1
|
Crocetto F, Falcone A, Mirto BF, Sicignano E, Pagano G, Dinacci F, Varriale D, Machiella F, Giampaglia G, Calogero A, Varlese F, Balsamo R, Trama F, Sciarra A, Del Giudice F, Busetto GM, Ferro M, Lucarelli G, Lasorsa F, Imbimbo C, Barone B. Unlocking Precision Medicine: Liquid Biopsy Advancements in Renal Cancer Detection and Monitoring. Int J Mol Sci 2024; 25:3867. [PMID: 38612677 PMCID: PMC11011885 DOI: 10.3390/ijms25073867] [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: 03/03/2024] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Renal cell carcinoma (RCC) remains a formidable diagnostic challenge, especially in the context of small renal masses. The quest for non-invasive screening tools and biomarkers has steered research towards liquid biopsy, focusing on microRNAs (miRNAs), exosomes, and circulating tumor cells (CTCs). MiRNAs, small non-coding RNAs, exhibit notable dysregulation in RCC, offering promising avenues for diagnosis and prognosis. Studies underscore their potential across various biofluids, including plasma, serum, and urine, for RCC detection and subtype characterization. Encouraging miRNA signatures show correlations with overall survival, indicative of their future relevance in RCC management. Exosomes, with their diverse molecular cargo, including miRNAs, emerge as enticing biomarkers, while CTCs, emanating from primary tumors into the bloodstream, provide valuable insights into cancer progression. Despite these advancements, clinical translation necessitates further validation and standardization, encompassing larger-scale studies and robust evidence generation. Currently lacking approved diagnostic assays for renal cancer, the potential future applications of liquid biopsy in follow-up care, treatment selection, and outcome prediction in RCC patients are profound. This review aims to discuss and highlight recent advancements in liquid biopsy for RCC, exploring their strengths and weaknesses in the comprehensive management of this disease.
Collapse
Affiliation(s)
- Felice Crocetto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Alfonso Falcone
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Benito Fabio Mirto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Enrico Sicignano
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Giovanni Pagano
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Fabrizio Dinacci
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Domenico Varriale
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Fabio Machiella
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Gaetano Giampaglia
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Armando Calogero
- Department of Advanced Biomedical Science, University of Naples “Federico II”, 80131 Naples, Italy; (A.C.); (F.V.)
| | - Filippo Varlese
- Department of Advanced Biomedical Science, University of Naples “Federico II”, 80131 Naples, Italy; (A.C.); (F.V.)
| | | | - Francesco Trama
- ASL Napoli 2 Nord, P.O. Santa Maria delle Grazie, 80078 Pozzuoli, Italy;
| | - Antonella Sciarra
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Francesco Del Giudice
- Department of Maternal Infant and Urological Sciences, Umberto I Polyclinic Hospital, Sapienza University, 00161 Rome, Italy;
| | - Gian Maria Busetto
- Department of Urology and Renal Transplantation, University of Foggia, 71122 Foggia, Italy;
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology (IEO)-IRCCS, 20141 Milan, Italy;
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy; (G.L.); (F.L.)
| | - Francesco Lasorsa
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy; (G.L.); (F.L.)
| | - Ciro Imbimbo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Biagio Barone
- Urology Unit, Department of Surgical Sciences, AORN Sant’Anna e San Sebastiano, 81100 Caserta, Italy
| |
Collapse
|
2
|
Zieren RC, Zondervan PJ, Pienta KJ, Bex A, de Reijke TM, Bins AD. Diagnostic liquid biopsy biomarkers in renal cell cancer. Nat Rev Urol 2024; 21:133-157. [PMID: 37758847 DOI: 10.1038/s41585-023-00818-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
The clinical presentation of renal cell cancer (RCC) is shifting towards incidental and early detection, creating new challenges in RCC diagnosis. Overtreatment might be reduced with the development of new diagnostic biomarkers to distinguish benign from malignant small renal masses (SRMs). Differently from tissue biopsies, liquid biopsies are obtained from a patient's blood or urine and, therefore, are minimally invasive and suitable for longitudinal monitoring. The most promising types of liquid biopsy biomarkers for RCC diagnosis are circulating tumour cells, extracellular vesicles (EVs) and cell-free DNA. Circulating tumour cell assays have the highest specificity, with low processing time and costs. However, the biological characteristics and low sensitivity limit the use of these markers in SRM diagnostics. Cell-free DNA might complement the diagnosis of high-volume RCC, but the potential for clinical application in SRMs is limited. EVs have the highest biological abundance and the highest sensitivity in identifying low-volume disease; moreover, the molecular characteristics of these markers make EVs suitable for multiple analytical applications. Thus, currently, EV assays have the greatest potential for diagnostic application in RCC (including identification of SRMs). All these liquid biomarkers have potential in clinical practice, pending validation studies. Biomarker implementation will be needed to also improve characterization of RCC subtypes. Last, diagnostic biomarkers might be extended to prognostic or predictive applications.
Collapse
Affiliation(s)
- Richard C Zieren
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Patricia J Zondervan
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, United Kingdom
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Theo M de Reijke
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan D Bins
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Circulating Cell-Free DNA in Renal Cell Carcinoma: The New Era of Precision Medicine. Cancers (Basel) 2022; 14:cancers14184359. [PMID: 36139519 PMCID: PMC9497114 DOI: 10.3390/cancers14184359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Early diagnosis of renal cell carcinoma (RCC) is challenging and typically incidental. Currently, several therapeutic strategies are used for the treatment; however, no established predictive biomarker has been established yet, and the optimal treatment choice and sequence of use remain unclear. Moreover, the recurrence occurs in about one-third of patients after tumor resection. Although several prognostic classification systems have been proposed, most of them showed only limited potential in recurrence prediction. Therefore, identifying simple, reliable, and easily accessible biomarkers to anticipate the diagnosis, effectively evaluate the risk of relapse, and predict the response to the therapeutic regimens is an unmet clinical need. Circulating cell-free DNA (cfDNA), released from cancer cells into the bloodstream, was shown to be a non-invasive, viable, inexpensive method to diagnose and monitor several solid malignancies, designed as a potential blood RCC biomarker. This review aims to summarize the state of the art of the current genetic and epigenetic techniques of plasma and serum cfDNA detection and outline the potential application of liquid biopsy in RCC. Abstract Tumor biopsy is still the gold standard for diagnosing and prognosis renal cell carcinoma (RCC). However, its invasiveness, costs, and inability to accurately picture tumor heterogeneity represent major limitations to this procedure. Analysis of circulating cell-free DNA (cfDNA) is a non-invasive cost-effective technique that has the potential to ease cancer detection and prognosis. In particular, a growing body of evidence suggests that cfDNA could be a complementary tool to identify and prognosticate RCC while providing contemporary mutational profiling of the tumor. Further, recent research highlighted the role of cfDNA methylation profiling as a novel method for cancer detection and tissue-origin identification. This review synthesizes current knowledge on the diagnostic, prognostic, and predictive applications of cfDNA in RCC, with a specific focus on the potential role of cell-free methylated DNA (cfMeDNA).
Collapse
|
4
|
Circulating Tumor DNA in Patients with Renal Cell Carcinoma. A Systematic Review of the Literature. EUR UROL SUPPL 2022; 37:27-35. [PMID: 35106503 PMCID: PMC8784339 DOI: 10.1016/j.euros.2021.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 11/24/2022] Open
Abstract
Context Over the past decade there has been increasing interest in the potential of liquid biopsies and systematic biomarkers in the diagnosis and management of kidney cancer, as they may provide a tool for early detection of disease and monitoring of treatment response. Objective To identify and summarize relevant published data on circulating tumor DNA (ctDNA) in patients with renal cell carcinoma (RCC). Evidence acquisition We performed a systematic review according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement of studies identified in PubMed, MEDLINE, EMBASE, and Cochrane Library up to January 15, 2021. Two reviewers independently screened all articles and performed the data extraction. Evidence synthesis Nineteen studies investigating ctDNA in RCC (1237 patients) were included and analyzed in the final review. The study size and design varied widely, and the studies were divided into five groups according to the method used for ctDNA detection. The outcome data included (1) the sensitivity/specificity if available; (2) the method used for ctDNA detection; and (3) the main findings in the studies. Conclusions The studies highlight that the level of ctDNA in RCC appears to be low. Studies using multiple methods for ctDNA detection indicate that tumor-guided analysis improves the ctDNA detection rate and suggest that cell-free methylated DNA immunoprecipitation and high-throughput sequencing may be a very sensitive method for ctDNA detection in RCC. Patient summary We systematically reviewed the literature to identify all relevant studies investigating circulating tumor DNA in patients with kidney cancer to investigate its use and potential in this highly malignant disease. We found that the level of circulating tumor DNA is low in kidney cancer and that very sensitive methods have to be used for detection in this disease.
Collapse
|
5
|
Ferguson SD, Fomchenko EI, Guerrieri RA, Glitza Oliva IC. Challenges and Advances in Diagnosis and Treatment of Leptomeningeal Disease (LMD). Front Oncol 2022; 11:800053. [PMID: 35096602 PMCID: PMC8789647 DOI: 10.3389/fonc.2021.800053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Leptomeningeal disease (LMD) is a devastating category of CNS metastasis with a very poor prognosis and limited treatment options. With maximal aggressive therapy, survival times remain short and, without treatment, prognosis is measured in weeks. Both LMD diagnosis and treatment are challenging topics within neuro-oncology. In this review, we discuss the advances in LMD diagnosis with a focus on the role of circulating tumor DNA (ctDNA) and discuss the role of targeted and immunotherapy in LMD treatment.
Collapse
Affiliation(s)
- Sherise D Ferguson
- Department of Neurosurgery, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Elena I Fomchenko
- Department of Neurosurgery, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Renato A Guerrieri
- Department of Melanoma Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
6
|
Renner AM, Derichsweiler C, Ilyas S, Gessner I, Fries JWU, Mathur S. High efficiency capture of biomarker miRNA15a for noninvasive diagnosis of malignant kidney tumors. Biomater Sci 2022; 10:1113-1122. [PMID: 35048092 DOI: 10.1039/d1bm01737c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To date, there are no preoperative and quantitative dynamics in clinical practice that can reliably differentiate between a benign and malignant renal cell carcinoma (RCC). For monitoring different analytes in body fluids, more than 40 different molecular biomarkers have been identified, however, they are associated with limited clinical sensitivity and/or non-optimal specificity due to their leaky nature. Previous work on RCC demonstrated the miRNA15a to be reliable and novel biomarker with 98.1% specificity and 100% sensitivity. Despite the high potential of miRNA15a biomarker, its clinical application is considerably hampered by the insensitive nature of the detection methods and low concentration of biomarker in samples that is aggravated by the high level of contamination due to other solutes present in body fluids. In this work, a non-invasive quantitative approach is demonstrated to overcome such diagnostics issues through biotin-streptavidin binding and fluorescence active magnetic nanocarriers that ensured prompt isolation, enrichment and purification of the biomarker miRNA15a from urine. The study demonstrates that detectable low levels of these miRNAs through miRNA capturing nanocarriers can potentially function as advanced diagnostic markers for the non-invasive investigation and early detection of renal cancer.
Collapse
Affiliation(s)
- Alexander M Renner
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Christina Derichsweiler
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Shaista Ilyas
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Isabel Gessner
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Jochen W U Fries
- Institute of Urology/Pathology, University Hospital of Cologne, Kerpenerstr. 62, 50924 Cologne, Germany
| | - Sanjay Mathur
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| |
Collapse
|
7
|
The Role of Circulating Biomarkers in the Oncological Management of Metastatic Renal Cell Carcinoma: Where Do We Stand Now? Biomedicines 2021; 10:biomedicines10010090. [PMID: 35052770 PMCID: PMC8773056 DOI: 10.3390/biomedicines10010090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/25/2021] [Accepted: 12/29/2021] [Indexed: 01/08/2023] Open
Abstract
Renal cell carcinoma (RCC) is an increasingly common malignancy that can progress to metastatic renal cell carcinoma (mRCC) in approximately one-third of RCC patients. The 5-year survival rate for mRCC is abysmally low, and, at the present time, there are sparingly few if any effective treatments. Current surgical and pharmacological treatments can have a long-lasting impact on renal function, as well. Thus, there is a compelling unmet need to discover novel biomarkers and surveillance methods to improve patient outcomes with more targeted therapies earlier in the course of the disease. Circulating biomarkers, such as circulating tumor DNA, noncoding RNA, proteins, extracellular vesicles, or cancer cells themselves potentially represent a minimally invasive tool to fill this gap and accelerate both diagnosis and treatment. Here, we discuss the clinical relevance of different circulating biomarkers in metastatic renal cell carcinoma by clarifying their potential role as novel biomarkers of response or resistance to treatments but also by guiding clinicians in novel therapeutic approaches.
Collapse
|
8
|
Lakshminarayanan H, Rutishauser D, Schraml P, Moch H, Bolck HA. Liquid Biopsies in Renal Cell Carcinoma-Recent Advances and Promising New Technologies for the Early Detection of Metastatic Disease. Front Oncol 2020; 10:582843. [PMID: 33194717 PMCID: PMC7656014 DOI: 10.3389/fonc.2020.582843] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) displays a highly varying clinical progression, from slow growing localized tumors to very aggressive metastatic disease (mRCC). Almost a third of all patients with ccRCC show metastatic dissemination at presentation while another third develop metastasis during the course of the disease. Survival rates of mRCC patients remain low despite the development of novel targeted treatment regimens. Biomarkers indicating disease progression could help to define its aggressive potential and thus guide patient management. However, molecular markers that can reliably assess metastatic dissemination and disease recurrence in ccRCC have not been recommended for clinical practice to date. Liquid biopsies could provide an attractive and non-invasive method to determine the risk of recurrence or metastatic dissemination during follow-up and thus assist the search for surveillance biomarkers in ccRCC tumors. A wide spectrum of circulating molecules have already shown considerable potential for ccRCC diagnosis and prognostication. In this review, we outline state of the art of the key circulating analytes such as cfDNA, cfRNA, proteins, and exosomes that may serve as biomarkers for the longitudinal monitoring of ccRCC progression to metastasis. Moreover, we address some of the prevailing limitations in the past approaches and present promising adoptable technologies that could help to pursue the implementation of liquid biopsies as a prognostic tool for mRCC.
Collapse
Affiliation(s)
| | | | | | - Holger Moch
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Hella A. Bolck
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Gläsker S, Vergauwen E, Koch CA, Kutikov A, Vortmeyer AO. Von Hippel-Lindau Disease: Current Challenges and Future Prospects. Onco Targets Ther 2020; 13:5669-5690. [PMID: 32606780 PMCID: PMC7305855 DOI: 10.2147/ott.s190753] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022] Open
Abstract
Understanding of molecular mechanisms of tumor growth has an increasing impact on the development of diagnostics and targeted therapy of human neoplasia. In this review, we summarize the current knowledge on molecular mechanisms and their clinical implications in von Hippel-Lindau (VHL) disease. This autosomal dominant tumor syndrome usually manifests in young adulthood and predisposes affected patients to the development of benign and malignant tumors of different organ systems mainly including the nervous system and internal organs. A consequent screening and timely preventive treatment of lesions are crucial for patients affected by VHL disease. Surgical indications and treatment have been evaluated and optimized over many years. In the last decade, pharmacological therapies have been evolving, but are largely still at an experimental stage. Effective pharmacological therapy as well as detection of biomarkers is based on the understanding of the molecular basis of disease. The molecular basis of von Hippel-Lindau disease is the loss of function of the VHL protein and subsequent accumulation of hypoxia-inducible factor with downstream effects on cellular metabolism and differentiation. Organs affected by VHL disease may develop frank tumors. More characteristically, however, they reveal multiple separate microscopic foci of neoplastic cell proliferation. The exact mechanisms of tumorigenesis in VHL disease are, however, still not entirely understood and knowledge on biomarkers and targeted therapy is scarce.
Collapse
Affiliation(s)
- Sven Gläsker
- Neurosurgical Practise Lake Constance, Singen (Hohentwiel), Germany.,Department of Neurosurgery, VUB University Medical Center Brussels, Brussels, Belgium
| | - Evelynn Vergauwen
- Department of Neurosurgery, VUB University Medical Center Brussels, Brussels, Belgium.,Department of Neurology, University Hospital Antwerp, Antwerp, Belgium
| | | | | | - Alexander O Vortmeyer
- Department of Pathology, Indiana University-Purdue University, Indianapolis, IN, USA
| |
Collapse
|
10
|
Smith CG, Moser T, Mouliere F, Field-Rayner J, Eldridge M, Riediger AL, Chandrananda D, Heider K, Wan JCM, Warren AY, Morris J, Hudecova I, Cooper WN, Mitchell TJ, Gale D, Ruiz-Valdepenas A, Klatte T, Ursprung S, Sala E, Riddick ACP, Aho TF, Armitage JN, Perakis S, Pichler M, Seles M, Wcislo G, Welsh SJ, Matakidou A, Eisen T, Massie CE, Rosenfeld N, Heitzer E, Stewart GD. Comprehensive characterization of cell-free tumor DNA in plasma and urine of patients with renal tumors. Genome Med 2020; 12:23. [PMID: 32111235 PMCID: PMC7048087 DOI: 10.1186/s13073-020-00723-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cell-free tumor-derived DNA (ctDNA) allows non-invasive monitoring of cancers, but its utility in renal cell cancer (RCC) has not been established. METHODS Here, a combination of untargeted and targeted sequencing methods, applied to two independent cohorts of patients (n = 91) with various renal tumor subtypes, were used to determine ctDNA content in plasma and urine. RESULTS Our data revealed lower plasma ctDNA levels in RCC relative to other cancers of similar size and stage, with untargeted detection in 27.5% of patients from both cohorts. A sensitive personalized approach, applied to plasma and urine from select patients (n = 22) improved detection to ~ 50%, including in patients with early-stage disease and even benign lesions. Detection in plasma, but not urine, was more frequent amongst patients with larger tumors and in those patients with venous tumor thrombus. With data from one extensively characterized patient, we observed that plasma and, for the first time, urine ctDNA may better represent tumor heterogeneity than a single tissue biopsy. Furthermore, in a subset of patients (n = 16), longitudinal sampling revealed that ctDNA can track disease course and may pre-empt radiological identification of minimal residual disease or disease progression on systemic therapy. Additional datasets will be required to validate these findings. CONCLUSIONS These data highlight RCC as a ctDNA-low malignancy. The biological reasons for this are yet to be determined. Nonetheless, our findings indicate potential clinical utility in the management of patients with renal tumors, provided improvement in isolation and detection approaches.
Collapse
Affiliation(s)
- Christopher G Smith
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
| | - Tina Moser
- Medical University of Graz, Diagnostic and Research Center for Molecular Biomedicine, Institute of Human Genetics, Graz, Austria
| | - Florent Mouliere
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Johanna Field-Rayner
- Cambridge Urology Translational Research and Clinical Trials Office, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Matthew Eldridge
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Anja L Riediger
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Dineika Chandrananda
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Katrin Heider
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Jonathan C M Wan
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Anne Y Warren
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - James Morris
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Irena Hudecova
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Wendy N Cooper
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Thomas J Mitchell
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Davina Gale
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Andrea Ruiz-Valdepenas
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Tobias Klatte
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK
- Department of Urology, Royal Bournemouth Hospital, Bournemouth, UK
| | - Stephan Ursprung
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Evis Sala
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Antony C P Riddick
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Tevita F Aho
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - James N Armitage
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Samantha Perakis
- Medical University of Graz, Diagnostic and Research Center for Molecular Biomedicine, Institute of Human Genetics, Graz, Austria
| | - Martin Pichler
- Department of Internal Medicine Graz, Austria Division of Oncology, Medical University of Graz, Graz, Austria
| | - Maximilian Seles
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Gabriel Wcislo
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Sarah J Welsh
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Athena Matakidou
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Tim Eisen
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, UK
- Department of Oncology, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Charles E Massie
- Hutchison/MRC Research Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Ellen Heitzer
- Medical University of Graz, Diagnostic and Research Center for Molecular Biomedicine, Institute of Human Genetics, Graz, Austria.
- Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Graz, Austria.
| | - Grant D Stewart
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK.
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
11
|
Zhao Y, He JY, Zou YL, Guo XS, Cui JZ, Guo L, Bu H. Evaluating the cerebrospinal fluid ctDNA detection by next-generation sequencing in the diagnosis of meningeal Carcinomatosis. BMC Neurol 2019; 19:331. [PMID: 31856745 PMCID: PMC6924020 DOI: 10.1186/s12883-019-1554-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background Meningeal carcinomatosis (MC) is the most severe form of brain metastasis and causes significant morbidity and mortality. Currently, the diagnosis of MC is routinely confirmed on the basis of clinical manifestation, positive cerebrospinal fluid (CSF) cytology, and/or neuroimaging features. However, negative rate of CSF cytology and neuroimaging findings often result in a failure to diagnose MC from the patients who actually have the disease. Here we evaluate the CSF circulating tumor DNA (ctDNA) in the diagnosis of MC. Methods A total of 35 CSF samples were collected from 35 patients with MC for CSF cytology examination, CSF ctDNA extraction and cancer-associated gene mutations detection by next-generation sequencing (NGS) at the same time. Results The most frequent primary tumor in this study was lung cancer (26/35, 74%), followed by gastric cancer (2/35, 6%), breast cancer (2/35, 6%), prostatic cancer (1/35, 3%), parotid gland carcinoma (1/35, 3%) and lymphoma (1/35, 3%) while no primary tumor could be found in the remaining 2 patients in spite of using various inspection methods. Twenty-five CSF samples (25/35; 71%) were found neoplastic cells in CSF cytology examination while all of the 35 CSF samples (35/35; 100%) were revealed having detectable ctDNA in which cancer-associated gene mutations were detected. All of 35 patients with MC in the study underwent contrast-enhanced brain MRI and/or CT and 22 neuroimaging features (22/35; 63%) were consistent with MC. The sensitivity of the neuroimaging was 88% (95% confidence intervals [95% CI], 75 to 100) (p = 22/25) and 63% (95% CI, 47 to 79) (p = 22/35) compared to those of CSF cytology and CSF ctDNA, respectively. The sensitivity of the CSF cytology was 71% (95% CI, 56 to 86) (n = 25/35) compared to that of CSF ctDNA. Conclusions This study suggests a higher sensitivity of CSF ctDNA than those of CSF cytology and neuroimaging findings. We find cancer-associated gene mutations in ctDNA from CSF of patients with MC at 100% of our cohort, and utilizing CSF ctDNA as liquid biopsy technology based on the detection of cancer-associated gene mutations may give additional information to diagnose MC with negative CSF cytology and/or negative neuroimaging findings.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Jun-Ying He
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Yue-Li Zou
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Xiao-Su Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Jun-Zhao Cui
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Li Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China.
| | - Hui Bu
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| |
Collapse
|
12
|
Cimadamore A, Massari F, Santoni M, Mollica V, Di Nunno V, Cheng L, Lopez-Beltran A, Scarpelli M, Montironi R, Moch H. Molecular characterization and diagnostic criteria of renal cell carcinoma with emphasis on liquid biopsies. Expert Rev Mol Diagn 2019; 20:141-150. [PMID: 31498685 DOI: 10.1080/14737159.2019.1665510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Over the past 6 years, important genomic and transcriptomic studies performed on RCC reported a comprehensive molecular description of RCC pathogenic alterations. Such molecular findings pave the way for an integrated classification, based on histopathology aspects and molecular alterations in order to personalize the clinical management of RCC.Areas covered: The aim of this review is to evaluate the current knowledge and the potential value of liquid biopsy in RCC. Studies on presence and analysis of circulating tumor DNA (ctDNA), circulating RNA, specific microRNA, long non-coding RNA, and circulating tumor cells are reported for each phase of disease, from the diagnostic setting to the localized disease and, lastly, in the metastatic stage.Expert opinion: Advantages of liquid biopsies compared to serial tissue sampling are numerous. However, some limitations must be addressed before considering liquid biopsy as a noninvasive biomarker of clinical utility. The suboptimal sensitivity depends on the assessment technique and genetic platforms used, the tumor organ, the tumor stage, tumor heterogeneity, and clonality. The rate of discordance with tumor tissue genotyping may depends on temporal heterogeneity, spatial heterogeneity, and/or assay error (false-negative or false-positive genotyping).
Collapse
Affiliation(s)
- Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | | | - Veronica Mollica
- Division of Oncology, S. Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW With this review, we describe the most recent advances in active surveillance as well as diagnosis and management of small renal masses (SRMs). RECENT FINDINGS We discuss diagnosis, differentiation of solid from cystic lesions, risk prediction and treatment of the SRM. A better understanding of the disease facilitates the use of more conservatory treatments, such as active surveillance. Active surveillance has been increasingly accepted not only for SRM, but also for larger tumors and even metastatic patients. Exiting advances in risk prediction will help us define which patients can be safely managed with active surveillance and which require immediate treatment. Meanwhile, the use of renal tumor biopsies is still an important tool for these cases. SUMMARY Active surveillance is an option for many patients with renal masses. Noninvasive methods for diagnosis and risk prediction are being developed, but meanwhile, renal tumor biopsy is a useful tool. A better understanding of the disease increases the number of patients who can undergo active surveillance fully certain of the safety of their management.
Collapse
|
14
|
The evolving role of cytoreductive nephrectomy: incorporating genomics of metastatic renal cell carcinoma into treatment decisions. Curr Opin Urol 2019; 29:531-539. [PMID: 31313716 DOI: 10.1097/mou.0000000000000663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Recent publications evaluating cytoreductive nephrectomy in the era of targeted therapy emphasize the importance of patient selection. We reviewed the predictive role of genetic alterations in patients with metastatic renal cell carcinoma (mRCC) undergoing cytoreductive nephrectomy. RECENT FINDINGS Studies evaluating the association between genetic alterations and outcomes following systemic treatment for mRCC include mainly patients after cytoreductive nephrectomy. Expression of proangiogenic genes, single nucleotide polymorphisms involving genes of the vascular-endothelial growth factor (VEGF) pathway and somatic mutations of chromatin remodeling genes were associated with response to VEGF-targeted therapy. Outcomes following treatment with mammalian target of rapamycin (mTOR) inhibitors were initially associated with mTOR/TSC1/TSC2 mutations; however, subsequent studies did not validate these findings but rather found an association between loss of PTEN expression and PBRM1 mutations and improved outcomes. Loss of PBRM1 was initially linked to response to immunotherapy; however, larger studies question this association and showed high expression of T-effector gene signature predicted improved outcome. Primary tumors with low intratumor heterogeneity but elevated somatic copy-number alterations were associated with rapid progression at multiple sites. SUMMARY Genetic alterations may help select patients for cytoreductive nephrectomy and optimize timing of treatment. Intratumor heterogeneity and genetic discordance between primary and metastatic tumors may limit clinical applicability. Future studies should evaluate approaches to overcome these limitations.
Collapse
|
15
|
Bolck HA, Corrò C, Kahraman A, von Teichman A, Toussaint NC, Kuipers J, Chiovaro F, Koelzer VH, Pauli C, Moritz W, Bode PK, Rechsteiner M, Beerenwinkel N, Schraml P, Moch H. Tracing Clonal Dynamics Reveals that Two- and Three-dimensional Patient-derived Cell Models Capture Tumor Heterogeneity of Clear Cell Renal Cell Carcinoma. Eur Urol Focus 2019; 7:152-162. [PMID: 31266731 DOI: 10.1016/j.euf.2019.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/16/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Extensive DNA sequencing has led to an unprecedented view of the diversity of individual genomes and their evolution among patients with clear cell renal cell carcinoma (ccRCC). OBJECTIVE To understand subclonal architecture and dynamics of patient-derived two-dimensional (2D) and three-dimensional (3D) ccRCC models in vitro, in order to determine whether they mirror ccRCC inter- and intratumor heterogeneity. DESIGN, SETTING, AND PARTICIPANTS We have established a comprehensive platform of living renal cancer cell models from ccRCC surgical specimens. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS We confirmed the concordance of 2D and 3D patient-derived cell (PDC) models with the original tumor tissue in terms of histology, biomarker expression, cancer driver mutations, and copy number alterations. We addressed inter- and intrapatient heterogeneity by analyzing clonal dynamics during serial passaging. RESULTS AND LIMITATIONS In-depth genetic characterization verified the presence of heterogeneous cell populations, and revealed a high degree of similarity between subclonal compositions of monolayer and organoid cell cultures and the corresponding parental ccRCCs. Clonal dynamics were evident during serial passaging of cells in vitro, suggesting that PDC cultures can offer insights into evolutionary potential and treatment susceptibility of ccRCC subclones in vivo. Proof-of-concept drug profiling using selected ccRCC-targeted therapy agents highlighted patient-specific vulnerabilities in PDC models that could not be anticipated by interrogating commercially available cell lines. CONCLUSIONS We demonstrate that PDC models mirror inter- and intratumor heterogeneity of ccRCC in vitro. Based on our findings, we envision that the use of these models will advance our understanding of the trajectories that cause genetic diversity and their consequences for treatment on an individual level. PATIENT SUMMARY In this study, we developed two- and three-dimensional patient-derived models from clear cell renal cell carcinoma (ccRCC) as "mini-tumors in a dish." We show that these cell models retain important features of the human ccRCCs such as the profound tumor heterogeneity, thus highlighting their importance for cancer research and precision medicine.
Collapse
Affiliation(s)
- Hella A Bolck
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Claudia Corrò
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Abdullah Kahraman
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Adriana von Teichman
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Jack Kuipers
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland; Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | - Peter K Bode
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Markus Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland; Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Peter Schraml
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland.
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Sigolaeva LV, Bulko TV, Kozin MS, Zhang W, Köhler M, Romanenko I, Yuan J, Schacher FH, Pergushov DV, Shumyantseva VV. Long-term stable poly(ionic liquid)/MWCNTs inks enable enhanced surface modification for electrooxidative detection and quantification of dsDNA. POLYMER 2019. [DOI: 10.1016/j.polymer.2019.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
17
|
Circulating Tumor Cells for the Management of Renal Cell Carcinoma. Diagnostics (Basel) 2018; 8:diagnostics8030063. [PMID: 30177639 PMCID: PMC6164661 DOI: 10.3390/diagnostics8030063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
Renal cell carcinoma is a highly malignant cancer that would benefit from non-invasive innovative markers providing early diagnosis and recurrence detection. Circulating tumor cells are a particularly promising marker of tumor invasion that could be used to improve the management of patients with RCC. However, the extensive genetic and immunophenotypic heterogeneity of cells from RCC and their trend to transition to the mesenchymal phenotype when they circulate in blood constitute a challenge for their sensitive and specific detection. This review analyzes published studies targeting CTC in patients with RCC, in the context of the biological, pathological, and molecular complexity of this particular cancer. Although further analytical and clinical studies are needed to pinpoint the most suitable approach for highly sensitive CTC detection in RCC patients, it is clear that this field can bring a relevant guide to clinicians and help to RCC patients. Furthermore, as described, a particular subtype of RCC-the ccRCC-can be used as a model to study the relationship between cytomorphological and genetic cellular markers of malignancy, an important issue for the study of CTC from any type of solid cancer.
Collapse
|
18
|
MicroRNAs as Urinary Biomarker for Oncocytoma. DISEASE MARKERS 2018; 2018:6979073. [PMID: 30116406 PMCID: PMC6079495 DOI: 10.1155/2018/6979073] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/22/2018] [Accepted: 06/05/2018] [Indexed: 12/28/2022]
Abstract
The identification of benign renal oncocytoma, its differentiation from malignant renal tumors, and their eosinophilic variants are a continuous challenge, influencing preoperative planning and being an unnecessary stress factor for patients. Regressive changes enhance the diagnostic dilemma, making evaluations by frozen sections or by immunohistology (on biopsies) unreliable. MicroRNAs (miRs) have been proposed as novel biomarkers to differentiate renal tumor subtypes. However, their value as a diagnostic biomarker of oncocytoma in urines based on mechanisms known in oncocytomas has not been exploited. We used urines from patients with renal tumors (oncocytoma, renal cell carcinoma: clear cell, papillary, chromophobe) and with other urogenital lesions. miRs were extracted and detected via qRT-PCR, the respective tumors analyzed by immunohistology. We found isocitrate dehydrogenase 2 upregulated in oncocytoma and oncocytic chromophobe carcinoma, indicating an increased Krebs cycle metabolism. Since we had shown that all renal tumors are stimulated by endothelin-1, we analyzed miRs preidentified by microarray after endothelin-1 stimulation of renal epithelial cells. Four miRs are proposed as presurgical urinary biomarkers due to their known regulatory mechanism in oncocytoma: miR-498 (formation of the oncocytoma-specific slice-form of vimentin, Vim3), miR-183 (associated with increased CO2 levels), miR-205, and miR-31 (signaling through downregulation of PKC epsilon, shown previously).
Collapse
|
19
|
He Z, Xia L, Deng Z, Lian A, Hu Z, Li B. Identification of a VHL gene mutation in a Chinese family with Von Hippel‑Lindau syndrome. Mol Med Rep 2018; 18:435-440. [PMID: 29749453 DOI: 10.3892/mmr.2018.8974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/16/2018] [Indexed: 11/05/2022] Open
Abstract
Von Hippel‑Lindau (VHL) syndrome is an autosomal dominant neoplastic disorder. The VHL tumor suppressor (VHL) gene has previously been identified to represent the causative gene of VHL. Previous studies have demonstrated that >506 different mutations in VHL are associated with VHL syndrome. The aim of the present study was to determine the VHL gene mutation present in a VHL syndrome pedigree and to investigate the pathogenesis of the mutant protein. Briefly, a family suffering from VHL syndrome in a Chinese Han population was recruited, and a missense mutation (c.345 C>A: p.H115Q) was revealed to be present within the VHL gene in the proband. Furthermore, Sanger sequencing revealed two carriers of the mutation within the family. The results of the present study also demonstrated a mutation in VHL associated with the VHL syndrome phenotype, which may be of future therapeutic benefit for the diagnosis of VHL syndrome. These results may also be relevant to further studies aiming to investigate the molecular pathogenesis of VHL syndrome.
Collapse
Affiliation(s)
- Zhengwen He
- Department of Neurosurgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya, School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lu Xia
- Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhiyong Deng
- Department of Neurosurgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya, School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Aojie Lian
- Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhengmao Hu
- Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, P.R. China
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
20
|
Alekseyev YO, Fazeli R, Yang S, Basran R, Maher T, Miller NS, Remick D. A Next-Generation Sequencing Primer-How Does It Work and What Can It Do? Acad Pathol 2018; 5:2374289518766521. [PMID: 29761157 PMCID: PMC5944141 DOI: 10.1177/2374289518766521] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 12/28/2022] Open
Abstract
Next-generation sequencing refers to a high-throughput technology that determines the nucleic acid sequences and identifies variants in a sample. The technology has been introduced into clinical laboratory testing and produces test results for precision medicine. Since next-generation sequencing is relatively new, graduate students, medical students, pathology residents, and other physicians may benefit from a primer to provide a foundation about basic next-generation sequencing methods and applications, as well as specific examples where it has had diagnostic and prognostic utility. Next-generation sequencing technology grew out of advances in multiple fields to produce a sophisticated laboratory test with tremendous potential. Next-generation sequencing may be used in the clinical setting to look for specific genetic alterations in patients with cancer, diagnose inherited conditions such as cystic fibrosis, and detect and profile microbial organisms. This primer will review DNA sequencing technology, the commercialization of next-generation sequencing, and clinical uses of next-generation sequencing. Specific applications where next-generation sequencing has demonstrated utility in oncology are provided.
Collapse
Affiliation(s)
- Yuriy O Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Roghayeh Fazeli
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Shi Yang
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Raveen Basran
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Thomas Maher
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Nancy S Miller
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Daniel Remick
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| |
Collapse
|
21
|
Broncy L, Njima BB, Méjean A, Béroud C, Romdhane KB, Ilie M, Hofman V, Muret J, Hofman P, Bouhamed HC, Paterlini-Bréchot AP. Single-cell genetic analysis validates cytopathological identification of circulating cancer cells in patients with clear cell renal cell carcinoma. Oncotarget 2018; 9:20058-20074. [PMID: 29732003 PMCID: PMC5929446 DOI: 10.18632/oncotarget.25102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 03/24/2018] [Indexed: 12/14/2022] Open
Abstract
CONTEXT Circulating Rare Cells (CRC) are non-haematological cells circulating in blood. They include Circulating Cancer Cells (CCC) and cells with uncertain malignant features (CRC-UMF) according to cytomorphology. Clear cell renal cell carcinomas frequently bear a mutated Von Hippel-Lindau (VHL) gene. AIM To match blind genetic analysis of CRC and tumor samples with CRC cytopathological diagnosis. RESULTS 29/30 patients harboured CRC (20 harboured CCC, 29 CRC-UMF) and 25/29 patients carried VHL mutations in their tumour. 205 single CRC (64 CCC, 141 CRC-UMF) provided genetic data. 57/57 CCC and 104/125 CRC-UMF from the 25 patients with VHL-mutated tumor carried the same VHL mutation detected in the tumor. Seven CCC and 16 CRC-UMF did not carry VHL mutations but were found in patients with wild-type VHL tumor tissue. CONCLUSIONS All the CCC and 83,2% (104/125) of the CRC-UMF were found to carry the same VHL mutation identified in the corresponding tumorous tissue, validating cytopathological identification of CCC in patients with clear cell renal cell carcinoma. METHODS The blood of 30 patients with clear cell renal cell carcinoma was treated by ISET® for CRC isolation, cytopathology and single-cell VHL mutations analysis, performed blindly and compared to VHL mutations of corresponding tumor tissues and leukocytes.
Collapse
Affiliation(s)
- Lucile Broncy
- INSERM Unit 1151, Faculté de Médecine Paris Descartes, Paris, France
| | - Basma Ben Njima
- Genetics and Pathology Departments, University of Tunis, Tunis, Tunisia
| | - Arnaud Méjean
- Service d'Urologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Christophe Béroud
- Aix Marseille University, INSERM, MMG, Marseille, France
- APHM, Hôpital TIMONE Enfants, Laboratoire de Génétique Moléculaire, Marseille, France
| | | | - Marius Ilie
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | - Veronique Hofman
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | - Jane Muret
- Institut Curie, PSL Research University, Département d'Anesthésie Réanimation Douleur, Paris, France
| | - Paul Hofman
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | | | - And Patrizia Paterlini-Bréchot
- INSERM Unit 1151, Faculté de Médecine Paris Descartes, Paris, France
- Laboratoire de Biochimie A, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
22
|
Gaiser MR, von Bubnoff N, Gebhardt C, Utikal JS. Liquid Biopsy zur Überwachung von Melanompatienten. J Dtsch Dermatol Ges 2018; 16:405-416. [DOI: 10.1111/ddg.13461_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/21/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Maria Rita Gaiser
- Klinische Kooperationseinheit Klinische Kooperationseinheit Dermatoonkologie des Deutschen; Krebsforschungszentrums (DKFZ); Heidelberg Deutschland
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsmedizin Mannheim, Ruprecht-Karls-Universität Heidelberg; Mannheim Deutschland
| | - Nikolas von Bubnoff
- Klinik für Hämatologie; Onkologie und Stammzelltransplantation; Universitätsklinikum Freiburg; Deutschland
- Deutsches Konsortium für Translationale Krebsforschung (DKTK); Deutsches Krebsforschungszentrum (DKFZ); Heidelberg Deutschland
| | - Christoffer Gebhardt
- Klinische Kooperationseinheit Klinische Kooperationseinheit Dermatoonkologie des Deutschen; Krebsforschungszentrums (DKFZ); Heidelberg Deutschland
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsmedizin Mannheim, Ruprecht-Karls-Universität Heidelberg; Mannheim Deutschland
| | - Jochen Sven Utikal
- Klinische Kooperationseinheit Klinische Kooperationseinheit Dermatoonkologie des Deutschen; Krebsforschungszentrums (DKFZ); Heidelberg Deutschland
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsmedizin Mannheim, Ruprecht-Karls-Universität Heidelberg; Mannheim Deutschland
| |
Collapse
|
23
|
Gaiser MR, von Bubnoff N, Gebhardt C, Utikal JS. Liquid biopsy to monitor melanoma patients. J Dtsch Dermatol Ges 2018. [PMID: 29512873 DOI: 10.1111/ddg.13461] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During the last six years, several innovative, systemic therapies for the treatment of metastatic malignant melanoma (MM) have emerged. Conventional chemotherapy has been superseded by novel first-line therapies, including systemic immunotherapies (anti-CTLA4 and anti-PD1; authorization of anti-PDL1 is anticipated) and therapies targeting specific mutations (BRAF, NRAS, and c-KIT). Thus, treating physicians are confronted with new challenges, such as stratifying patients for appropriate treatments and monitoring long-term responders for progression. Consequently, reliable methods for monitoring disease progression or treatment resistance are necessary. Localized and advanced cancers may generate circulating tumor cells and circulating tumor DNA (ctDNA) that can be detected and quantified from peripheral blood samples (liquid biopsy). For melanoma patients, liquid biopsy results may be useful as novel predictive biomarkers to guide therapeutic decisions, particularly in the context of mutation-based targeted therapies. The challenges of using liquid biopsy include strict criteria for the phenotypic nature of circulating MM cells or their fragments and the instability of ctDNA in blood. The limitations of liquid biopsy in routine diagnostic testing are discussed in this review.
Collapse
Affiliation(s)
- Maria Rita Gaiser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Nikolas von Bubnoff
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center University of Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoffer Gebhardt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Jochen Sven Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| |
Collapse
|
24
|
El-Deiry WS, Taylor B, Neal JW. Tumor Evolution, Heterogeneity, and Therapy for Our Patients With Advanced Cancer: How Far Have We Come? Am Soc Clin Oncol Educ Book 2017; 37:e8-e15. [PMID: 28746017 DOI: 10.1200/edbk_175524] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The clinical and molecular heterogeneity of various cancer types is well documented. In the era of precision oncology whereby molecular profiling of tumors is incorporated into clinical care, both intra- and intertumoral molecular and genetic heterogeneity have been described. Together, they impact patient treatment and outcomes. Host genetics and the tumor microenvironment impact on tumor evolution and heterogeneity through variations in immune cell infiltration, stromal variations, and selection pressures from hypoxia or nutrient stress, among others. Tumor progression and exposure to therapeutic agents lead to further molecular evolution and heterogeneity that is clinically relevant. Moreover, tumors that evolve after diagnosis and as a function of therapy generally become more aggressive and refractory to available therapeutics, including targeted agents and immunotherapy. The evolving clinical and molecular heterogeneity of patient tumors can be explored with various clinical and research-based specimens and testing such as pre- and post-treatment biopsies; serial liquid biopsies; single cell analysis; PDX and organoid models; anatomic, functional, and molecular imaging; and rapid postmortem studies. Other factors that influence tumor heterogeneity include immune checkpoints, cancer stem cells, therapy-acquired resistance mechanisms that may occur through secondary mutations, and adaptive responses. Modern technologic advances for tumor characterization provide opportunities to understand tumor evolution and its impact on clinical outcomes to improve therapeutic regimens. Characterization of novel targets and development of effective therapeutics are needed to target heterogeneity and the evolution of resistance mechanisms.
Collapse
Affiliation(s)
- Wafik S El-Deiry
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; Stanford University, Palo Alto, CA
| | - Barry Taylor
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; Stanford University, Palo Alto, CA
| | - Joel W Neal
- From the Fox Chase Cancer Center, Philadelphia, PA; Memorial Sloan Kettering Cancer Center, New York, NY; Stanford University, Palo Alto, CA
| |
Collapse
|