of the treatment group compared to control cells, a significant decrease in cell viability, ability to migrate and cell growth and proliferation was observed. Conclusion Inactivation of the NEAT1 gene in ACHN cell line using CRISPR/Cas9 technology elevated apoptosis and reduced cell survival and proliferation which makes it a novel target for kidney cancer therapeutics.
Collapse
Affiliation(s)
- Nastaran Haghighi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Jafar Kiani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
26
|
Chrabańska M, Rynkiewicz M, Kiczmer P, Drozdzowska B. Immunohistochemical Expression of CD44, MMP-2, MMP-9, and Ki-67 as the Prognostic Markers in Non-Clear Cell Renal Cell Carcinomas-A Prospective Cohort Study. J Clin Med 2022; 11:jcm11175196. [PMID: 36079127 PMCID: PMC9457518 DOI: 10.3390/jcm11175196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
CD44 is the most frequently reported marker of the cancer stem cells in renal cell carcinoma (RCC). Matrix metalloproteinases MMP-2 and MMP-9 are key regulators of tumor invasion and metastasis. The aim of this study was to investigate the clinicopathologic and prognostic values of the immunohistochemical expression of CD44, MMP2, MMP9, and Ki-67 in papillary and chromophobe RCCs. In the case of papillary RCC, MMP-2 expression was positively correlated with patient age (p < 0.05), while CD44 expression was positively correlated with tumor stage (τ = 0.26, p < 0.05). Moreover, CD44 expression positively correlated with MMP-9 (τ = 0.39, p < 0.05). In the case of chromophobe RCC, only Ki-67 expression was negatively correlated with tumor stage (τ = −0.44, p < 0.05). During follow-up, a death was documented in 6 patients with papillary RCC. In these patients, CD44 expression was not a significant factor affecting the overall survival of patients (p > 0.05), whereas there was a positive correlation between increased MMP-9 expression and shorter overall survival (p < 0.05). Taken together, carcinogenesis in papillary RCC is probably dependent on both cancer stem cells and metalloproteinases activity. Expression of CD44 and MMP-9 can significantly improve the prediction of papillary RCC prognosis in the future.
Collapse
|
27
|
Renal cell carcinoma management: A step to nano-chemoprevention. Life Sci 2022; 308:120922. [PMID: 36058262 DOI: 10.1016/j.lfs.2022.120922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022]
Abstract
Renal cell carcinoma (RCC) is one of the most common kidney cancers, responsible for nearly 90 % of all renal malignancies. Despite the availability of many treatment strategies, RCC still remains to be an incurable disease due to its resistivity towards conventional therapies. Nanotechnology is an emerging field of science that offers newer possibilities in therapeutics including cancer medicine, specifically by targeted delivery of anticancer drugs. Several phytochemicals are known for their anti-cancer properties and have been regarded as chemopreventive agents. However, the hydrophobic nature of many phytochemicals decreases its bioavailability and distribution, thus showing limited therapeutic effect. Application of nanotechnology to enhance chemoprevention is an effective strategy to increase the bioavailability of phytochemicals and thereby its therapeutic efficacy. The present review focuses on the utility of nanotechnology in RCC treatment and chemopreventive agents of RCC. We have also visualized the future prospects of nanomolecules in the prevention and cure of RCC.
Collapse
|
28
|
Sriram JD, van de Beek I, Johannesma PC, van Werkum MH, van der Wel TJWT, Wessels EM, Gille HJJP, Houweling AC, Postmus PE, Smit HJM. Birt-Hogg-Dubé syndrome in apparent primary spontaneous pneumothorax patients; results and recommendations for clinical practice. BMC Pulm Med 2022; 22:325. [PMID: 36028846 PMCID: PMC9414409 DOI: 10.1186/s12890-022-02107-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background Birt-Hogg-Dubé syndrome (BHD) is an inherited disease caused by pathogenic variants in the FLCN gene. One of the characteristics is the increased risk for spontaneous pneumothorax, likely due to the presence of pulmonary cysts mainly distributed under the carina. Due to variable expression and lack of awareness, BHD is likely to be underdiagnosed. We aimed to examine the prevalence of BHD in patients presenting with an apparent primary spontaneous pneumothorax and to evaluate the contribution of chest CT in establishing the diagnosis. Methods Patients who presented with apparent primary spontaneous pneumothorax between 2004 and 2017 in a large Dutch teaching hospital were enrolled in this quantitative cross-sectional study. A questionnaire was sent to eligible patients. Patients who completed the questionnaire and consented to further participation were invited to visit the hospital for genetic testing and low dose, volumetric chest CT. Results Genetic testing was performed in 88 patients with apparent primary spontaneous pneumothorax. Three patients were found to have a pathogenic variant in the FLCN gene (3.4%). No variants of unknown significance were detected. Pulmonary cysts were detected in 14 out of 83 participants with an available chest CT, six had more than one cyst. All three patients with BHD had multiple pulmonary cysts. Conclusions Based on previous literature and the present study, we believe that performing a chest CT in every patient presenting with primary spontaneous pneumothorax is justified. Subsequent genetic testing of the FLCN gene should be considered when multiple pulmonary cysts are present. Trial registration The study was registered at clinicaltrials.gov with reference NCT02916992. Summary at a glance Three out of 88 patients with an apparent primary spontaneous pneumothorax were diagnosed with Birt-Hogg-Dubé syndrome in this study and all three had multiple pulmonary cysts. We believe that performing a chest CT in every patient with an apparent primary spontaneous pneumothorax is justified to identify underlying diseases.
Collapse
Affiliation(s)
- Jincey D Sriram
- Department of Pulmonology, Rijnstate Hospital Arnhem, Wagnerlaan 55, 6815 AD, Arnhem, The Netherlands.
| | - Irma van de Beek
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Paul C Johannesma
- Department of Pulmonology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.,Department of Surgery, Gelderse Vallei Ziekenhuis, Willy Brandtlaan 10, 6716 RP, Ede, The Netherlands
| | - Michiel H van Werkum
- Department of Radiology, Rijnstate Hospital, Wagnerlaan 55, 6815 AD, Arnhem, The Netherlands
| | - Tijmen J W T van der Wel
- Department of Pulmonology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Elise M Wessels
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Hans J J P Gille
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Pieter E Postmus
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Hans J M Smit
- Department of Pulmonology, Rijnstate Hospital Arnhem, Wagnerlaan 55, 6815 AD, Arnhem, The Netherlands
| |
Collapse
|
29
|
Sreepada A, Tiwari M, Pal K. Adhesion G protein-coupled receptor gluing action guides tissue development and disease. J Mol Med (Berl) 2022; 100:1355-1372. [PMID: 35969283 DOI: 10.1007/s00109-022-02240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [ |