1
|
Rosellini M, Mollica V, Marchetti A, Coluccelli S, Giunchi F, Tassinari E, Danielli L, Maloberti T, Ricci C, Fiorentino M, Biase DD, Massari F. Insights into the impact of chromosome 3p mutations in advanced renal cell carcinoma treated with immune-based combinations or targeted therapy: A single-center experience. Pathol Res Pract 2025; 270:155964. [PMID: 40233528 DOI: 10.1016/j.prp.2025.155964] [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: 02/28/2025] [Revised: 03/18/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Immune-based combinations have transformed first-line treatment for metastatic renal cell carcinoma (mRCC), but reliable biomarkers for patient selection remain elusive. Chromosome 3p mutations (e.g., VHL, PBRM1, SETD2, BAP1) have shown inconsistent prognostic and predictive value. This retrospective study assessed the prognostic impact of tissue-based biomarkers, focusing on 3p mutations in mRCC. PATIENTS AND METHODS A single-center retrospective analysis included mRCC patients treated with immunocombinations or tyrosine kinase inhibitors (TKIs). We evaluated mutations in 14 genes, including VHL, PBRM1, SETD2, and BAP1. Primary endpoints were overall survival (OS) and progression-free survival (PFS). Prognostic factors were analyzed using univariate and multivariate models. RESULTS Among the included 38 patients, the most common mutations were in VHL (45 %), PBRM1 (42 %), and SETD2 (26 %), with these latter more frequent in males (p = 0.012). Most patients (74 %) received immune-based combinations; 26 % received TKIs. SETD2 mutations were associated with primary refractoriness (30 %). Median OS was not reached; brain metastases (p = 0.001) and BAP1 mutations (p = 0.025) predicted worse OS, while PBRM1 mutations trended toward improved OS (p = 0.845). Median PFS was 14.1 months. Higher tumor grade (p = 0.038) and worse ECOG PS (p = 0.008) negatively impacted PFS, while 3p mutations showed no significant effect on PFS. CONCLUSIONS ECOG PS and brain metastases were confirmed as poor prognostic factors. VHL and PBRM1 mutations may suggest a better prognosis, while SETD2 and BAP1 mutations portend worse outcomes. Larger studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Matteo Rosellini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Marchetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Sara Coluccelli
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| | - Francesca Giunchi
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| | - Elisa Tassinari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Linda Danielli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| | - Costantino Ricci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Pathology Unit, Maggiore Hospital-AUSL Bologna, Bologna, Italy
| | - Michelangelo Fiorentino
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Pathology Unit, Maggiore Hospital-AUSL Bologna, Bologna, Italy
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy; Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna 40126, Italy
| | - Francesco Massari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| |
Collapse
|
2
|
Hein D, Christie A, Holcomb M, Xie B, Jain AJ, Vento J, Rakheja N, Hamza Shakur A, Christley S, Cowell LG, Brugarolas J, Jamieson A, Kapur P. Prompts to Table: Specification and Iterative Refinement for Clinical Information Extraction with Large Language Models. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322107. [PMID: 39990557 PMCID: PMC11844613 DOI: 10.1101/2025.02.11.25322107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Extracting structured data from free-text medical records at scale is laborious, and traditional approaches struggle in complex clinical domains. We present a novel, end-to-end pipeline leveraging large language models (LLMs) for highly accurate information extraction and normalization from unstructured pathology reports, focusing initially on kidney tumors. Our innovation combines flexible prompt templates, the direct production of analysis-ready tabular data, and a rigorous, human-in-the-loop iterative refinement process guided by a comprehensive error ontology. Applying the finalized pipeline to 2,297 kidney tumor reports with pre-existing templated data available for validation yielded a macro-averaged F1 of 0.99 for six kidney tumor subtypes and 0.97 for detecting kidney metastasis. We further demonstrate flexibility with multiple LLM backbones and adaptability to new domains utilizing publicly available breast and prostate cancer reports. Beyond performance metrics or pipeline specifics, we emphasize the critical importance of task definition, interdisciplinary collaboration, and complexity management in LLM-based clinical workflows.
Collapse
Affiliation(s)
- David Hein
- University of Texas Southwestern Medical Center (UTSW) Lyda Hill Department of Bioinformatics
| | | | - Michael Holcomb
- University of Texas Southwestern Medical Center (UTSW) Lyda Hill Department of Bioinformatics
| | - Bingqing Xie
- UTSW Department of Internal Medicine, Division of Hematology & Oncology
| | - AJ Jain
- University of Texas Southwestern Medical Center (UTSW) Lyda Hill Department of Bioinformatics
| | - Joseph Vento
- UTSW Department of Internal Medicine, Division of Hematology & Oncology
| | - Neil Rakheja
- UTSW Harold C Simmons Comprehensive Cancer Center
| | - Ameer Hamza Shakur
- University of Texas Southwestern Medical Center (UTSW) Lyda Hill Department of Bioinformatics
| | - Scott Christley
- Department of Health Data Science and Biostatistics, Peter O’Donnell Jr. School of Public Health, UTSW
| | - Lindsay G. Cowell
- Department of Health Data Science and Biostatistics, Peter O’Donnell Jr. School of Public Health, UTSW
| | | | - Andrew Jamieson
- University of Texas Southwestern Medical Center (UTSW) Lyda Hill Department of Bioinformatics
| | - Payal Kapur
- UTSW Harold C Simmons Comprehensive Cancer Center
- UTSW Department of Pathology
| |
Collapse
|
3
|
Gupta S, Cheville JC. Renal Neoplasia: Rare Subtypes and Uncommon Clinical Presentations. Surg Pathol Clin 2025; 18:157-174. [PMID: 39890302 DOI: 10.1016/j.path.2024.09.001] [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] [Indexed: 02/03/2025]
Abstract
Herein, the authors have discussed a series of uncommon familial kidney cancer syndromes (including hyperparathyroidism-jaw tumor syndrome and PTEN hamartoma tumor syndrome), sporadically occurring tumors (BRAF and MTOR pathway-mutated tumors, and juxtaglomerular cell tumors), and uncommon patterns of well-established subtypes of kidney cancer (mucinous tubular spindle cell carcinoma, fumarate hydratase-deficient, and TFE3-rearranged renal cell carcinoma). The rarity of these tumors often leads to diagnostic odysseys for pathologists and patients. Appropriate classification of these rare tumors has implications for screening at-risk family members in the case of hereditary tumor predisposition syndromes, accurate prognostication, and appropriate patient selection for clinical trials.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
4
|
Rinaldi L, Senatore E, Feliciello S, Chiuso F, Insabato L, Feliciello A. Kidney cancer: From tumor biology to innovative therapeutics. Biochim Biophys Acta Rev Cancer 2025; 1880:189240. [PMID: 39674419 DOI: 10.1016/j.bbcan.2024.189240] [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: 07/01/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Renal cell carcinoma (RCC) constitutes the most frequent kidney cancer of the adult population and one of the most lethal malignant tumors worldwide. RCC often presents without early symptoms, leading to late diagnosis. Prognosis varies widely based on the stage of cancer at diagnosis. In the early-stage, localized RCC has a relatively good prognosis, while advanced or metastatic RCC has a poor outcome. Obesity, smoking, genetic mutations and family history are all considered risk factors for RCC, while inherited disorders, such as Tuberous Sclerosis and von Hippel-Lindau syndrome, are causally associated with RCC development. Genetic screening, deep sequencing analysis, quantitative proteomics and immunostaining analysis on RCC tissues, biological fluids and blood samples have been employed to identify novel biomarkers, predisposing factors and therapeutic targets for RCC with important clinical implications for patient treatment. Combined approaches of gene-targeting strategies coupled to a deep functional analysis of cancer cell biology, both in vitro and in appropriate animal models of RCC, significantly contributed to identify and characterize relevant pathogenic mechanisms underlying development and progression of RCC. These studies provided also important cues for the generation of novel target-specific therapeutics that selectively restore deranged cancer cell signalling and dysfunctional immune checkpoints, positively impacting on the survival rate of treated RCC patients. In this review, we will describe the recent discoveries concerning the most relevant pathogenic mechanisms of RCC and will highlight novel therapeutic strategies that interrupt oncogenic pathways and restore immune defences in RCC patients.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Emanuela Senatore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Stella Feliciello
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Luigi Insabato
- Department of Advanced Biomedical Sciences, University Hospital Federico II, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
5
|
Wagh H, Bhattacharya S. Targeted therapy with polymeric nanoparticles in PBRM1-mutant biliary tract cancers: Harnessing DNA damage repair mechanisms. Crit Rev Oncol Hematol 2024; 204:104505. [PMID: 39255911 DOI: 10.1016/j.critrevonc.2024.104505] [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: 07/01/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Biliary tract cancers (BTCs) are aggressive malignancies with a dismal prognosis that require intensive targeted therapy. Approximately 10 % of BTCs have PBRM1 mutations, which impede DNA damage repair pathways and make cancer cells more susceptible to DNA-damaging chemicals. This review focus on development of poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles targeting delivery system to selectively deliver chemotherapy into PBRM1-deficient BTC cells. These nanoparticles improve therapy efficacy by increasing medication targeting and retention at tumour locations. In preclinical studies, pharmacokinetic profile of this nanoparticle was encouraging and supported its ability to achieve extended circulation time with high drug accumulation in tumor. The review also highlights potential of Pou3F3:I54N to expedite bioassays for patient selection in BTC targeted therapies.
Collapse
Affiliation(s)
- Hrushikesh Wagh
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India.
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India.
| |
Collapse
|
6
|
Camp SY, He MX, Cuoco MS, Saad E, Pimenta E, Meli K, Bakouny Z, Labaki C, Titchen BM, Kang YJ, Horst J, Trowbridge R, Shannon E, Helvie K, Thorner AR, Vigneau S, Mayorga A, Kodali J, Lachmayr H, Bemus M, Park J, Choueiri T, Bi K, Van Allen EM. Single-cell epigenetic profiling reveals an interferon response-high program associated with BAP1 deficiency in kidney cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623837. [PMID: 39605676 PMCID: PMC11601305 DOI: 10.1101/2024.11.15.623837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Renal cell carcinoma (RCC) is characterized by recurrent somatic mutations in epigenetic regulators, which stratify patients into clinically significant subgroups with distinct prognoses and treatment responses. However, the cell type-specific epigenetic landscape of RCC-broadly and in the context of these mutations-is incompletely understood. To investigate these open questions, we integrated single nucleus ATAC sequencing data from RCC tumors across four independent cohorts. In clear cell RCC tumors, we identified four shared malignant epigenetic programs related to angiogenesis, proximal tubule-like features, interferon (IFN) signaling, and one that lacked distinct genomic regions with increased accessibility. Among the mutated epigenetic regulators, BAP1 mutation exhibited the most significant impact on chromatin accessibility in tumor cells, and the associated epigenetic changes were linked to IFN response. We identify multiple potential sources of elevated IFN signaling in these lesions, such as increased immune infiltration and increased accessibility and expression of an IFN-associated ERV, ERV3-16A3_LTR. We find that the expression of ERV3-16A3_LTR may itself be a negative prognostic biomarker in ccRCC. Our findings highlight the convergence of malignant epigenetic programs across ccRCC tumors and suggest that BAP1 loss, potentially through ERV3-16A3_LTR dysregulation, is associated with an IFN response-high epigenetic program.
Collapse
Affiliation(s)
- Sabrina Y. Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Graduate Program in Biophysics, Boston, MA 02115, USA
| | - Michael S. Cuoco
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, CA 92093, USA
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Erica Pimenta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Meli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Chris Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Breanna M. Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Yun Jee Kang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jack Horst
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Rachel Trowbridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Karla Helvie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Aaron R. Thorner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sébastien Vigneau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Angie Mayorga
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jahnavi Kodali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hannah Lachmayr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Meredith Bemus
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Toni Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
7
|
Anderson AC, Ho J, Hall ET, Hannan R, Liao JJ, Louie AV, Ma TM, Psutka SP, Rengan R, Siva S, Swaminath A, Tachiki L, Tang C, Teh BS, Tsai J, Tykodi SS, Weg E, Zaorsky NG, Lo SS. Focal therapy for oligometastatic and oligoprogressive renal cell carcinoma: a narrative review. Future Oncol 2024; 20:2573-2588. [PMID: 39258792 PMCID: PMC11534104 DOI: 10.1080/14796694.2024.2389769] [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: 05/22/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Metastatic renal cell carcinoma (RCC) can present with oligometastatic disease and/or develop oligoprogression following systemic therapy. Cytoreductive and focal metastasis-directed therapy options include resection, stereotactic ablative radiation and thermal ablation. Aggressive focal therapy may allow delay in initiation of or modification to systemic therapy and improve clinical outcomes. In this narrative review we synthesize current practice guidelines and prospective data on focal therapy management options and highlight future research. Patient selection and the choice of focal treatment techniques are controversial due to limited and heterogeneous data and patients may benefit from multidisciplinary evaluation. Prospective comparative trials with clearly defined inclusion criteria and relevant end points are needed to clarify the risks and benefits of different approaches.
Collapse
Affiliation(s)
- August C Anderson
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Joel Ho
- Pfizer Inc., Bothell, WA 98011, USA
| | - Evan T Hall
- University of Washington, Division of Hematology & Oncology, Seattle, WA 98195,USA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
| | - Raquibul Hannan
- The University of Texas Southwestern Medical Center, Radiation Oncology, Dallas, TX 75235, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Kidney Cancer Program, Dallas, TX75235, USA
| | - Jay J Liao
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Alexander V Louie
- Sunnybrook Health Sciences Centre, Department of Radiation Oncology, Toronto, ON, M4N 3M5, Canada
| | - Ting Martin Ma
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Sarah P Psutka
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
- University of Washington, Department of Urology, Seattle, WA 98195, USA
| | - Ramesh Rengan
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Division of Radiation Oncology & Cancer Imaging, Melbourne, VIC, 3052, Australia
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, VIC, 3052, Australia
| | - Anand Swaminath
- Juravinski Cancer Centre, Radiation Therapy, Hamilton, ON, L8V 5C2, Canada
- McMaster University, Division of Radiation Oncology, Hamilton, ON,L8S 4L8,Canada
| | - Lisa Tachiki
- University of Washington, Division of Hematology & Oncology, Seattle, WA 98195,USA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
| | - Chad Tang
- The University of Texas MD Anderson Cancer Center, Genitourinary Radiation Oncology, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, Investigational Cancer Therapeutics, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, Translational Molecular Pathology, Houston, TX 77030, USA
| | - Bin Sing Teh
- Houston Methodist Hospital, Radiation Oncology, Houston, TX 77030, USA
| | - Joseph Tsai
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Scott S Tykodi
- University of Washington, Division of Hematology & Oncology, Seattle, WA 98195,USA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA 98195, USA
| | - Emily Weg
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| | - Nicholas G Zaorsky
- University Hospitals Seidman Cancer Center, Case Western Reserve University, Department of Radiation Oncology, Cleveland, OH 44106, USA
| | - Simon S Lo
- University of Washington, Department of Radiation Oncology, Seattle, WA 98195, USA
- Fred Hutchinson Cancer Center, Radiation Oncology Division, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Zhang M, Li J, Liu S, Zhou F, Zhang L. UCHL5 is a putative prognostic marker in renal cell carcinoma: a study of UCHL family. MOLECULAR BIOMEDICINE 2024; 5:28. [PMID: 39034372 PMCID: PMC11265068 DOI: 10.1186/s43556-024-00192-0] [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: 04/19/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
A macroscopic perspective is indispensable for understanding the intricate relationship between deubiquitinases and tumorigenesis. Proteomics has been proposed as a viable approach for elucidating the complex role of deubiquitylation in cellular progression. Instead of studying the function of a single ubiquitinase, research on a deubiquitinase family with similar catalytic core(s) may provide a new perspective for the pathological understanding of cancer. The Ubiquitin C-terminal hydrolase L (UCHL) family consists of four members: UCHL1, UCHL3, UCHL5, and BRAC1 associated protein-1 (BAP1), and they have been implicated in tumorigenesis and metastasis. Some members are considered hallmarks of intracranial lesions, colon cancer, chromatin remodeling, and histone stability. The present study uncovered an unknown correlation between the UCHL family and renal cancer. We discovered that UCHLs exhibit diverse regulatory effects in renal cancer, establishing connections between the renal cancer and truncated gene mutations, mitochondrial energetic metastasis, immune cell infiltration, and chromosomal stability of UCHLs family. Notably, we found that the increase of UCHL5 expression in renal cancer cells decreases the antigen processing and presentation of RCC tumor-infiltrating B cells. Further research identified that the expression of UCHL5 in RCC tumors is correlated with transport proteins, which led us to find that the abundance of UCHL5 in the blood of late-stage renal cell cancer patients is upregulated from 18 ng/L to 500 ng/L. Therefore, we propose that the abundance of UCHL5 in patients' blood can be a possible indicator of poor prognosis for renal cell cancer.
Collapse
Affiliation(s)
- Mengdi Zhang
- Life Sciences Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Jingxian Li
- Life Sciences Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Sijia Liu
- International Biomed-X Research Center, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, PR China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310058, PR China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, 215123, PR China
| | - Long Zhang
- Life Sciences Institute, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
9
|
Greco F, D’Andrea V, Buoso A, Cea L, Bernetti C, Beomonte Zobel B, Mallio CA. Advancements in Radiogenomics for Clear Cell Renal Cell Carcinoma: Understanding the Impact of BAP1 Mutation. J Clin Med 2024; 13:3960. [PMID: 38999524 PMCID: PMC11242378 DOI: 10.3390/jcm13133960] [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: 05/20/2024] [Revised: 06/23/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024] Open
Abstract
Recent advancements in understanding clear cell renal cell carcinoma (ccRCC) have underscored the critical role of the BAP1 gene in its pathogenesis and prognosis. While the von Hippel-Lindau (VHL) mutation has been extensively studied, emerging evidence suggests that mutations in BAP1 and other genes significantly impact patient outcomes. Radiogenomics with and without texture analysis based on CT imaging holds promise in predicting BAP1 mutation status and overall survival outcomes. However, prospective studies with larger cohorts and standardized imaging protocols are needed to validate these findings and translate them into clinical practice effectively, paving the way for personalized treatment strategies in ccRCC. This review aims to summarize the current knowledge on the role of BAP1 mutation in ccRCC pathogenesis and prognosis, as well as the potential of radiogenomics in predicting mutation status and clinical outcomes.
Collapse
Affiliation(s)
- Federico Greco
- Department of Radiology, Cittadella della Salute, Azienda Sanitaria Locale di Lecce, Piazza Filippo Bottazzi, 2, 73100 Lecce, Italy
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
| | - Valerio D’Andrea
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Andrea Buoso
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Laura Cea
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Caterina Bernetti
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Bruno Beomonte Zobel
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Carlo Augusto Mallio
- Research Unit of Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (V.D.); (A.B.); (L.C.); (C.B.); (B.B.Z.); (C.A.M.)
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| |
Collapse
|
10
|
Vlachostergios PJ, Papathanassiou M, Anagnostou M, Thodou E, Tamposis I, Mitrakas L, Zachos I, Koukoulis GK, Samara M, Tzortzis V. Mutational profile of primary clear cell renal cell carcinoma predicts recurrence and potential candidacy for adjuvant immune checkpoint inhibition. F1000Res 2024; 12:918. [PMID: 38933491 PMCID: PMC11200057 DOI: 10.12688/f1000research.136087.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The risk of recurrence after nephrectomy for primary clear cell renal cell carcinoma (ccRCC) is estimated in daily practice solely based on clinical criteria. The aim of this study was to assess the prognostic relevance of common somatic mutations with respect to tumor aggressiveness and outcomes of ccRCC patients after definitive treatment. METHODS Primary tumors from 37 patients with ccRCC who underwent radical nephrectomy were analyzed for presence of somatic mutations using a 15-gene targeted next-generation sequencing (NGS) panel. Associations to histopathologic characteristics and outcomes were investigated in the study cohort (n=37) and validated in The Cancer Genome Atlas (TCGA) ccRCC cohort (n=451). RESULTS VHL was the most frequently mutated gene (51%), followed by PBRM1 (27%), BAP1 (13%), SETD2 (13%), KDM5C (5%), ATM (5%), MTOR (5%), and PTEN (3%). One-third of patients did not have any somatic mutations within the 15-gene panel. The vast majority of tumors harboring no mutations at all or VHL-only mutations (51%) were more frequently of smaller size (pT1-2) and earlier stage (I/II), whereas presence of any other gene mutations in various combinations with or without VHL was enriched in larger (pT3) and higher stage tumors (III) (p=0.02). No recurrences were noted in patients with unmutated tumors or VHL-only mutations as opposed to three relapses in patients with non- VHL somatic mutations (p=0.06). Presence of somatic mutations in PBRM1, BAP1, SETD2, KDM5C, ATM, MTOR, or PTEN genes in 451 TCGA ccRCC patients was associated with a significantly shorter disease-free survival (DFS) compared to those with unaltered tumors (q=0.01). CONCLUSIONS Preliminary findings from this ongoing study support the prognostic value of non- VHL mutations including PBRM1, BAP1, SETD2, KDM5C, ATM, MTOR, and PTEN in primary ccRCC tumors as surrogates of earlier recurrence and potential selection for adjuvant immune checkpoint inhibition.
Collapse
Affiliation(s)
- Panagiotis J Vlachostergios
- Urology, University of Thessaly, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa, Larissa, Thessalia, Greece
- Medical Oncology, IASO Thessalias Hospital, Larissa, Thessalia, Greece
- Hematology & Medical Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Maria Papathanassiou
- Pathology, University of Thessaly, Faculty of Medicine, School of Health Sciences, Larissa, Greece
| | - Maria Anagnostou
- Pathology, University of Thessaly, Faculty of Medicine, School of Health Sciences, Larissa, Greece
| | - Eleni Thodou
- Pathology, University of Thessaly, Faculty of Medicine, School of Health Sciences, Larissa, Greece
| | - Ioannis Tamposis
- Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Lampros Mitrakas
- Urology, University of Thessaly, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa, Larissa, Thessalia, Greece
| | - Ioannis Zachos
- Urology, University of Thessaly, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa, Larissa, Thessalia, Greece
| | - George K Koukoulis
- Pathology, University of Thessaly, Faculty of Medicine, School of Health Sciences, Larissa, Greece
| | - Maria Samara
- Pathology, University of Thessaly, Faculty of Medicine, School of Health Sciences, Larissa, Greece
| | - Vassilios Tzortzis
- Urology, University of Thessaly, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa, Larissa, Thessalia, Greece
| |
Collapse
|
11
|
Eule CJ, Hu J, Hedges D, Jani A, Pshak T, Manley BJ, Sanchez A, Dreicer R, Myint ZW, Zakharia Y, Lam ET. Clinical and Genomic Features of Patients with Renal Cell Carcinoma and Advanced Chronic Kidney Disease: Analysis of a Multi-Institutional Database. Cancers (Basel) 2024; 16:1920. [PMID: 38791999 PMCID: PMC11119962 DOI: 10.3390/cancers16101920] [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: 04/22/2024] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Patients with advanced chronic kidney disease (ACKD) are at an increased risk of developing renal cell carcinoma (RCC), but molecular alterations in RCC specimens arising from ACKD and overall survival (OS) in affected patients are not well defined. PATIENTS AND METHODS Using the Oncology Research Information Exchange Network (ORIEN) Total Cancer Care® protocol, 296 consented adult patients with RCC and somatic tumor whole exome sequencing were included. Patients with ACKD were defined as those with serum creatinine ≥1.5 mg/dL prior to RCC diagnosis. RESULTS Of 296 patients with RCC, 61 met the criteria for ACKD. The most common somatic mutations in the overall cohort were in VHL (126, 42.6%), PBRM1 (102, 34.5%), and SETD2 (54, 18.2%). BAP1 had a decreased mutational frequency in RCC specimens from patients without ACKD as compared to those with ACKD (10.6% versus 1.6%), but this was not statistically significant in univariable (OR 0.14, p = 0.056) or multivariable (OR 0.15, p = 0.067) analysis. Median OS was not reached in either cohort. CONCLUSIONS Using the clinicogenomic ORIEN database, our study found lower rates of BAP1 mutations in RCC specimens from patients with ACKD, which may reflect a BAP1-independent mutational driver of RCC in patients with ACKD.
Collapse
Affiliation(s)
- Corbin J. Eule
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Junxiao Hu
- Biostatistics Core, University of Colorado Cancer Center, Aurora, CO 80045, USA
| | | | - Alkesh Jani
- Division of Nephrology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Thomas Pshak
- Division of Urology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brandon J. Manley
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Alejandro Sanchez
- Division of Urology, Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Robert Dreicer
- Division of Medical Oncology, Department of Medicine, University of Virginia Comprehensive Cancer Center, Charlottesville, VA 22908, USA
| | - Zin W. Myint
- Division of Medical Oncology, Department of Internal Medicine, Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
| | - Yousef Zakharia
- Division of Hematology, Oncology, and Blood and Bone Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Elaine T. Lam
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
12
|
Jia L, Cowell LG, Kapur P. Understanding Factors that Influence Prognosis and Response to Therapy in Clear Cell Renal Cell Carcinoma. Adv Anat Pathol 2024; 31:96-104. [PMID: 38179997 DOI: 10.1097/pap.0000000000000428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
In this review, we highlight and contextualize emerging morphologic prognostic and predictive factors in renal cell carcinoma. We focus on clear cell renal cell carcinoma (ccRCC), the most common histologic subtype. Our understanding of the molecular characterization of ccRCC has dramatically improved in the last decade. Herein, we highlight how these discoveries have laid the foundation for new approaches to prognosis and therapeutic decision-making for patients with ccRCC. We explore the clinical relevance of common mutations, established gene expression signatures, intratumoral heterogeneity, sarcomatoid/rhabdoid morphology and PD-L1 expression, and discuss their impact on predicting response to therapy.
Collapse
Affiliation(s)
| | - Lindsay G Cowell
- Peter O'Donnell School of Public Health
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| | - Payal Kapur
- Department of Pathology
- Department of Urology, University of Texas Southwestern Medical Center
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, Dallas, TX
| |
Collapse
|
13
|
Rosellini M, Mollica V, Marchetti A, Coluccelli S, Giunchi F, Tassinari E, Ricci C, Fiorentino M, Tallini G, De Biase D, Massari F. Chromosome 3p gene alterations as biomarkers for immunocombinations in metastatic renal cell carcinoma: A hypothesis-generating analysis. Pathol Res Pract 2024; 254:155142. [PMID: 38277752 DOI: 10.1016/j.prp.2024.155142] [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: 08/22/2023] [Revised: 12/31/2023] [Accepted: 01/13/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Identifying biomarkers for metastatic renal cell carcinoma (mRCC) is an unmet need in actual immunotherapy era. Available data regarding chromosome 3p genes (i.e., VHL, PBRM1, SETD2) mutations as potential predictors for therapy response is conflicting. We describe the impact of these mutations on clinical outcomes in mRCC patients treated with immune checkpoint inhibitor (ICI)-doublet or ICI/tyrosine kinase inhibitor (TKI) combinations. METHODS We performed a single-center retrospective analysis on mRCC patients treated with first line ICI/ICI or ICI/TKI. A multi-gene panel was used, allowing the amplification of 841 amplicons (54.93 kb, human reference sequence hg19/GRCh37) in the coding sequences of the following genes: ATM, BAP1, KDM5C, MET, MTOR, NF2, PBRM1, PIK3CA, PTEN, SETD2, SMARCB1, TP53, TSC1, TSC2, VHL. RESULTS 18 patients undergoing ICI/ICI and ICI/TKI who had tumor tissue adequate for molecular analysis were included. Histology was 100% clear cell. IMDC risk was 50% intermediate, 33.4% good, 16.6% poor. First line therapy was 89% ICI/TKI, 11% ICI/ICI. 83.3% pts (n = 15) carried genomic alterations (GA). Most common GA included VHL in 44% (n = 8; 7 pathogenic - PAT and 1 variant of unknown significance - VUS), PBRM1 in 44% (n = 8; 5 PAT and 3 VUS) and SETD2 in 33% (n = 6; 4 PAT and 2 VUS). With the limit of a small sample that did not allow proper statistical analyses, SETD2-mutated patients had lower median progression free (mPFS) and overall survival (mOS) than non-SETD2 mutated patients. Higher mPFS and mOS were shown with VHL or PBRM1 GA, especially in PBRM1 +VHL mutated pts. CONCLUSIONS Our data shows a possible negative predictive role of SETD2 GA for ICI-based therapy in RCC. Concomitant VHL and PBRM1 GA could act as a predictor for ICI/TKI efficacy. Our hypothesis-generating analysis highlights the need of an integrated evaluation of these genes as promising biomarkers in RCC. Further larger studies are required.
Collapse
Affiliation(s)
- Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Sara Coluccelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Giunchi
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Costantino Ricci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Pathology Unit, Maggiore Hospital-AUSL Bologna, Bologna, Italy
| | - Michelangelo Fiorentino
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Pathology Unit, Maggiore Hospital-AUSL Bologna, Bologna, Italy
| | - Giovanni Tallini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Dario De Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Pharmacy and Biotechnology (FaBit), University of Bologna, 40126 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
| |
Collapse
|
14
|
Jang A, Lichterman JN, Zhong JY, Shoag JE, Garcia JA, Zhang T, Barata PC. Immune approaches beyond traditional immune checkpoint inhibitors for advanced renal cell carcinoma. Hum Vaccin Immunother 2023; 19:2276629. [PMID: 37947202 PMCID: PMC10653627 DOI: 10.1080/21645515.2023.2276629] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Renal cell carcinoma (RCC), especially clear cell RCC, is generally considered an immunotherapy-responsive cancer. Recently, the prognosis for patients with locally advanced and metastatic RCC has significantly improved with the regulatory approvals of anti-PD-1/PD-L1/CTLA-4 immune checkpoint inhibitor (ICI)-based regimens. Yet in most cases, RCC will remain initially unresponsive to treatment or will develop resistance over time. Hence, there remains an unmet need to understand what leads to ICI resistance and to develop novel immune and nonimmune treatments to enhance the response to ICIs. In this review, we highlight recently published studies and the latest clinical studies investigating the next generation of immune approaches to locally advanced and metastatic RCC beyond traditional ICIs. These trials include cytokines, gut microbiota-based therapies, novel immune checkpoint agents, vaccines, and chimeric antigen receptor T cells. These agents are being evaluated as monotherapy or in combination with traditional ICIs and will hopefully provide improved outcomes to patients with RCC soon.
Collapse
Affiliation(s)
- Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jake N. Lichterman
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey Y. Zhong
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jonathan E. Shoag
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jorge A. Garcia
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Tian Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pedro C. Barata
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
15
|
Lee J, Thuzar M, DiPoto-Brahmbhatt AC, Chaudoir C, Tanner A, Fazili T, Yu X, Constantinescu M, Yeh YA. Clear cell renal cell carcinoma with BAP1 mutation: a report of two cases. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:429-434. [PMID: 37941648 PMCID: PMC10628621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/31/2023] [Indexed: 11/10/2023]
Abstract
Clear cell renal cell carcinoma is the most common subtype of renal cell carcinomas (RCCs) and accounts for 60%-70% of all RCCs cases in adults. Aberrations in the von Hippel-Lindau (VHL) gene on chromosome 3p occurred in > 90% of clear cell RCCs. Other tumor suppressor genes located on chromosome 3p, such as BAP1, PBRM1, and SETD2, also contribute to tumorigenesis. Clear cell RCCs with both BAP1 and VHL mutations may display distinctive histopathological features. Here, we report two cases of clear cell RCCs with BAP1 mutation. One tumor had VHL, BAP-1, and RAF1 mutations and the tumor nests and alveoli of tumor cells were surrounded by proliferative vessels and the optically clear cytoplasm contained numerous eosinophilic granules and hyaline globules of varying sizes. The other tumor had BAP1 and ATM mutations, and demonstrated clear cells with numerous eosinophilic granules and other typical histopathological features of conventional clear cell RCC. Furthermore, many tumor nodules with dense peripheral lymphocytic infiltrates contained rhabdoid cells. Sarcomatoid cells were also observed. Both tumor cells showed high-grade nuclei. Clear cell RCCs with BAP1 mutation exhibit aggressive clinical behaviors.
Collapse
Affiliation(s)
- Jennifer Lee
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Moe Thuzar
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Angela C DiPoto-Brahmbhatt
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Catherine Chaudoir
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Anthony Tanner
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
| | - Tajammul Fazili
- Department of Urology, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Urology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Xiuping Yu
- Department of Urology, Louisiana State University Health ShreveportShreveport, LA, USA
- Department of Biochemistry and Molecular Biology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Michael Constantinescu
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health ShreveportShreveport, LA, USA
| | - Y Albert Yeh
- Pathology and Laboratory Medicine Service, Overton Brooks VA Medical CenterShreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health ShreveportShreveport, LA, USA
- Department of Urology, Louisiana State University Health ShreveportShreveport, LA, USA
| |
Collapse
|
16
|
Pini GM, Lucianò R, Colecchia M. Cystic Clear Cell Renal Cell Carcinoma: A Morphological and Molecular Reappraisal. Cancers (Basel) 2023; 15:3352. [PMID: 37444462 DOI: 10.3390/cancers15133352] [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: 05/31/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
A wide variety of renal neoplasms can have cystic areas. These can occur for different reasons: some tumors have an intrinsic cystic architecture, while others exhibit pseudocystic degeneration of necrotic foci or they have cystically dilated renal tubules constrained by stromal neoplastic cells. Clear cell renal cell carcinoma (CCRCC), either solid or cystic, is the most frequent type of renal cancer. While pseudocysts are found in high-grade aggressive CCRCC, cystic growth is associated with low-grade indolent cases. The latter also form through a cyst-dependent molecular pathway, and they are more frequent in patients suffering from VHL disease. The differential diagnosis of multilocular cystic renal neoplasm of low malignant potential and clear cell papillary renal cell tumor can be especially hard and requires a focused macroscopical and microscopical pathological analysis. As every class of renal tumor includes cystic forms, knowledge of the criteria required for a differential diagnosis is mandatory.
Collapse
Affiliation(s)
- Giacomo Maria Pini
- Department of Pathology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Roberta Lucianò
- Department of Pathology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Maurizio Colecchia
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
17
|
Gupta S, Cheng L, Erickson LA. Contemporary updates in urologic pathology: a special issue of renal, urinary tract, prostate, penile, and testicular pathology. Hum Pathol 2023; 133:1-4. [PMID: 36265596 DOI: 10.1016/j.humpath.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/04/2022]
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Lifespan Academic Medical Center, Providence, RI, 02903, USA.
| | - Lori A Erickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
BAP1 and PTEN mutations shape the immunological landscape of clear cell renal cell carcinoma and reveal the intertumoral heterogeneity of T cell suppression: a proof-of-concept study. Cancer Immunol Immunother 2022; 72:1603-1618. [PMID: 36562826 DOI: 10.1007/s00262-022-03346-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is an immunologically vulnerable tumor entity, and immune checkpoint inhibitors are now widely used to treat patients with advanced disease. Whether and to what extent immune responses in ccRCC are shaped by genetic alterations, however, is only beginning to emerge. In this proof-of-concept study, we performed a detailed correlative analysis of the mutational and immunological landscapes in a series of 23 consecutive kidney cancer patients. We discovered that a high infiltration with CD8 + T cells was not dependent on the number of driver mutations but rather on the presence of specific mutational events, namely pathogenic mutations in PTEN or BAP1. This observation encouraged us to compare mechanisms of T cell suppression in the context of four different genetic patterns, i.e., the presence of multiple drivers, a PTEN or BAP1 mutation, or the absence of detectable driver mutations. We found that ccRCCs harboring a PTEN or BAP1 mutation showed the lowest level of Granzyme B positive tumor-infiltrating lymphocytes (TILs). A multiplex immunofluorescence analysis revealed a significant number of CD8 + TILs in the vicinity of CD68 + macrophages/monocytes in the context of a BAP1 mutation but not in the context of a PTEN mutation. In line with this finding, direct interactions between CD8 + TILs and CD163 + M2-polarized macrophages were found in BAP1-mutated ccRCC but not in tumors with other mutational patterns. While an absence of driver mutations was associated with more CD8 + TILs in the vicinity of FOXP3 + Tregs and CD68 + monocytes/macrophages, the presence of multiple driver mutations was, to our surprise, not found to be strongly associated with immunosuppressive mechanisms. Our results highlight the role of genetic alterations in shaping the immunological landscape of ccRCC. We discovered a remarkable heterogeneity of mechanisms that can lead to T cell suppression, which supports the need for personalized immune oncological approaches.
Collapse
|