1
|
Gouda MA, Ballesteros PA, Garrido-Laguna I, Rodon J. Efficacy assessment in phase I clinical trials: endpoints and challenges. Ann Oncol 2025; 36:507-519. [PMID: 40049448 DOI: 10.1016/j.annonc.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
The scope of phase I clinical trials in oncology goes beyond the conventional safety evaluation-only objectives of these trials in other specialties. Rather, most first-in-human oncology clinical trials have therapeutic intent, and efficacy signals observed in phase I trials can drive a go/no-go decision of advancing a new molecule to phase II testing. The complexity of efficacy assessment in the context of a small, heterogeneous patient population and a complex study design requires a more liberal perspective compared with later trial phases when looking into efficacy endpoints. Classically, in later-phase clinical trials, these endpoints would include the objective response rate, progression-free survival, and overall survival. However, new, evolving endpoints may be worth investigating when looking into the antitumor activity signals in phase I trials. Integration of all these endpoints into trial designs can improve the assessment of therapeutic efficacy during early drug development and guide decisions related to the further advancement of novel molecules into later phases. In this review, we discuss the advantages and pitfalls of different classic efficacy endpoints when evaluated as part of phase I trials in oncology and describe how challenges in assessing the antitumor activity of new drugs can be overcome through the incorporation of novel endpoints that have thus far proven successful in clinical trials.
Collapse
Affiliation(s)
- M A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - P A Ballesteros
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - I Garrido-Laguna
- Department of Medical Oncology, Huntsman Cancer Institute, Salt Lake City, USA
| | - J Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
2
|
Rahimi A, Baghernejadan Z, Hazrati A, Malekpour K, Samimi LN, Najafi A, Falak R, Khorramdelazad H. Combination therapy with immune checkpoint inhibitors in colorectal cancer: Challenges, resistance mechanisms, and the role of microbiota. Biomed Pharmacother 2025; 186:118014. [PMID: 40157004 DOI: 10.1016/j.biopha.2025.118014] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Colorectal cancer (CRC) is still one of the leading causes of cancer deaths worldwide. Even though there has been progress in cancer immunotherapy, the results of applying immune checkpoint inhibitors (ICIs) have been unsatisfactory, especially in microsatellite stable (MSS) CRC. Single-agent ICIs that target programmed cell death-1 (PD-1)/ PD-L1, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), T cell Ig- and mucin-domain-containing molecule-3 (TIM-3), and lymphocyte activation gene (LAG)-3 have emerged as having specific benefits. However, many primary and secondary resistance mechanisms are available in the tumor microenvironment (TME) that prevent it from happening. Combination strategies, such as the use of anti-PD-1 and anti-CTLA-4, can be effective in overcoming these resistance pathways, but toxicities remain a significant concern. Moreover, ICIs have been integrated with various treatment modalities, including chemotherapy, radiotherapy, antibiotics, virotherapy, polyadenosine diphosphate-ribose polymerase (PARP) inhibitors, and heat shock protein 90 (HSP90) inhibitors. The outcomes observed in both preclinical and clinical settings have been encouraging. Interestingly, manipulating gut microbiota via fecal microbiota transplantation (FMT) has been identified as a new strategy to increase the efficacy of immunotherapy in CRC patients. Therefore, integrating ICIs with other treatment approaches holds promise in enhancing the prognosis of CRC patients. This review focuses on the unmet need for new biomarkers to select patients for combination therapies and the ongoing work to overcome resistance and immune checkpoint blockade.
Collapse
Affiliation(s)
- Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
3
|
Lauzon-Young C, Silva A, Sadikovic B. Epigenomic insights and computational advances in hematologic malignancies. Mol Cytogenet 2025; 18:9. [PMID: 40221777 PMCID: PMC11993968 DOI: 10.1186/s13039-025-00712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
Hematologic malignancies (HMs) encompass a diverse spectrum of cancers originating from the blood, bone marrow, and lymphatic systems, with myeloid malignancies representing a significant and complex subset. This review provides a focused analysis of their classification, prevalence, and incidence, highlighting the persistent challenges posed by their intricate genetic and epigenetic landscapes in clinical diagnostics and therapeutics. The genetic basis of myeloid malignancies, including chromosomal translocations, somatic mutations, and copy number variations, is examined in detail, alongside epigenetic modifications with a specific emphasis on DNA methylation. We explore the dynamic interplay between genetic and epigenetic factors, demonstrating how these mechanisms collectively shape disease progression, therapeutic resistance, and clinical outcomes. Advances in diagnostic modalities, particularly those integrating epigenomic insights, are revolutionizing the precision diagnosis of HMs. Key approaches such as nano-based contrast agents, optical imaging, flow cytometry, circulating tumor DNA analysis, and somatic mutation testing are discussed, with particular attention to the transformative role of machine learning in epigenetic data analysis. DNA methylation episignatures have emerged as a pivotal tool, enabling the development of highly sensitive and specific diagnostic and prognostic assays that are now being adopted in clinical practice. We also review the impact of computational advancements and data integration in refining diagnostic and therapeutic strategies. By combining genomic and epigenomic profiling techniques, these innovations are accelerating biomarker discovery and clinical translation, with applications in precision oncology becoming increasingly evident. Comprehensive genomic datasets, coupled with artificial intelligence, are driving actionable insights into the biology of myeloid malignancies and facilitating the optimization of patient management strategies. Finally, this review emphasizes the translational potential of these advancements, focusing on their tangible benefits for patient care and outcomes. By synthesizing current knowledge and recent innovations, we underscore the critical role of precision medicine and epigenomic research in transforming the diagnosis and treatment of myeloid malignancies, setting the stage for ongoing advancements and broader clinical implementation.
Collapse
Affiliation(s)
- Carolyn Lauzon-Young
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, Canada
- Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Ananilia Silva
- Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Bekim Sadikovic
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON, Canada.
- Pathology and Laboratory Medicine, Western University, London, ON, Canada.
| |
Collapse
|
4
|
Sassorossi C, Evangelista J, Stefani A, Chiappetta M, Martino A, Campanella A, De Paolis E, Nachira D, Del Re M, Guerrera F, Boldrini L, Urbani A, Margaritora S, Minucci A, Bria E, Lococo F. The Role of ctDNA for Diagnosis and Histological Prediction in Early Stage Non-Small-Cell Lung Cancer: A Narrative Review. Diagnostics (Basel) 2025; 15:904. [PMID: 40218254 PMCID: PMC11988553 DOI: 10.3390/diagnostics15070904] [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: 02/04/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Circulating tumor DNA (ctDNA) may be released from neoplastic cells into biological fluids through apoptosis, necrosis, or active release. In patients with non-small-cell lung cancer (NSCLC), ctDNA analysis is being introduced in clinical practice only for advanced disease management. Nevertheless, an interesting and promising field of application is the analysis of ctDNA in the management of early stage non-small-cell lung cancer, both for evaluation before treatment, such as diagnosis and screening, and for prediction of histology or pathological features. Methods: A thorough review of the literature published between 2000 and 2024 was performed on PubMed, utilizing the advanced search feature to narrow down titles and abstracts containing the following keywords: ctDNA, early stage, and NSCLC. A total of 20 studies that met all inclusion criteria were chosen for this review. Results: In this review, we summarize the increasing evidence suggesting that ctDNA has potential clinical applications in the management of patients with early stage NSCLC. ctDNA levels in early stage cancers are very low, posing many technical challenges in improving the detection rate and sensitivity, especially in clinical practice, if it is to be implemented for early detection. Presently, the main limitation of ctDNA experimental and clinical studies, especially in early stage settings, is the lack of definitive standardization and consensus regarding methodology, the absence of systematically validated analyses, and the lack of adoption of sensitive approaches. Conclusions: Possible applications of this analyte open up new fields of diagnosis, treatment, and follow up, which are less invasive and more precise than other approaches currently in use, especially in early stage NSCLC patients.
Collapse
Affiliation(s)
- Carolina Sassorossi
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
| | - Jessica Evangelista
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
| | - Alessio Stefani
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.S.); (E.B.)
| | - Marco Chiappetta
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
- Thoracic Surgery Unit, University “Magna Graecia”, 88100 Catanzaro, Italy
| | - Antonella Martino
- Radiotherapy Unit, A. Gemelli University Hospital Foundation IRCCS, 00168 Rome, Italy;
| | - Annalisa Campanella
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
| | - Elisa De Paolis
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (A.M.)
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Dania Nachira
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
- Thoracic Surgery Unit, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marzia Del Re
- Department of Faculty Medicine, Saint Camillus International University of Medical and Health Sciences, 00131 Rome, Italy;
| | - Francesco Guerrera
- Department of Cardio-Thoracic and Vascular Surgery, Azienda Ospedaliera-Universitaria Città Della Salute e Della Scienza di Torino, 10126 Torino, Italy;
- Department of Surgical Sciences, University of Torino, 10126 Torino, Italy
| | - Luca Boldrini
- Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Andrea Urbani
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Stefano Margaritora
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
- Thoracic Surgery Unit, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Angelo Minucci
- Departmental Unit of Molecular and Genomic Diagnostics, Genomics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (A.M.)
| | - Emilio Bria
- Medical Oncology, Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.S.); (E.B.)
- UOC Oncologia Medica, Isola Tiberina Gemelli Isola, 00186 Rome, Italy
| | - Filippo Lococo
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (M.C.); (A.C.); (D.N.); (S.M.); (F.L.)
- Thoracic Surgery Unit, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
5
|
Cottrell TR, Lotze MT, Ali A, Bifulco CB, Capitini CM, Chow LQM, Cillo AR, Collyar D, Cope L, Deutsch JS, Dubrovsky G, Gnjatic S, Goh D, Halabi S, Kohanbash G, Maecker HT, Maleki Vareki S, Mullin S, Seliger B, Taube J, Vos W, Yeong J, Anderson KG, Bruno TC, Chiuzan C, Diaz-Padilla I, Garrett-Mayer E, Glitza Oliva IC, Grandi P, Hill EG, Hobbs BP, Najjar YG, Pettit Nassi P, Simons VH, Subudhi SK, Sullivan RJ, Takimoto CH. Society for Immunotherapy of Cancer (SITC) consensus statement on essential biomarkers for immunotherapy clinical protocols. J Immunother Cancer 2025; 13:e010928. [PMID: 40054999 DOI: 10.1136/jitc-2024-010928] [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] [Accepted: 02/05/2025] [Indexed: 03/12/2025] Open
Abstract
Immunotherapy of cancer is now an essential pillar of treatment for patients with many individual tumor types. Novel immune targets and technical advances are driving a rapid exploration of new treatment strategies incorporating immune agents in cancer clinical practice. Immunotherapies perturb a complex system of interactions among genomically unstable tumor cells, diverse cells within the tumor microenvironment including the systemic adaptive and innate immune cells. The drive to develop increasingly effective immunotherapy regimens is tempered by the risk of immune-related adverse events. Evidence-based biomarkers that measure the potential for therapeutic response and/or toxicity are critical to guide optimal patient care and contextualize the results of immunotherapy clinical trials. Responding to the lack of guidance on biomarker testing in early-phase immunotherapy clinical trials, we propose a definition and listing of essential biomarkers recommended for inclusion in all such protocols. These recommendations are based on consensus provided by the Society for Immunotherapy of Cancer (SITC) Clinical Immuno-Oncology Network (SCION) faculty with input from the SITC Pathology and Biomarker Committees and the Journal for ImmunoTherapy of Cancer readership. A consensus-based selection of essential biomarkers was conducted using a Delphi survey of SCION faculty. Regular updates to these recommendations are planned. The inaugural list of essential biomarkers includes complete blood count with differential to generate a neutrophil-to-lymphocyte ratio or systemic immune-inflammation index, serum lactate dehydrogenase and albumin, programmed death-ligand 1 immunohistochemistry, microsatellite stability assessment, and tumor mutational burden. Inclusion of these biomarkers across early-phase immunotherapy clinical trials will capture variation among trials, provide deeper insight into the novel and established therapies, and support improved patient selection and stratification for later-phase clinical trials.
Collapse
Affiliation(s)
- Tricia R Cottrell
- Queen's University Sinclair Cancer Research Institute, Kingston, Ontario, Canada
| | | | - Alaa Ali
- Stem Cell Transplant and Cellular Immunotherapy Program, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, Washington, DC, USA
| | - Carlo B Bifulco
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Christian M Capitini
- University of Wisconsin School of Medicine and Public Health and Carbone Cancer Center, Madison, Wisconsin, USA
| | | | - Anthony R Cillo
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Collyar
- Patient Advocates In Research (PAIR), Danville, California, USA
| | - Leslie Cope
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Susan Halabi
- Duke School of Medicine and Duke Cancer Institute, Durham, North Carolina, USA
| | - Gary Kohanbash
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Holden T Maecker
- Stanford University School of Medicine, Stanford, California, USA
| | - Saman Maleki Vareki
- Department of Oncology and Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Sarah Mullin
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Barbara Seliger
- Campus Brandenburg an der Havel, Brandenburg Medical School, Halle, Germany
| | - Janis Taube
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Wim Vos
- Radiomics.bio, Liège, Belgium
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Kristin G Anderson
- Department of Microbiology, Immunology and Cancer Biology, Department of Obstetrics and Gynecology, Beirne B. Carter Center for Immunology Research and the University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| | - Tullia C Bruno
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Codruta Chiuzan
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | | | | | | | | | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian P Hobbs
- Dell Medical School, The University of Texas, Austin, Texas, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Sumit K Subudhi
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Needham, Massachusetts, USA
| | | |
Collapse
|
6
|
Oscar-Thompson L, Riveros C, Sonpavde G, Apolo AB, Lalani AKA, Wallis CJD, Satkunasivam R. Adjuvant immunotherapy in high-risk muscle invasive urothelial carcinoma: A systematic review and meta-analysis of randomized clinical trials. Urol Oncol 2025; 43:156-163. [PMID: 39289120 PMCID: PMC11875964 DOI: 10.1016/j.urolonc.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/15/2024] [Accepted: 08/04/2024] [Indexed: 09/19/2024]
Abstract
Despite surgical resection, many patients with muscle invasive urothelial carcinoma (MIUC) experience recurrence. Adjuvant immune checkpoint inhibition (ICI) following radical resection in patients with MIUC demonstrates disparate outcomes among phase III randomized controlled trials (RCTs). Our objective was to synthesize available data regarding the disease-free survival (DFS) benefit of adjuvant ICIs for patients with MIUC and evaluate the overall safety profile of ICIs in this setting. The protocol was registered with PROSPERO, CRD42022352587. We searched MEDLINE, Embase, CENTRAL, and relevant conference proceedings from inception up to January 29, 2024. Only phase III RCTs comparing adjuvant ICI versus placebo/observation were selected. Study screening and selection, along with data extraction was performed in duplicate according to a predefined registered protocol. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline was used. Quality assessment was performed using the Cochrane risk-of-bias (RoB 2) tool for randomized trials. The primary and secondary endpoints were DFS and serious adverse events, respectively. All outcomes were analyzed using random-effects meta-analysis owing to inter-study heterogeneity. Sensitivity and subgroup analyses were performed to identify potential sources of heterogeneity. A priori defined subgroups of interest included positive program death-ligand 1 (PD-L1) expression, previous use of neoadjuvant chemotherapy (NAC), primary tumor origin, pathologic lymph node status, and baseline Eastern Cooperative Oncology Group performance status. Pooled results across the 3 RCTs (2,220 patients) demonstrated significantly improved DFS for patients treated with ICI in the intention-to-treat cohorts (HR 0.76, 95% CI 0.65-0.90). There was considerable clinical and statistical heterogeneity (I2 = 44%) due to differences in inclusion criteria and interventions. Overall, there was a low risk of bias among the RCTs. Regarding subgroup analyses, there was significant benefit among patients with negative PD-L1 expression (HR 0.76, 95% CI 0.64-0.90), those who received prior NAC (HR 0.69, 95% CI 0.52-0.91), and patients with lower tract (HR 0.71, 95% CI 0.55-0.92) but not upper tract disease (HR 1.21, 95% CI 0.87-1.68). This pooled analysis of DFS and safety provides support for ICI utilization in the setting of high-risk resected MIUC.
Collapse
Affiliation(s)
| | - Carlos Riveros
- Department of Urology, Houston Methodist Hospital, Houston, TX
| | - Guru Sonpavde
- Department of Hematology and Oncology, Genitourinary Oncology Program, AdventHealth Cancer Institute, Orlando, FL
| | - Andrea B Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Aly-Khan A Lalani
- Department of Oncology, Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Christopher J D Wallis
- Division of Urology and Surgical Oncology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada; Division of Urology, University of Toronto, Toronto, Ontario, Canada; Division of Urology, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | |
Collapse
|
7
|
Chen X, Wang HY, Yu L, Liu JQ, Sun H. Correlation of multiple peripheral blood parameters with metastasis and invasion of papillary thyroid cancer: a retrospective cohort study. Endocrine 2025:10.1007/s12020-025-04194-y. [PMID: 40025307 DOI: 10.1007/s12020-025-04194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE Papillary thyroid cancer (PTC) progression is characterized by lymph node metastasis and thyroid capsular invasion. This study aimed to identify high-risk PTC populations for these events based on peripheral blood test parameters and to determine the associated factors. METHODS This retrospective study analyzed data from 4557 PTC patients. Principal component analysis (PCA) and cluster analysis were performed on 45 peripheral blood test results. High- and low-risk clusters were defined based on metastasis and invasion prevalence. Univariate and multivariate analyses identified parameters significantly differentiating the clusters, examining their association with tumor progression. RESULTS Preoperative blood tests stratified patients into two distinct clusters. Cluster 0 demonstrated significantly higher rates of metastasis and invasion than Cluster 1, defining it as the high-risk group. PCA identified four principal components significantly differentiating the clusters. Analysis of these components revealed key peripheral blood parameters. Multivariable logistic regression identified six parameters associated with increased risk of Cluster 0: alanine aminotransferase, free triiodothyronine, thrombin time, hemoglobin, hematocrit, and leukocyte count. Conversely, aspartate aminotransferase and neutrophil count were associated with decreased risk. CONCLUSION These findings suggest that peripheral blood parameters may provide insights into the progression of thyroid tumors and highlight potential avenues for exploring the underlying mechanisms of PTC. However, given the retrospective nature of this study and the potential for selection bias, further prospective studies are necessary to validate these results and confirm their predictive value in clinical practice.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Medical Clinical Research Center for Diabetes and Metabolic Diseases, Wuhan, China
| | - Han-Yu Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Medical Clinical Research Center for Diabetes and Metabolic Diseases, Wuhan, China
| | - Lu Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Medical Clinical Research Center for Diabetes and Metabolic Diseases, Wuhan, China
| | - Jia-Qi Liu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Medical Clinical Research Center for Diabetes and Metabolic Diseases, Wuhan, China
| | - Hui Sun
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Medical Clinical Research Center for Diabetes and Metabolic Diseases, Wuhan, China.
| |
Collapse
|
8
|
Jang T, Kim GP, George TJ. Select updates from ASCO and ESMO 2024 for gastrointestinal cancer care. Oncologist 2025; 30:oyaf020. [PMID: 39998903 PMCID: PMC11853594 DOI: 10.1093/oncolo/oyaf020] [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: 09/01/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Gastrointestinal (GI) malignancies remain the culprit behind a substantial portion of cancer-related mortality worldwide, and outcomes for patients with advanced or metastatic GI cancers remain poor despite continued efforts to improve care. In 2024, the ongoing clinical trials to optimize and improve GI cancer care showcased progress in molecular diagnostics, systemic therapies, and localized treatment approaches, providing hope for continued progress toward improved patient outcomes. MATERIALS AND METHODS This review summarizes selected updates in GI cancer care from the 2024 ASCO and ESMO Annual Meetings, including both positive and negative trials that, while not universally practice-changing, contribute to shaping GI cancer care, clinical management, or address key questions in the field. The selected trials cover early detection and diagnostic advances, perioperative management, metastatic disease management, and immune checkpoint inhibitor (ICI)'s emerging role in GI cancer. RESULTS Various clinical trials in perioperative management and their results continue to reshape or strengthen the current treatment paradigms. The use of ICIs for microsatellite instability-high colorectal cancer (CRC) presented a notable advancement with the potential to improve patient outcomes. Localized treatments such as thermal ablation appear to benefit some patients with CRC and liver metastases. CONCLUSIONS The collection of trial results presented at the 2024 ASCO and ESMO Annual Meetings denote the ongoing efforts of the medical and scientific community for optimizing GI cancer care. The ongoing efforts of the GI cancer research and patient community provide hope for continued progress toward improved patient outcomes and new standards of care.
Collapse
Affiliation(s)
- Tim Jang
- University of Florida Health Cancer Center, Gainesville, FL 32608, United States
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608, United States
| | - George P Kim
- University of Florida Health Cancer Center, Gainesville, FL 32608, United States
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608, United States
| | - Thomas J George
- University of Florida Health Cancer Center, Gainesville, FL 32608, United States
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32608, United States
| |
Collapse
|
9
|
Piao M, Zhang N, Li J, Li C, Xun Z, Zhang L, Wang S, Sun B, Li S, Yang X, Yang X, Wang H, Zhao H. Peripheral blood PD-1 + T lymphocytes as biomarkers in liquid biopsies for solid tumors: Clinical significance and prognostic applications. Int Immunopharmacol 2025; 147:114052. [PMID: 39799737 DOI: 10.1016/j.intimp.2025.114052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
A shift toward a T cell exhaustion phenotype is associated with the upregulation of expression of programmed cell death protein 1 (PD-1) on T lymphocytes in patients with malignant solid tumors. The interaction between PD-1 and programmed death-ligand 1 (PD-L1) inhibits PD-1+ T lymphocyte function, impacting their anti-tumor immune activity. Immune checkpoint inhibitors targeting PD-1/PD-L1 have revolutionized the treatment of various solid malignancies, improving therapeutic efficacy and survival outcomes. Peripheral blood analysis of liquid biopsies is being increasingly used to identify populations most likely to benefit from various treatment modalities. PD-1+ T lymphocytes represent the primary cell population responsive to immunotherapeutic interventions for patients with solid malignancies, as evidenced by the altered PD-1 expression levels and proportion of cells comprising the overall population of immunocytes. PD-1+ T cells in peripheral blood exert an associative and reciprocal predictive effect on homologous intratumoral cells. Distinct subpopulations of PD-1+ T cells exhibit differential ability to proliferate in the periphery and can be characterized by tumor antigen-specific and exhaustion phenotypes. These characteristics have prognostic implications, aiding in the prediction of the efficacy of antitumor therapy and predicting survival outcomes. We highlight distinct subpopulations of PD-1+ T cells, their exhaustion and antigen-specific phenotypes, and their dynamic changes over treatment, providing insights into their utility for tailoring personalized therapies. For the first time, this review discusses the role of peripheral PD-1+ T lymphocytes as prognostic biomarkers in liquid biopsies, focusing on their clinical significance, predictive value during therapy, and future research directions.
Collapse
Affiliation(s)
- Mingjian Piao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Jiongyuan Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Chengjie Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Ziyu Xun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Boyu Sun
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Shuofeng Li
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Xu Yang
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China.
| | - Hanping Wang
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences &Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Tian H, Shi H, Chen J, Zhu T, Huang Z, Zong C, Jia S, Ruan J, Ge S, Yuan H, Zhang Y, Jiang B, Liu R, Jia R, Fan X, Xu S. Circulating Tumor DNA in Conjunctival Melanoma: Landscape and Surveillance Value. Am J Ophthalmol 2025; 270:67-76. [PMID: 39424026 DOI: 10.1016/j.ajo.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE To evaluate the surveillance value of circulating tumor DNA (ctDNA) for detecting distant metastasis and indicating systemic therapeutic efficacy in conjunctival melanoma (CoM). DESIGN Retrospective, observational case series. METHODS From July 2021 to June 2023, 30 CoM patients in our center underwent plasma ctDNA assessment, out of which 12 individuals presented with distant metastases. We employed a 437-gene panel containing common mutations in CoM and common drug-sensitive mutations using next-generation sequencing (NGS) technology to analyze ctDNA mutations in plasma. Clinical and radiological records were used to assess tumor status. The relationship between ctDNA characteristics, tissue gene mutations, and clinical manifestations were explored. RESULTS CoM-related driver mutations were detected in ctDNA of 11 patients with distant metastasis. The ctDNA were highly consistent with tissue sequencing, mutual driver mutation including BRAF, NRAS, KRAS, NF1, CTNNB1, and TP53 mutation. those with a higher VAF had shorter progression-free survival (PFS, p = .0475) and overall survival (OS, p = .0043). The ctDNA variant allele fraction (VAF) was not correlated with the sum of the longest diameters (SLD, p = .8192) in distant metastasis patients. CONCLUSIONS Positive plasma ctDNA reflected the presence of metastases. The ctDNA could be used as a complement or alternative to tissue sequencing. High VAF ctDNA might indicate rapid disease progression in distant metastasis patients.
Collapse
Affiliation(s)
- Hao Tian
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Hanhan Shi
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Jie Chen
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Tianyu Zhu
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Ziyue Huang
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Chunyan Zong
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Shichong Jia
- Tianjin Eye Hospital (S.J.), Tianjin Key Lab of Ophthalmology and Visual Science, Nankai University Affiliated Eye Hospital, Tianjin Eye Institute, Tianjin, China
| | - Jing Ruan
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Shengfang Ge
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Haihua Yuan
- Department of Oncology (H.Y., Y.Z., B.J.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjie Zhang
- Department of Oncology (H.Y., Y.Z., B.J.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Jiang
- Department of Oncology (H.Y., Y.Z., B.J.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Liu
- Opthalmology Department of Tongji Hospital (R.L.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renbing Jia
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China
| | - Xianqun Fan
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China.
| | - Shiqiong Xu
- From the Department of Ophthalmology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., X.F., S.X.), Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology (H.T., H.S., J.C., T.Z., Z.H., C.Z., J.R., S.G., R.J., S.X.), Shanghai, China.
| |
Collapse
|
11
|
Lutfi A, Afghan MK, Kasi PM. CTCs and liquid biopsies in patients with colorectal cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2025; 392:101-117. [PMID: 40287217 DOI: 10.1016/bs.ircmb.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Liquid biopsy, which includes both circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs) has become a valuable tool for cancer diagnosis and monitoring. It offers a less invasive approach than traditional tissue biopsy and recent technological advancements have enabled their use in comprehensive analysis of tumor molecular characteristics. By capturing the dynamic nature of tumors through repeated sampling, liquid biopsy addresses the limitations of tissue biopsy and provides insights into tumor heterogeneity over time. It is being extensively studied in patients with advanced colorectal cancer because it can aid in diagnosis, predict disease course, and guide treatment selection. Furthermore, as personalized medicine becomes more common, identifying genetic changes that cause cancer cells to become resistant to treatment is crucial. This chapter explores the emerging field of liquid biopsy, with a particular focus on the role and potential of circulating tumor cells (CTCs) in the context of colorectal cancer.
Collapse
Affiliation(s)
- Areeb Lutfi
- Department of Hematology and Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Maaz Khan Afghan
- Department of Hematology and Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Pashtoon Murtaza Kasi
- Department of Oncology and Therapeutics Research, City of Hope, Irvine, CA, United States.
| |
Collapse
|
12
|
Cao L, Duan Q, Zhu Z, Xu X, Liu J, Li B. Liquid biopsy technologies: innovations and future directions in breast cancer biomarker detection. Biomed Microdevices 2025; 27:4. [PMID: 39849252 DOI: 10.1007/s10544-025-00734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 01/25/2025]
Abstract
Globally, breast cancer is the most frequent type of cancer, and its early diagnosis and screening can significantly improve the probability of survival and quality of life of those affected. Liquid biopsy-based targets such as circulating tumor cells, circulating tumor DNA, and exosomes have been instrumental in the early discovery of cancer, and have been found to be effective in stage therapy, recurrence monitoring, and drug selection. Biosensors based on these target related biomarkers convert the tested substances into quantifiable signals such as electrical and optical signals through signal transduction, which has the advantages of high sensitivity, simple operation, and low invasiveness. This review provides an overview of the latest progress of liquid biopsy biomarkers in the diagnosis, prognosis and treatment of breast cancer, compares the application and advantages of different biosensors based on these biomarkers in the diagnosis of breast cancer, and analyzes the limitations and solutions of biosensor based methods.
Collapse
Affiliation(s)
- Linhong Cao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Qingli Duan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Zixin Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Xuejing Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| |
Collapse
|
13
|
Martínez-Vila C, Teixido C, Aya F, Martín R, González-Navarro EA, Alos L, Castrejon N, Arance A. Detection of Circulating Tumor DNA in Liquid Biopsy: Current Techniques and Potential Applications in Melanoma. Int J Mol Sci 2025; 26:861. [PMID: 39859576 PMCID: PMC11766255 DOI: 10.3390/ijms26020861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
The treatment landscape for advanced melanoma has transformed significantly with the advent of BRAF and MEK inhibitors (BRAF/MEKi) targeting BRAFV600 mutations, as well as immune checkpoint inhibitors (ICI) like anti-PD-1 monotherapy or its combinations with anti-CTLA-4 or anti-LAG-3. Despite that, many patients still do not benefit from these treatments at all or develop resistance mechanisms. Therefore, prognostic and predictive biomarkers are needed to identify patients who should switch or escalate their treatment strategies or initiate an intensive follow-up. In melanoma, liquid biopsy has shown promising results, with a potential role in predicting relapse in resected high-risk patients or in disease monitoring during the treatment of advanced disease. Several components in peripheral blood have been analyzed, such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), and circulant tumoral DNA (ctDNA), which have turned out to be particularly promising. To analyze ctDNA in blood, different techniques have proven to be useful, including digital droplet polymerase chain reaction (ddPCR) to detect specific mutations and, more recently, next-generation sequencing (NGS) techniques, which allow analyzing a broader repertoire of the mutation landscape of each patient. In this review, our goal is to update the current understanding of liquid biopsy, focusing on the use of ctDNA as a biological material in the daily clinical management of melanoma patients, in particular those with advanced disease treated with ICI.
Collapse
Affiliation(s)
- Clara Martínez-Vila
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Dr. Joan Soler, 1–3, 08243 Manresa, Spain;
- Programa de Doctorat en Medicina i Recerca Translacional, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), Roda 70, 08500 Vic, Spain
| | - Cristina Teixido
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (C.T.); (L.A.); (N.C.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
| | - Francisco Aya
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Roberto Martín
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Europa Azucena González-Navarro
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Immunology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Llucia Alos
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (C.T.); (L.A.); (N.C.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
| | - Natalia Castrejon
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (C.T.); (L.A.); (N.C.)
| | - Ana Arance
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (F.A.); (R.M.); (E.A.G.-N.)
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain
| |
Collapse
|
14
|
Ahuja S, Zaheer S. Advancements in pathology: Digital transformation, precision medicine, and beyond. J Pathol Inform 2025; 16:100408. [PMID: 40094037 PMCID: PMC11910332 DOI: 10.1016/j.jpi.2024.100408] [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: 09/24/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 01/02/2025] Open
Abstract
Pathology, a cornerstone of medical diagnostics and research, is undergoing a revolutionary transformation fueled by digital technology, molecular biology advancements, and big data analytics. Digital pathology converts conventional glass slides into high-resolution digital images, enhancing collaboration and efficiency among pathologists worldwide. Integrating artificial intelligence (AI) and machine learning (ML) algorithms with digital pathology improves diagnostic accuracy, particularly in complex diseases like cancer. Molecular pathology, facilitated by next-generation sequencing (NGS), provides comprehensive genomic, transcriptomic, and proteomic insights into disease mechanisms, guiding personalized therapies. Immunohistochemistry (IHC) plays a pivotal role in biomarker discovery, refining disease classification and prognostication. Precision medicine integrates pathology's molecular findings with individual genetic, environmental, and lifestyle factors to customize treatment strategies, optimizing patient outcomes. Telepathology extends diagnostic services to underserved areas through remote digital pathology. Pathomics leverages big data analytics to extract meaningful insights from pathology images, advancing our understanding of disease pathology and therapeutic targets. Virtual autopsies employ non-invasive imaging technologies to revolutionize forensic pathology. These innovations promise earlier diagnoses, tailored treatments, and enhanced patient care. Collaboration across disciplines is essential to fully realize the transformative potential of these advancements in medical practice and research.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
15
|
Prkačin I, Mokos M, Ferara N, Šitum M. Melanoma's New Frontier: Exploring the Latest Advances in Blood-Based Biomarkers for Melanoma. Cancers (Basel) 2024; 16:4219. [PMID: 39766118 PMCID: PMC11727356 DOI: 10.3390/cancers16244219] [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: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025] Open
Abstract
Melanoma is one of the most malignant cancers, and the global incidence of cutaneous melanoma is increasing. While melanomas are highly prone to metastasize if diagnosed late, early detection and treatment significantly reduce the risk of mortality. Identifying patients at higher risk of metastasis, who might benefit from early adjuvant therapies, is particularly important, especially with the advent of new melanoma treatments. Therefore, there is a pressing need to develop additional prognostic biomarkers for melanoma to improve early stratification of patients and accurately identify high-risk subgroups, ultimately enabling more effective personalized treatments. Recent advances in melanoma therapy, including targeted treatments and immunotherapy, have underscored the importance of biomarkers in determining prognosis and predicting treatment response. The clinical application of these markers holds the potential for significant advancements in melanoma management. Various tumor-derived genetic, proteomic, and cellular components are continuously released into the bloodstream of cancer patients. These molecules, including circulating tumor DNA and RNA, proteins, tumor cells, and immune cells, are emerging as practical and precise liquid biomarkers for cancer. In the current era of effective molecular-targeted therapies and immunotherapies, there is an urgent need to integrate these circulating biomarkers into clinical practice to facilitate personalized treatment. This review highlights recent discoveries in circulating melanoma biomarkers, explores the challenges and potentials of emerging technologies for liquid biomarker discovery, and discusses future directions in melanoma biomarker research.
Collapse
Affiliation(s)
- Ivana Prkačin
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
- School of Medicine, University of Split, 21000 Split, Croatia
| | - Mislav Mokos
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
| | - Nikola Ferara
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
| | - Mirna Šitum
- Department of Dermatology and Venereology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (M.M.); (N.F.); (M.Š.)
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Croatian Academy of Sciences and Arts, 10000 Zagreb, Croatia
| |
Collapse
|
16
|
Kou M, Deng Y. Circulating tumor DNA as a predictive biomarker for treatment response and survival in metastatic colorectal cancer. Int J Colorectal Dis 2024; 39:203. [PMID: 39681775 DOI: 10.1007/s00384-024-04785-7] [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] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
PURPOSE To explore the potential of circulating tumor DNA (ctDNA) as a prognostic biomarker to predict treatment response and survival outcomes in patients with metastatic colorectal cancer (mCRC). METHODS A retrospective analysis was conducted on 134 patients with mCRC who were treated between January 2020 and December 2021. The patients were classified into ctDNA-negative and ctDNA-positive groups based on plasma ctDNA detection. Demographic, clinical, and laboratory parameters, treatment response, survival outcomes, and adverse events were recorded and analyzed. RESULTS No significant differences were observed in baseline characteristics between the two groups. Compared to the ctDNA-positive patients, ctDNA-negative patients exhibited superior outcomes, including a higher objective response rate (65.22% vs. 46.15%), disease control rate (81.16% vs. 63.08%), progression-free survival (8.24 ± 1.02 vs. 7.86 ± 0.91 months), overall survival (24.58 ± 3.58 vs. 23.27 ± 3.46 months), and 1-year survival rate (73.91% vs. 55.38%). The ctDNA-positive group had a significantly higher incidence of adverse events. Correlation analyses revealed significant associations between ctDNA status, tumor markers, treatment response, and survival outcomes. CONCLUSIONS ctDNA is a promising noninvasive biomarker for predicting treatment response, survival, and adverse events in mCRC, potentially guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Kou
- Department of Oncology, Maoming People's Hospital, Maoming City, Guangdong Province, China
| | - Ying Deng
- Department of Gastroenterology, Maoming People's Hospital, No.101 Weimin Road, Maonan District, Maoming City, Guangdong Province, 525000, China.
| |
Collapse
|
17
|
Si HQ, Wang P, Long F, Zhong W, Meng YD, Rong Y, Meng XY, Wang FB. Cancer liquid biopsies by Oxford Nanopore Technologies sequencing of cell-free DNA: from basic research to clinical applications. Mol Cancer 2024; 23:265. [PMID: 39614371 PMCID: PMC11605934 DOI: 10.1186/s12943-024-02178-6] [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: 10/09/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024] Open
Abstract
Liquid biopsies, in particular, analysis of cell-free DNA, are expected to revolutionize the current landscape of cancer diagnostics and treatment. However, the existing methods for cfDNA-based liquid biopsies for cancer have certain limitations, such as fragment interruption and GC bias, which are likely to be resolved by the emerging Oxford Nanopore Technologies (ONT), characterized by long read-length, fast read-times, high throughput, and polymerase chain reaction-free. In this review, we summarized the current literatures regarding the feasibility and applications of cfDNA-based liquid biopsies using ONT for cancer management, a possible game-changer that we believe is promising in detecting multimodal biomarkers and can be applied in a wide range of oncology utilities including early screening, diagnosis, and treatment monitoring.
Collapse
Affiliation(s)
- Hua-Qi Si
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fei Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Zhong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan-Dong Meng
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Hubei Minzu University, Enshi, China
| | - Yuan Rong
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Xiang-Yu Meng
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Hubei Minzu University, Enshi, China.
| | - Fu-Bing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
18
|
Yin X, Song Y, Deng W, Blake N, Luo X, Meng J. Potential predictive biomarkers in antitumor immunotherapy: navigating the future of antitumor treatment and immune checkpoint inhibitor efficacy. Front Oncol 2024; 14:1483454. [PMID: 39655071 PMCID: PMC11625675 DOI: 10.3389/fonc.2024.1483454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment modality, offering promising outcomes for various malignancies. However, the efficacy of ICIs varies among patients, highlighting the essential need of accurate predictive biomarkers. This review synthesizes the current understanding of biomarkers for ICI therapy, and discusses the clinical utility and limitations of these biomarkers in predicting treatment outcomes. It discusses three US Food and Drug Administration (FDA)-approved biomarkers, programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), and microsatellite instability (MSI), and explores other potential biomarkers, including tumor immune microenvironment (TIME)-related signatures, human leukocyte antigen (HLA) diversity, non-invasive biomarkers such as circulating tumor DNA (ctDNA), and combination biomarker strategies. The review also addresses multivariable predictive models integrating multiple features of patients, tumors, and TIME, which could be a promising approach to enhance predictive accuracy. The existing challenges are also pointed out, such as the tumor heterogeneity, the inconstant nature of TIME, nonuniformed thresholds and standardization approaches. The review concludes by emphasizing the importance of biomarker research in realizing the potential of personalized immunotherapy, with the goal of improving patient selection, treatment strategies, and overall outcomes in cancer treatment.
Collapse
Affiliation(s)
- Xiangyu Yin
- Department of Biological Sciences, School of Science, AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Yunjie Song
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Wanglong Deng
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Neil Blake
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Xinghong Luo
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Jia Meng
- Department of Biological Sciences, School of Science, AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
19
|
Tegeler CM, Hartkopf AD, Banys-Paluchowski M, Krawczyk N, Fehm T, Jaeger BAS. Circulating Tumor DNA in Early and Metastatic Breast Cance-Current Role and What Is Coming Next. Cancers (Basel) 2024; 16:3919. [PMID: 39682108 DOI: 10.3390/cancers16233919] [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: 09/15/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 12/18/2024] Open
Abstract
The progress that has been made in recent years in relation to liquid biopsies in general and circulating tumor DNA (ctDNA) in particular can be seen as groundbreaking for the future of breast cancer treatment, monitoring and early detection. Cell-free DNA (cfDNA) consists of circulating DNA fragments released by various cell types into the bloodstream. A portion of this cfDNA, known as ctDNA, originates from malignant cells and carries specific genetic mutations. Analysis of ctDNA provides a minimally invasive method for diagnosis, monitoring response to therapy, and detecting the emergence of resistance. Several methods are available for the analysis of ctDNA, each with distinct advantages and limitations. Quantitative polymerase chain reaction is a well-established technique widely used due to its high sensitivity and specificity, particularly for detecting known mutations. In addition to the detection of individual mutations, multigene analyses were developed that could detect several mutations at once, including rarer mutations. These methods are complementary and can be used strategically depending on the clinical question. In the context of metastatic breast cancer, ctDNA holds particular promise as it allows for the dynamic monitoring of tumor evolution. Through ctDNA analysis, mutations in the ESR1 or PIK3CA genes, which are associated with therapy resistance, can be identified. This enables the early adjustment of treatment and has the potential to significantly enhance clinical outcome. The application of ctDNA in early breast cancer is an ongoing investigation. In (neo)adjuvant settings, there is preliminary data indicating that ctDNA can be used for therapy monitoring and risk stratification to decide on post-neoadjuvant strategies. In the monitoring of aftercare, the detection of ctDNA appears to be several months ahead of routine imaging. However, the feasibility of implementing this approach in a clinical setting remains to be seen. While the use of ctDNA as a screening method for the asymptomatic population would be highly advantageous due to its minimally invasive nature, the available data on its clinical benefit are still insufficient. Nevertheless, ctDNA represents the most promising avenue for fulfilling this potential future need.
Collapse
Affiliation(s)
- Christian Martin Tegeler
- Department of Obstetrics and Gynecology, University Hospital Tübingen, 72076 Tübingen, Germany
- Department of Peptide-Based Immunotherapy, Institute of Immunology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas Daniel Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Maggie Banys-Paluchowski
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Luebeck, 23538 Luebeck, Germany
| | - Natalia Krawczyk
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, 40225 Duesseldorf, Germany
- Center for Integrated Oncology (CIO) ABCD, 40225 Duesseldorf, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, 40225 Duesseldorf, Germany
- Center for Integrated Oncology (CIO) ABCD, 40225 Duesseldorf, Germany
| | - Bernadette Anna Sophia Jaeger
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, 40225 Duesseldorf, Germany
- Center for Integrated Oncology (CIO) ABCD, 40225 Duesseldorf, Germany
| |
Collapse
|
20
|
Li S, Yuan T, Yuan J, Zhu B, Chen D. Opportunities and challenges of using circulating tumor DNA to predict lung cancer immunotherapy efficacy. J Cancer Res Clin Oncol 2024; 150:501. [PMID: 39545998 PMCID: PMC11568038 DOI: 10.1007/s00432-024-06030-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Immune checkpoint inhibitors (ICIs), particularly anti-programmed death 1 (PD-1)/ programmed death ligand 1 (PD-L1) antibodies, have led to significant progress in lung cancer treatment. However, only a minority of patients have responses to these therapies. Detecting peripheral blood of circulating tumor DNA (ctDNA) allows minimally invasive diagnosis, characterization, and monitoring of lung cancer. ctDNA has potential to be a prognostic biomarker and a predictor of the response to ICI therapy since it can indicate the genomic status and tumor burden. Recent studies on lung cancer have shown that pretreatment ctDNA analysis can detect residual proliferative disease in the adjuvant immunotherapy setting and evaluate tumor burden in patients with metastatic disease. Early ctDNA dynamics can not only predict the clinical outcome of ICI therapy but also help distinguish between pseudoprogression and real progression. Furthermore, in addition to quantitative assessment, ctDNA can also detect genetic predictors of response to ICI therapy. However, barriers still exist in the application of ctDNA analysis in clinical lung cancer treatment. The predictive value of ctDNA in lung cancer immunotherapy requires further identification and resolution of these challenges. This review aims to summarize the existing data of ctDNA analysis in patients receiving immunotherapy for lung cancer, understand the limitations of clinical treatment, and discuss future research directions.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jing Yuan
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
21
|
Wyatt AW, Litiere S, Bidard FC, Cabel L, Dyrskjøt L, Karlovich CA, Pantel K, Petrie J, Philip R, Andrews HS, Vellanki PJ, Tolmeijer SH, Villalobos Alberu X, Alfano C, Bogaerts J, Calvo E, Chen AP, Toledo RA, de Vries EGE, Seymour L, Laurie SA, Garralda E. Plasma ctDNA as a Treatment Response Biomarker in Metastatic Cancers: Evaluation by the RECIST Working Group. Clin Cancer Res 2024; 30:5034-5041. [PMID: 39269996 DOI: 10.1158/1078-0432.ccr-24-1883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Early indicators of metastatic cancer response to therapy are important for evaluating new drugs and stopping ineffective treatment. The RECIST guidelines based on repeat cancer imaging are widely adopted in clinical trials, are used to identify active regimens that may change practice, and contribute to regulatory approvals. However, these criteria do not provide insight before 6 to 12 weeks of treatment and typically require that patients have measurable disease. Recent data suggest that measuring on-treatment changes in the amount or proportion of ctDNA in peripheral blood plasma may accurately identify responding and nonresponding cancers at earlier time points. Over the past year, the RECIST working group has evaluated current evidence for plasma ctDNA kinetics as a treatment response biomarker in metastatic cancers and early endpoint in clinical trials to identify areas of focus for future research and validation. Here, we outline the requirement for large standardized trial datasets, greater scrutiny of optimal ctDNA collection time points and assay thresholds, and consideration of regulatory body guidelines and patient opinions. In particular, clinically meaningful changes in plasma ctDNA abundance are likely to differ by cancer type and therapy class and must be assessed before ctDNA can be considered a potential pan-cancer response evaluation biomarker. Despite the need for additional data, minimally invasive on-treatment ctDNA measurements hold promise to build upon existing response assessments such as RECIST and offer opportunities for developing novel early endpoints for modern clinical trials.
Collapse
Affiliation(s)
- Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre and Clinical Cancer Genomics Program, BC Cancer, Vancouver, British Columbia, Canada
| | - Saskia Litiere
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Francois-Clément Bidard
- Department of Medical Oncology, Institut Curie, Université Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Université Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Lars Dyrskjøt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Chris A Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Klaus Pantel
- Department of Tumor Biology, Center for Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joan Petrie
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | - Reena Philip
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | | | - Paz J Vellanki
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Sofie H Tolmeijer
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Christian Alfano
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | | | - Elisabeth G E de Vries
- University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Scott A Laurie
- Division of Medical Oncology, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Elena Garralda
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
22
|
Gang X, Yan J, Li X, Shi S, Xu L, Liu R, Cai L, Li H, Zhao M. Immune checkpoint inhibitors rechallenge in non-small cell lung cancer: Current evidence and future directions. Cancer Lett 2024; 604:217241. [PMID: 39260670 DOI: 10.1016/j.canlet.2024.217241] [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: 04/25/2024] [Revised: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Immunotherapy, remarkably immune checkpoint inhibitors (ICIs), has significantly altered the treatment landscape for non-small cell lung cancer (NSCLC). Despite their success, the discontinuation of ICIs therapy may occur due to factors such as prior treatment completion, disease progression during ICIs treatment, or immune-related adverse events (irAEs). As numerous studies highlight the dynamic nature of immune responses and the sustained benefits of ICIs, ICIs rechallenge has become an attractive and feasible option. However, the decision-making process for ICIs rechallenge in clinical settings is complicated by numerous uncertainties. This review systematically analyses existing clinical research evidence, classifying ICIs rechallenge into distinct clinical scenarios, exploring methods to overcome ICIs resistance in rechallenge instances, and identifying biomarkers to select patients likely to benefit from rechallenge. By integrating recent studies and new technologies, we offer crucial recommendations for future clinical trial design and provide a practical guideline to maximize the therapeutic benefits of immunotherapy for NSCLC patients.
Collapse
Affiliation(s)
- Xiaoyu Gang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jinshan Yan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xin Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Sha Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Lu Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ruotong Liu
- Clinical Medicine, Shenyang Medical College, Shenyang, 110001, China
| | - Lutong Cai
- Psychological Medicine, Shenyang Medical College, Shenyang, 110001, China
| | - Heming Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China; Guangdong Association of Clinical Trials (GACT)/Chinese Thoracic Oncology Group (CTONG) and Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, Guangzhou, 510000, China.
| | - Mingfang Zhao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
23
|
Hu Q, Chen L, Li K, Liu R, Sun L, Han T. Circulating tumor DNA: current implementation issues and future challenges for clinical utility. Clin Chem Lab Med 2024; 62:2094-2110. [PMID: 38109307 DOI: 10.1515/cclm-2023-1157] [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: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Over the past decades, liquid biopsy, especially circulating tumor DNA (ctDNA), has received tremendous attention as a noninvasive detection approach for clinical applications, including early diagnosis of cancer and relapse, real-time therapeutic efficacy monitoring, potential target selection and investigation of drug resistance mechanisms. In recent years, the application of next-generation sequencing technology combined with AI technology has significantly improved the accuracy and sensitivity of liquid biopsy, enhancing its potential in solid tumors. However, the increasing integration of such promising tests to improve therapy decision making by oncologists still has complexities and challenges. Here, we propose a conceptual framework of ctDNA technologies and clinical utilities based on bibliometrics and highlight current challenges and future directions, especially in clinical applications such as early detection, minimal residual disease detection, targeted therapy, and immunotherapy. We also discuss the necessities of developing a dynamic field of translational cancer research and rigorous clinical studies that may support therapeutic strategy decision making in the near future.
Collapse
Affiliation(s)
- Qilin Hu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Lujun Chen
- The General Hospital of Northern Theater Command Training Base for Graduate, China Medical University, Shenyang, P.R. China
| | - Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Ruotong Liu
- Clinical Medicine, Shenyang Medical College, Shenyang, P.R. China
| | - Lei Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, P.R. China
| |
Collapse
|
24
|
Fairley JA, Badrick T, Denis MG, Dimitrova L, Goodall R, Maas J, Normanno N, Patton SJ, Rouleau E, Russo A, Stockley TL, Deans ZC. Implementation of circulating tumour DNA multi-target mutation testing in plasma: a perspective from an external quality assessment providers' survey. Virchows Arch 2024; 485:717-722. [PMID: 37202567 PMCID: PMC11522039 DOI: 10.1007/s00428-023-03558-x] [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: 02/23/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/20/2023]
Abstract
Demand for large-scale tumour profiling across cancer types has increased in recent years, driven by the emergence of targeted drug therapies. Analysing alternations in plasma circulating tumour DNA (ctDNA) for cancer detection can improve survival; ctDNA testing is recommended when tumour tissue is unavailable. An online survey of molecular pathology testing was circulated by six external quality assessment members of IQN Path to registered laboratories and all IQN Path collaborative corporate members. Data from 275 laboratories across 45 countries were collected; 245 (89%) perform molecular pathology testing, including 177 (64%) which perform plasma ctDNA diagnostic service testing. The most common tests were next-generation sequencing-based (n = 113). Genes with known stratified treatment options, including KRAS (n = 97), NRAS (n = 84), and EGFR (n = 130), were common targets. The uptake of ctDNA plasma testing and plans to implement further testing demonstrates the importance of support from a well-designed EQA scheme.
Collapse
Affiliation(s)
- Jennifer A Fairley
- GenQA, Department of Laboratory Medicine, NHS Lothian, Nine Bioquarter, Little France Rd, Edinburgh, EH16 4UX, UK
| | - Tony Badrick
- The Royal College of Pathologists of Australasia, Quality Assurance Programs (RCPAQAP), St. Leonards, Australia
| | - Marc G Denis
- Nantes Université, CHU Nantes, Department of Biochemistry, INSERM, CNRS, Immunology and New Concepts in Immunotherapy, Nantes, France
| | | | - Rebecca Goodall
- EMQN CIC, Unit 4, Enterprise House, Manchester Science Park, Pencroft Way, Manchester, M15 6SE, UK
| | - Joerg Maas
- Deutsche Gesellschaft für Pathologie E.V. (DGP), Berlin, Germany
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - "Fondazione G. Pascale", Via Mariano Semola, 80131, Napoli, Italy
| | - Simon J Patton
- EMQN CIC, Unit 4, Enterprise House, Manchester Science Park, Pencroft Way, Manchester, M15 6SE, UK
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Gustave Roussy, Cancer Genetics Laboratory, Gustave Roussy, 94800, Villejuif, France
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Tracy L Stockley
- Laboratory Medicine Program, University Health Network; Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Zandra C Deans
- GenQA, Department of Laboratory Medicine, NHS Lothian, Nine Bioquarter, Little France Rd, Edinburgh, EH16 4UX, UK.
| |
Collapse
|
25
|
Chen C, Douglas MP, Ragavan MV, Phillips KA, Jansen JP. Clinical Validity and Utility of Circulating Tumor DNA (ctDNA) Testing in Advanced Non-small Cell Lung Cancer (aNSCLC): A Systematic Literature Review and Meta-analysis. Mol Diagn Ther 2024; 28:525-536. [PMID: 39093546 PMCID: PMC11349784 DOI: 10.1007/s40291-024-00725-x] [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: 06/18/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE Circulating tumor DNA (ctDNA) testing has become a promising tool to guide first-line (1L) targeted treatment for advanced non-small cell lung cancer (aNSCLC). This study aims to estimate the clinical validity (CV) and clinical utility (CU) of ctDNA-based next-generation sequencing (NGS) for oncogenic driver mutations to inform 1L treatment decisions in aNSCLC through a systematic literature review and meta-analysis. METHODS A systematic literature search was conducted in PubMed/MEDLINE and Embase to identify randomized control trials or observational studies reporting CV/CU on ctDNA testing in patients with aNSCLC. Meta-analyses were performed using bivariate random-effects models to estimate pooled sensitivity and specificity. Progression-free/overall survival (PFS/OS) was summarized for CU studies. RESULTS A total of 20 studies were identified: 17 CV only, 2 CU only, and 1 both, and 13 studies were included for the meta-analysis on multi-gene detection. The overall sensitivity and specificity for ctDNA detection of any mutation were 0.69 (95% CI 0.63-0.74) and 0.99 (95% CI 0.97-1.00), respectively. However, sensitivity varied greatly by driver gene, ranging from 0.29 (95% CI 0.13-0.53) for ROS1 to 0.77 (95% CI 0.63-0.86) for KRAS. Two studies that compared PFS with ctDNA versus tissue-based testing followed by 1L targeted therapy found no significant differences. One study reported OS curves on ctDNA-matched and tissue-matched therapies but no hazard ratios were provided. CONCLUSIONS ctDNA testing demonstrated an overall acceptable diagnostic accuracy in patients with aNSCLC, however, sensitivity varied greatly by driver mutation. Further research is needed, especially for uncommon driver mutations, to better understand the CU of ctDNA testing in guiding targeted treatments for aNSCLC.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
| | - Michael P Douglas
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
| | - Meera V Ragavan
- Division of Hematology and Oncology, UCSF Department of Medicine, San Francisco, CA, USA
| | - Kathryn A Phillips
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
- UCSF Philip R. Lee Institute for Health Policy, San Francisco, CA, USA
| | - Jeroen P Jansen
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA.
- UCSF Philip R. Lee Institute for Health Policy, San Francisco, CA, USA.
- Department of Clinical Pharmacy, School of Pharmacy, University of California San Francisco, 490 Illinois St. Valley Tower, 3rd Floor, Box 0613, San Francisco, CA, 94143, USA.
| |
Collapse
|
26
|
HOU Y, ZHANG T, WANG H. [Advancements in Radiomics for Immunotherapy of Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:637-644. [PMID: 39318257 PMCID: PMC11425675 DOI: 10.3779/j.issn.1009-3419.2024.102.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Indexed: 09/26/2024]
Abstract
Lung cancer is the main cause of cancer-related deaths, with non-small cell lung cancer (NSCLC) being the predominant subtype. At present, immunotherapy represented by immune checkpoint inhibitors (ICIs) of programmed cell death receptor 1 or its ligand has been widely used in the clinical diagnosis and treatment of patients with NSCLC. However, only a few patients can benefit from it, and reliable predictive markers for immunotherapy are lacking. Radiomics is a tool that uses computer software and algorithms to extract a large amount of quantitative information from biomedical images. A large number of studies have confirmed that the radiomic model that predicts the immune efficacy of NSCLC can be used as a new type of immune efficacy predictive marker, which is expected to guide the individualized diagnosis and treatment decisions for patients with lung cancer and has a bright application prospect. This article reviews the research progress of radiomics in predicting the immune therapy response of NSCLC, identifying pseudo-progression and hyperprogression, ICIs-related pneumonia, cachexia risk, and combining with other genomics.
.
Collapse
|
27
|
Rina A, Maffeo D, Minnai F, Esposito M, Palmieri M, Serio VB, Rosati D, Mari F, Frullanti E, Colombo F. The Genetic Analysis and Clinical Therapy in Lung Cancer: Current Advances and Future Directions. Cancers (Basel) 2024; 16:2882. [PMID: 39199653 PMCID: PMC11352260 DOI: 10.3390/cancers16162882] [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: 07/30/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Lung cancer, including both non-small cell lung cancer and small cell lung cancer, remains the leading cause of cancer-related mortality worldwide, representing 18% of the total cancer deaths in 2020. Many patients are identified already at an advanced stage with metastatic disease and have a worsening prognosis. Recent advances in the genetic understanding of lung cancer have opened new avenues for personalized treatments and targeted therapies. This review examines the latest discoveries in the genetics of lung cancer, discusses key biomarkers, and analyzes current clinical therapies based on this genetic information. It will conclude with a discussion of future prospects and potential research directions.
Collapse
Affiliation(s)
- Angela Rina
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- UOC Laboratorio di Assistenza e Ricerca Traslazionale, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
| | - Debora Maffeo
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Minnai
- Institute of Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (F.M.); (M.E.)
| | - Martina Esposito
- Institute of Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (F.M.); (M.E.)
| | - Maria Palmieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Viola Bianca Serio
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Diletta Rosati
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Mari
- UOC Laboratorio di Assistenza e Ricerca Traslazionale, Azienda Ospedaliero-Universitaria Senese, 53100 Siena, Italy;
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (A.R.); (D.M.); (M.P.); (V.B.S.); (D.R.); (E.F.)
- Cancer Genomics and Systems Biology Laboratory, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Colombo
- Institute of Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (F.M.); (M.E.)
| |
Collapse
|
28
|
Li K, Zhu Q, Yang J, Zheng Y, Du S, Song M, Peng Q, Yang R, Liu Y, Qi L. Imaging and Liquid Biopsy for Distinguishing True Progression From Pseudoprogression in Gliomas, Current Advances and Challenges. Acad Radiol 2024; 31:3366-3383. [PMID: 38614827 DOI: 10.1016/j.acra.2024.03.019] [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: 12/10/2023] [Revised: 01/14/2024] [Accepted: 03/18/2024] [Indexed: 04/15/2024]
Abstract
RATIONALE AND OBJECTIVES Gliomas are aggressive brain tumors with a poor prognosis. Assessing treatment response is challenging because magnetic resonance imaging (MRI) may not distinguish true progression (TP) from pseudoprogression (PsP). This review aims to discuss imaging techniques and liquid biopsies used to distinguish TP from PsP. MATERIALS AND METHODS This review synthesizes existing literature to examine advances in imaging techniques, such as magnetic resonance diffusion imaging (MRDI), perfusion-weighted imaging (PWI) MRI, and liquid biopsies, for identifying TP or PsP through tumor markers and tissue characteristics. RESULTS Advanced imaging techniques, including MRDI and PWI MRI, have proven effective in delineating tumor tissue properties, offering valuable insights into glioma behavior. Similarly, liquid biopsy has emerged as a potent tool for identifying tumor-derived markers in biofluids, offering a non-invasive glimpse into tumor evolution. Despite their promise, these methodologies grapple with significant challenges. Their sensitivity remains inconsistent, complicating the accurate differentiation between TP and PSP. Furthermore, the absence of standardized protocols across platforms impedes the reliability of comparisons, while inherent biological variability adds complexity to data interpretation. CONCLUSION Their potential applications have been highlighted, but gaps remain before routine clinical use. Further research is needed to develop and validate these promising methods for distinguishing TP from PsP in gliomas.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China; Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), 1# Jiazi Road, Foshan, Guangdong 528300, China.; Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qihui Zhu
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Junyi Yang
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Yin Zheng
- Department of Neurosurgery, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Siyuan Du
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Meihui Song
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Qian Peng
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China
| | - Runwei Yang
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), 1# Jiazi Road, Foshan, Guangdong 528300, China
| | - Yawei Liu
- Department of Neurosurgery & Medical Research Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), 1# Jiazi Road, Foshan, Guangdong 528300, China
| | - Ling Qi
- Institute of Digestive Disease of Guangzhou Medical University, Affiliated Qingyuan Hospital,Guangzhou Medical University,Qingyuan People's Hospital, Qingyuan 511518, China.
| |
Collapse
|
29
|
Megquier K, Husted C, Rhoades J, White ME, Genereux DP, Chen FL, Xiong K, Kwon E, Swofford R, Painter C, Adalsteinsson V, London CA, Gardner HL, Karlsson EK. Impact of preanalytical factors on liquid biopsy in the canine cancer model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605605. [PMID: 39131379 PMCID: PMC11312437 DOI: 10.1101/2024.07.29.605605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
While liquid biopsy has potential to transform cancer diagnostics through minimally-invasive detection and monitoring of tumors, the impact of preanalytical factors such as the timing and anatomical location of blood draw is not well understood. To address this gap, we leveraged pet dogs with spontaneous cancer as a model system, as their compressed disease timeline facilitates rapid diagnostic benchmarking. Key liquid biopsy metrics from dogs were consistent with existing reports from human patients. The tumor content of samples was higher from venipuncture sites closer to the tumor and from a central vein. Metrics also differed between lymphoma and non-hematopoietic cancers, urging cancer-type-specific interpretation. Liquid biopsy was highly sensitive to disease status, with changes identified soon after post chemotherapy administration, and trends of increased tumor fraction and other metrics observed prior to clinical relapse in dogs with lymphoma or osteosarcoma. These data support the utility of pet dogs with cancer as a relevant system for advancing liquid biopsy platforms.
Collapse
Affiliation(s)
- Kate Megquier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christopher Husted
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genomics and Computational Biology, UMass Chan Medical School, Worcester, MA, USA
- Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, USA
| | | | | | | | - Frances L. Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genomics and Computational Biology, UMass Chan Medical School, Worcester, MA, USA
| | - Kan Xiong
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Euijin Kwon
- Genomics and Computational Biology, UMass Chan Medical School, Worcester, MA, USA
- Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA, USA
| | - Ross Swofford
- Genomics and Computational Biology, UMass Chan Medical School, Worcester, MA, USA
| | | | | | - Cheryl A. London
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, USA
| | | | - Elinor K. Karlsson
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genomics and Computational Biology, UMass Chan Medical School, Worcester, MA, USA
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
30
|
Zhu S, Wu R, Liu X, Xie B, Xie C, Li S, Wu Z, Zhang Z, Tang Z, Gu L. Clinical application of ctDNA in early diagnosis, treatment and prognosis of patients with non-small cell lung cancer. Future Oncol 2024; 20:2213-2224. [PMID: 39073412 PMCID: PMC11514542 DOI: 10.1080/14796694.2024.2376513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Lung cancer is one of the most common malignancies worldwide, with non-small cell lung cancer (NSCLC) being the most common type. As understanding of precise treatment options for NSCLC deepens, circulating tumor DNA (ctDNA) has emerged as a potential biomarker that has become a research hotspot and may represent a new approach for the individualized diagnosis and treatment of NSCLC. This article reviews the applications of ctDNA for the early screening of patients with NSCLC, guiding targeted therapy and immunotherapy, evaluating chemotherapy and postoperative efficacy, assessing prognosis and monitoring recurrence. With the in-depth study of the pathogenesis of NSCLC, plasma ctDNA may become an indispensable part of the precise treatment of NSCLC, which has great clinical application prospects.
Collapse
Affiliation(s)
- Shenyu Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Rongqian Wu
- Department of Endocrinology and Metabolism, Gaoxin Hospital of The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiangjin Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Bin Xie
- First Clinical Medical College, The Gannan Medical University, Ganzhou, China
| | - Chunfa Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Shulin Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Zhicheng Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Zuxiong Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Zhixian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Liang Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| |
Collapse
|
31
|
Dang Q, Zuo L, Hu X, Zhou Z, Chen S, Liu S, Ba Y, Zuo A, Xu H, Weng S, Zhang Y, Luo P, Cheng Q, Liu Z, Han X. Molecular subtypes of colorectal cancer in the era of precision oncotherapy: Current inspirations and future challenges. Cancer Med 2024; 13:e70041. [PMID: 39054866 PMCID: PMC11272957 DOI: 10.1002/cam4.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most hackneyed malignancies. Even patients with identical clinical symptoms and the same TNM stage still exhibit radically different clinical outcomes after receiving equivalent treatment regimens, indicating extensive heterogeneity of CRC. Myriad molecular subtypes of CRC have been exploited for decades, including the most compelling consensus molecular subtype (CMS) classification that has been broadly applied for patient stratification and biomarker-drug combination formulation. Encountering barriers to clinical translation, however, CMS classification fails to fully reflect inter- or intra-tumor heterogeneity of CRC. As a consequence, addressing heterogeneity and precisely managing CRC patients with unique characteristics remain arduous tasks for clinicians. REVIEW In this review, we systematically summarize molecular subtypes of CRC and further elaborate on their clinical applications, limitations, and future orientations. CONCLUSION In recent years, exploration of subtypes through cell lines, animal models, patient-derived xenografts (PDXs), organoids, and clinical trials contributes to refining biological insights and unraveling subtype-specific therapies in CRC. Therapeutic interventions including nanotechnology, clustered regulatory interspaced short palindromic repeat/CRISPR-associated nuclease 9 (CRISPR/Cas9), gut microbiome, and liquid biopsy are powerful tools with the possibility to shift the immunologic landscape and outlook for CRC precise medicine.
Collapse
Affiliation(s)
- Qin Dang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Lulu Zuo
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xinru Hu
- Department of Cardiology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhaokai Zhou
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuang Chen
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Peng Luo
- Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Quan Cheng
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
32
|
Zhu L, Zhang L, Qi J, Ye Z, Nie G, Leng S. Machine learning-derived immunosenescence index for predicting outcome and drug sensitivity in patients with skin cutaneous melanoma. Genes Immun 2024; 25:219-231. [PMID: 38811681 DOI: 10.1038/s41435-024-00278-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
The functions of immunosenescence are closely related to skin cutaneous melanoma (SKCM). The aim of this study is to uncover the characteristics of immunosenescence index (ISI) to identify novel biomarkers and potential targets for treatment. Firstly, integrated bioinformatics analysis was carried out to identify risk prognostic genes, and their expression and prognostic value were evaluated. Then, we used the computational algorithm to estimate ISI. Finally, the distribution characteristics and clinical significance of ISI in SKCM by using multi-omics analysis. Patients with a lower ISI had a favorable survival rate, lower chromosomal instability, lower somatic copy-number alterations, lower somatic mutations, higher immune infiltration, and sensitive to immunotherapy. The ISI exhibited robust, which was validated in multiple datasets. Besides, the ISI is more effective than other published signatures in predicting survival outcomes for patients with SKCM. Single-cell analysis revealed higher ISI was specifically expressed in monocytes, and correlates with the differentiation fate of monocytes in SKCM. Besides, individuals exhibiting elevated ISI levels could potentially receive advantages from chemotherapy, and promising compounds with the potential to target high ISI were recognized. The ISI model is a valuable tool in categorizing SKCM patients based on their prognosis, gene mutation signatures, and response to immunotherapy.
Collapse
Affiliation(s)
- Linyu Zhu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Lvya Zhang
- Traditional Chinese Medicine department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Junhua Qi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Department of Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhiyu Ye
- Traditional Chinese Medicine department, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Gang Nie
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
33
|
Liu H, Chen J, Huang Y, Zhang Y, Ni Y, Xu N, Zhao F, Tang Y, Liu H, Sun G, Shen P, Liu Z, Huang J, Liao B, Zeng H. Prognostic significance of circulating tumor DNA in urothelial carcinoma: a systematic review and meta-analysis. Int J Surg 2024; 110:3923-3936. [PMID: 38573063 PMCID: PMC11175790 DOI: 10.1097/js9.0000000000001372] [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: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) has emerged as a noninvasive technique that provides valuable insights into molecular profiles and tumor disease management. This study aimed to evaluate the prognostic significance of circulating tumor DNA (ctDNA) in urothelial carcinoma (UC) through a systematic review and meta-analysis. METHODS A comprehensive search was conducted in MEDLINE, EMBASE, and the Cochrane Library from the inception to December 2023. Studies investigating the prognostic value of ctDNA in UC were included. Hazard ratios (HRs) of disease-free survival (DFS) and overall survival (OS) were extracted. Overall meta-analysis and subgroup exploration stratified by metastatic status, ctDNA sampling time, treatment type, and detection method was performed using the R software (version 4.2.2). RESULTS A total of 16 studies with 1725 patients were included. Fourteen studies assessed the association between baseline ctDNA status and patient outcomes. Patients with elevated ctDNA levels exhibited significantly worse DFS (HR=6.26; 95% CI: 3.71-10.58, P <0.001) and OS (HR=4.23; 95% CI: 2.72-6.57, P <0.001) regardless of metastatic status, ctDNA sampling time, treatment type, and detection methods. Six studies evaluated the prognostic value of ctDNA dynamics in UC. Patients who showed a decrease or clearance in ctDNA levels during treatment or observation demonstrated more favorable DFS (HR=0.26, 95% CI: 0.17-0.41, P <0.001) and OS (HR=0.21, 95% CI: 0.11-0.38, P <0.001) compared to those who did not. The association remained consistent across the subgroup analysis based on metastatic status and detection methods. In the immune checkpoint inhibitor-treated setting, both lower baseline ctDNA level and ctDNA decrease during the treatment were significantly associated with more favorable oncologic outcomes. Furthermore, specific gene mutations such as FGFR3 identified in ctDNA also demonstrated predictive value in UC patients. CONCLUSION This meta-analysis demonstrates a strong association of ctDNA status and its dynamic change with survival outcomes in UC, suggesting substantial clinical utility of ctDNA testing in prognosis prediction and decision making in this setting.
Collapse
Affiliation(s)
- Haoyang Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yuchen Huang
- Department of Cardiothoracic Surgery, West China Hospital, Sichuan University
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yuchao Ni
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Nanwei Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Fengnian Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Yanfeng Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Haolin Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Jin Huang
- Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Banghua Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University
| |
Collapse
|
34
|
Wang H, Zhang Y, Zhang H, Cao H, Mao J, Chen X, Wang L, Zhang N, Luo P, Xue J, Qi X, Dong X, Liu G, Cheng Q. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm (Beijing) 2024; 5:e564. [PMID: 38807975 PMCID: PMC11130638 DOI: 10.1002/mco2.564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Currently, tumor treatment modalities such as immunotherapy and targeted therapy have more stringent requirements for obtaining tumor growth information and require more accurate and easy-to-operate tumor information detection methods. Compared with traditional tissue biopsy, liquid biopsy is a novel, minimally invasive, real-time detection tool for detecting information directly or indirectly released by tumors in human body fluids, which is more suitable for the requirements of new tumor treatment modalities. Liquid biopsy has not been widely used in clinical practice, and there are fewer reviews of related clinical applications. This review summarizes the clinical applications of liquid biopsy components (e.g., circulating tumor cells, circulating tumor DNA, extracellular vesicles, etc.) in tumorigenesis and progression. This includes the development process and detection techniques of liquid biopsies, early screening of tumors, tumor growth detection, and guiding therapeutic strategies (liquid biopsy-based personalized medicine and prediction of treatment response). Finally, the current challenges and future directions for clinical applications of liquid biopsy are proposed. In sum, this review will inspire more researchers to use liquid biopsy technology to promote the realization of individualized therapy, improve the efficacy of tumor therapy, and provide better therapeutic options for tumor patients.
Collapse
Affiliation(s)
- Hao Wang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Yi Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Hao Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Hui Cao
- Department of PsychiatryThe School of Clinical Medicine, Hunan University of Chinese MedicineChangshaChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province)ChangshaChina
| | - Jinning Mao
- Health Management CenterThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xinxin Chen
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Liangchi Wang
- Department of NeurosurgeryFengdu People's Hospital, ChongqingChongqingChina
| | - Nan Zhang
- College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Peng Luo
- Department of OncologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Ji Xue
- Department of NeurosurgeryTraditional Chinese Medicine Hospital Dianjiang ChongqingChongqingChina
| | - Xiaoya Qi
- Health Management CenterThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xiancheng Dong
- Department of Cerebrovascular DiseasesDazhou Central HospitalSichuanChina
| | - Guodong Liu
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Quan Cheng
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
35
|
O'Sullivan NJ, Temperley HC, Kyle ET, Sweeney KJ, O'Neill M, Gilham C, O'Sullivan J, O'Kane G, Mehigan B, O'Toole S, Larkin J, Gallagher D, McCormick P, Kelly ME. Assessing circulating tumour DNA (ctDNA) as a prognostic biomarker in locally advanced rectal cancer: a systematic review and meta-analysis. Int J Colorectal Dis 2024; 39:82. [PMID: 38809315 PMCID: PMC11136793 DOI: 10.1007/s00384-024-04656-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION Circulating tumour DNA (ctDNA) has emerged as a promising biomarker in various cancer types, including locally advanced rectal cancer (LARC), offering potential insights into disease progression, treatment response and recurrence. This review aims to comprehensively evaluate the utility of ctDNA as a prognostic biomarker in LARC. METHODS PubMed, EMBASE and Web of Science were searched as part of our review. Studies investigating the utility of ctDNA in locally advanced rectal cancer (LARC) were assessed for eligibility. Quality assessment of included studies was performed using the Newcastle Ottawa Scale (NOS) risk of bias tool. Outcomes extracted included basic participant characteristics, ctDNA details and survival data. A meta-analysis was performed on eligible studies to determine pooled recurrence-free survival (RFS). RESULTS Twenty-two studies involving 1676 participants were included in our analysis. Methodological quality categorised by the Newcastle Ottawa Scale was generally satisfactory across included studies. ctDNA detected at various time intervals was generally associated with poor outcomes across included studies. Meta-analysis demonstrated a pooled hazard ratio of 8.87 (95% CI 4.91-16.03) and 15.15 (95% CI 8.21-27.95), indicating an increased risk of recurrence with ctDNA positivity in the post-neoadjuvant and post-operative periods respectively. CONCLUSION Our systematic review provides evidence supporting the prognostic utility of ctDNA in patients with LARC, particularly in identifying patients at higher risk of disease recurrence in the post-neoadjuvant and post-operative periods.
Collapse
Affiliation(s)
- Niall J O'Sullivan
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland.
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Hugo C Temperley
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Eimear T Kyle
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
| | - Kevin J Sweeney
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
| | - Maeve O'Neill
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
| | - Charles Gilham
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- Department of Radiation Oncology, St. James's Hospital, Dublin 8, Ireland
| | - Jacintha O'Sullivan
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- Trinity Translational Medicine Institute, Trinity St. James's Cancer Institute, Trinity College, St. James's Hospital, Dublin, Ireland
| | - Grainne O'Kane
- Department of Medical Oncology, St. James's Hospital, Dublin 8, Ireland
| | - Brian Mehigan
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
| | - Sharon O'Toole
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- Trinity Translational Medicine Institute, Trinity St. James's Cancer Institute, Trinity College, St. James's Hospital, Dublin, Ireland
| | - John Larkin
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
| | - David Gallagher
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- Department of Medical Oncology, St. James's Hospital, Dublin 8, Ireland
- Department of Genetics, St. James's Hospital, Dublin 8, Ireland
| | - Paul McCormick
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
| | - Michael E Kelly
- Department of Surgery, St. James's Hospital, Dublin 8, Ireland
- School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Dublin 8, Ireland
| |
Collapse
|
36
|
Stibbards-Lyle M, Malinovska J, Badawy S, Schedin P, Rinker KD. Status of breast cancer detection in young women and potential of liquid biopsy. Front Oncol 2024; 14:1398196. [PMID: 38835377 PMCID: PMC11148378 DOI: 10.3389/fonc.2024.1398196] [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: 03/11/2024] [Accepted: 05/01/2024] [Indexed: 06/06/2024] Open
Abstract
Young onset breast cancer (YOBC) is an increasing demographic with unique biology, limited screening, and poor outcomes. Further, women with postpartum breast cancers (PPBCs), cancers occurring up to 10 years after childbirth, have worse outcomes than other young breast cancer patients matched for tumor stage and subtype. Early-stage detection of YOBC is critical for improving outcomes. However, most young women (under 45) do not meet current age guidelines for routine mammographic screening and are thus an underserved population. Other challenges to early detection in this population include reduced performance of standard of care mammography and reduced awareness. Women often face significant barriers in accessing health care during the postpartum period and disadvantaged communities face compounding barriers due to systemic health care inequities. Blood tests and liquid biopsies targeting early detection may provide an attractive option to help address these challenges. Test development in this area includes understanding of the unique biology involved in YOBC and in particular PPBCs that tend to be more aggressive and deadly. In this review, we will present the status of breast cancer screening and detection in young women, provide a summary of some unique biological features of YOBC, and discuss the potential for blood tests and liquid biopsy platforms to address current shortcomings in timely, equitable detection.
Collapse
Affiliation(s)
- Maya Stibbards-Lyle
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
| | - Julia Malinovska
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
| | - Seleem Badawy
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
| | - Pepper Schedin
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Kristina D Rinker
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
- Cellular and Molecular Bioengineering Research Lab, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
37
|
Kött J, Zimmermann N, Zell T, Rünger A, Heidrich I, Geidel G, Smit DJ, Hansen I, Abeck F, Schadendorf D, Eggermont A, Puig S, Hauschild A, Gebhardt C. Sentinel lymph node risk prognostication in primary cutaneous melanoma through tissue-based profiling, potentially redefining the need for sentinel lymph node biopsy. Eur J Cancer 2024; 202:113989. [PMID: 38518535 DOI: 10.1016/j.ejca.2024.113989] [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] [Accepted: 03/04/2024] [Indexed: 03/24/2024]
Abstract
PURPOSE OF REVIEW The role of Sentinel Lymph Node Biopsy (SLNB) is pivotal in the contemporary staging of cutaneous melanoma. In this review, we examine advanced molecular testing platforms like gene expression profiling (GEP) and immunohistochemistry (IHC) as tools for predicting the prognosis of sentinel lymph nodes. We compare these innovative approaches with traditional staging assessments. Additionally, we delve into the shared genetic and protein markers between GEP and IHC tests and their relevance to melanoma biology, exploring their prognostic and predictive characteristics. Finally, we assess alternative methods to potentially obviate the need for SLNB altogether. RECENT FINDINGS Progress in adjuvant melanoma therapy has diminished the necessity of Sentinel Lymph Node Biopsy (SLNB) while underscoring the importance of accurately identifying high-risk stage I and II melanoma patients who may benefit from additional anti-tumor interventions. The clinical application of testing through gene expression profiling (GEP) or immunohistochemistry (IHC) is gaining traction, with platforms such as DecisionDx, Merlin Assay (CP-GEP), MelaGenix GEP, and Immunoprint coming into play. Currently, extensive validation studies are in progress to incorporate routine molecular testing into clinical practice. However, due to significant methodological limitations, widespread clinical adoption of tissue-based molecular testing remains elusive at present. SUMMARY While various tissue-based molecular testing platforms have the potential to stratify the risk of sentinel lymph node positivity (SLNP), most suffer from significant methodological deficiencies, including limited sample size, lack of prospective validation, and limited correlation with established clinicopathological variables. Furthermore, the genes and proteins identified by individual gene expression profiling (GEP) or immunohistochemistry (IHC) tests exhibit minimal overlap, even when considering the most well-established melanoma mutations. However, there is hope that the ongoing prospective trial for the Merlin Assay may safely reduce the necessity for SLNB procedures if successful. Additionally, the MelaGenix GEP and Immunoprint tests could prove valuable in identifying high-risk stage I-II melanoma patients and potentially guiding their selection for adjuvant therapy, thus potentially reducing the need for SLNB. Due to the diverse study designs employed, effective comparisons between GEP or IHC tests are challenging, and to date, there is no study directly comparing the clinical utility of these respective GEP or IHC tests.
Collapse
Affiliation(s)
- Julian Kött
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Noah Zimmermann
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Tim Zell
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Alessandra Rünger
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Isabel Heidrich
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Glenn Geidel
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Daniel J Smit
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Inga Hansen
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Finn Abeck
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Dirk Schadendorf
- Department of Dermatology & Westdeutsches Tumorzentrum Essen (WTZ), University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany; National Center for Tumor Diseases (NCT-West), Campus Essen, Germany; Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Alexander Eggermont
- Princess Máxima Center and University Medical Center Utrecht, 3584 CS Utrecht, the Netherlands; Comprehensive Cancer Center Munich, Technical University Munich & Ludwig Maximilian University, Munich, Germany
| | - Susana Puig
- Department of Dermatology, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain; Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Christoffer Gebhardt
- University Skin Cancer Center Hamburg, Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| |
Collapse
|
38
|
Sei S, Srivastava S, Kelly HR, Miller MS, Leitner WW, Shoemaker RH, Szabo E, Castle PE. NCI Resources for Cancer Immunoprevention Research. Cancer Immunol Res 2024; 12:387-392. [PMID: 38562082 DOI: 10.1158/2326-6066.cir-23-0708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/29/2023] [Accepted: 01/31/2024] [Indexed: 02/03/2024]
Abstract
Cancer prevention and early detection, the first two of the eight primary goals of the National Cancer Plan released in April 2023, are at the forefront of the nation's strategic efforts to reduce cancer incidence and mortality. The Division of Cancer Prevention (DCP) of the NCI is the federal government's principal component devoted to promoting and supporting innovative cancer prevention research. Recent advances in tumor immunology, cancer immunotherapy, and vaccinology strongly suggest that the host immune system can be effectively harnessed to elicit protective immunity against the development of cancer, that is, cancer immunoprevention. Cancer immunoprevention may be most effective if the intervention is given before or early in the carcinogenic process while the immune system remains relatively uncompromised. DCP has increased the emphasis on immunoprevention research in recent years and continues to expand program resources and interagency collaborations designed to facilitate research in the immunoprevention field. These resources support a wide array of basic, translational, and clinical research activities, including discovery, development, and validation of biomarkers for cancer risk assessment and early detection (Early Detection Research Network), elucidation of biological and pathophysiological mechanistic determinants of precancer growth and its control (Translational and Basic Science Research in Early Lesions), spatiotemporal multiomics characterization of precancerous lesions (Human Tumor Atlas Network/Pre-Cancer Atlas), discovery of immunoprevention pathways and immune targets (Cancer Immunoprevention Network), and preclinical and clinical development of novel agents for immunoprevention and interception (Cancer Prevention-Interception Targeted Agent Discovery Program, PREVENT Cancer Preclinical Drug Development Program, and Cancer Prevention Clinical Trials Network).
Collapse
Affiliation(s)
- Shizuko Sei
- Division of Cancer Prevention, NCI, NIH, Bethesda, Maryland
| | | | - Halonna R Kelly
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Wolfgang W Leitner
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Eva Szabo
- Division of Cancer Prevention, NCI, NIH, Bethesda, Maryland
| | - Philip E Castle
- Division of Cancer Prevention, NCI, NIH, Bethesda, Maryland
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland
| |
Collapse
|
39
|
Egger ME, Alexander E, Van Meter T, Kong M, Maung AA, Valdes R, Hall MB, Linder MW. Corresponding ctDNA and tumor burden dynamics in metastatic melanoma patients on systemic treatment. Transl Oncol 2024; 42:101883. [PMID: 38306914 PMCID: PMC10850110 DOI: 10.1016/j.tranon.2024.101883] [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: 04/13/2023] [Revised: 12/15/2023] [Accepted: 01/11/2024] [Indexed: 02/04/2024] Open
Abstract
Radiographic imaging is the current standard for monitoring progression of tumor-burden and therapeutic resistance in patients with metastatic melanoma. Plasma circulating tumor DNA (ctDNA) has shown promise as a survelience tool, but longitudinal data on the dynamics between plasma ctDNA concentrations and radiographic imaging is lacking. We evaluated the relationship between longitudinal radiographic measures of tumor burden and ctDNA concentrations in plasma on 30 patients with metastatic melanoma on systemic treatment. In 9 patients with no radiographic evidence of disease over a total of 15 time points, ctDNA concentrations were undetectable. In 21 patients with radiographic tumor burden, ctDNA was detected in 81 % of 58 time points. Plasma ctDNA concentrations demonstrated a modest positive correlation with total tumor burden (TTB) measurements (R2= 0.49, p < 0.001), with the greatest degree of correlation observed under conditions of progressive disease (PD) (R2 = 0.91, p = 0.032). Plasma ctDNA concentrations were significantly greater at times of RECIST v1.1 progression (PD; 22.1 % ± 5.7 %) when compared to samples collected during stable disease (SD; 4.99 % ± 3.0 %) (p = 0.012); this difference was independent of total tumor burden (p = 0.997). Changes in plasma ctDNA showed a strong correlation with changes in TTB (R2= 0.88, p<0.001). These data suggest that measurements of plasma ctDNA during therapy are a better surrogate for responding versus non-responding disease compared to absolute tumor burden.
Collapse
Affiliation(s)
- Michael E Egger
- Department of Surgery, Division of Surgical Oncology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Evan Alexander
- Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Tracy Van Meter
- Department of Radiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Maiying Kong
- Biostatistics and Bioinformatics Facility, James Graham Brown Cancer Center, Louisville, KY, USA; Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Aye Aye Maung
- Biostatistics and Bioinformatics Facility, James Graham Brown Cancer Center, Louisville, KY, USA; Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Roland Valdes
- Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Melissa Barousse Hall
- UofL Health Brown Cancer Center, Louisville, KY, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Mark W Linder
- Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, Louisville, KY, USA; UofL Health Brown Cancer Center, Louisville, KY, USA.
| |
Collapse
|
40
|
Díaz del Arco C, Fernández Aceñero MJ, Ortega Medina L. Liquid biopsy for gastric cancer: Techniques, applications, and future directions. World J Gastroenterol 2024; 30:1680-1705. [PMID: 38617733 PMCID: PMC11008373 DOI: 10.3748/wjg.v30.i12.1680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
After the study of circulating tumor cells in blood through liquid biopsy (LB), this technique has evolved to encompass the analysis of multiple materials originating from the tumor, such as nucleic acids, extracellular vesicles, tumor-educated platelets, and other metabolites. Additionally, research has extended to include the examination of samples other than blood or plasma, such as saliva, gastric juice, urine, or stool. LB techniques are diverse, intricate, and variable. They must be highly sensitive, and pre-analytical, patient, and tumor-related factors significantly influence the detection threshold, diagnostic method selection, and potential results. Consequently, the implementation of LB in clinical practice still faces several challenges. The potential applications of LB range from early cancer detection to guiding targeted therapy or immunotherapy in both early and advanced cancer cases, monitoring treatment response, early identification of relapses, or assessing patient risk. On the other hand, gastric cancer (GC) is a disease often diagnosed at advanced stages. Despite recent advances in molecular understanding, the currently available treatment options have not substantially improved the prognosis for many of these patients. The application of LB in GC could be highly valuable as a non-invasive method for early diagnosis and for enhancing the management and outcomes of these patients. In this comprehensive review, from a pathologist's perspective, we provide an overview of the main options available in LB, delve into the fundamental principles of the most studied techniques, explore the potential utility of LB application in the context of GC, and address the obstacles that need to be overcome in the future to make this innovative technique a game-changer in cancer diagnosis and treatment within clinical practice.
Collapse
Affiliation(s)
- Cristina Díaz del Arco
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - M Jesús Fernández Aceñero
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Luis Ortega Medina
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| |
Collapse
|
41
|
Fietz S, Diekmann E, de Vos L, Zarbl R, Hunecke A, Glosch AK, Färber M, Sirokay J, Hoffmann F, Fröhlich A, Franzen A, Strieth S, Landsberg J, Dietrich D. Circulating Cell-Free SHOX2 DNA Methylation Is a Predictive, Prognostic, and Monitoring Biomarker in Adjuvant and Palliative Anti-PD-1-Treated Melanoma. Clin Chem 2024; 70:516-527. [PMID: 38300881 DOI: 10.1093/clinchem/hvad230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND The majority of metastatic melanoma patients initially do not respond or acquire resistance to anti-programmed cell death 1 (PD-1) immunotherapy. Liquid biopsy biomarkers might provide useful early response information and allow for personalized treatment decisions. METHODS We prospectively assessed circulating cell-free SHOX2 DNA methylation (SHOX2 ccfDNAm) levels and their dynamic changes in blood plasma of melanoma patients by quantitative methylation-specific polymerase chain reaction. Patients were treated with either palliative (n = 42) or adjuvant (n = 55) anti-PD-1 immunotherapy. Moreover, we included n = 126 control patients without evidence of malignant disease. We analyzed SHOX2 ccfDNAm status prior to and 4 weeks after palliative treatment initiation with regard to outcome [objective response, progression-free survival (PFS), and overall survival (OS)]. In the adjuvant setting, we associated longitudinal SHOX2 ccfDNAm status with disease recurrence. RESULTS Sensitivity was 60% with 25/42 melanoma patients showing increased SHOX2 ccfDNAm levels, whereas specificity was 98% with 123/126 (P < 0.001) control patients having SHOX2 ccfDNAm levels below cut-off. Pretreatment SHOX2 ccfDNAm status did not correlate with outcome; however, SHOX2 ccfDNAm negativity 4 weeks after palliative treatment initiation was strongly associated with improved survival [PFS: hazard ratio (HR) = 0.25, P = 0.002; OS: HR = 0.12, P = 0.007]. Pretreatment positive patients who reached SHOX2 ccfDNAm clearance after 4 weeks of immunotherapy showed an exceptionally beneficial outcome. SHOX2 ccfDNAm testing allowed for an early detection of distant metastases in adjuvant-treated melanoma patients. CONCLUSIONS Our study suggests SHOX2 ccfDNAm to be an early predictor of outcome in anti-PD-1 treated melanoma patients. SHOX2 ccfDNAm testing may aid individualized treatment decision-making.
Collapse
Affiliation(s)
- Simon Fietz
- Department of Dermatology and Allergology, University Hospital Bonn, Bonn, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Eric Diekmann
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Luka de Vos
- Department of Dermatology and Allergology, University Hospital Bonn, Bonn, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Romina Zarbl
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Alina Hunecke
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Ann-Kathrin Glosch
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Moritz Färber
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Judith Sirokay
- Department of Dermatology and Allergology, University Hospital Bonn, Bonn, Germany
| | - Friederike Hoffmann
- Department of Dermatology and Allergology, University Hospital Bonn, Bonn, Germany
| | - Anne Fröhlich
- Department of Dermatology and Allergology, University Hospital Bonn, Bonn, Germany
| | - Alina Franzen
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Sebastian Strieth
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| | - Jennifer Landsberg
- Department of Dermatology and Allergology, University Hospital Bonn, Bonn, Germany
| | - Dimo Dietrich
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
42
|
Homburg S, Christensen CB, Pedersen M, Sørensen SG, Donia M, Svane IM, Hendel HW, Ellebaek E. Prospective Assessment of Fluorine-18-Fluorodeoxyglucose-Positron Emission Tomography/Computed Tomography (FDG-PET/CT) for Early Identification of Checkpoint-Inhibitor-Induced Pseudoprogression. Cancers (Basel) 2024; 16:964. [PMID: 38473325 DOI: 10.3390/cancers16050964] [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: 02/03/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The activity of immune checkpoint inhibitors (ICIs) in patients with metastatic melanoma is often monitored using fluorine-18-fluorodeoxyglucose-positron emission tomography/computed tomography (FDG-PET/CT) scans. However, distinguishing disease progression (PD) from pseudoprogression (PsPD), where increased FDG uptake might reflect immune cell activity rather than tumor growth, remains a challenge. This prospective study compared the efficacy of dual-time point (DTP) FDG-PET/CT with modified response criteria (PERCIMT) in differentiating PsPD from PD. From July 2017-January 2021, 41 patients suspected to have PsPD on an evaluation scan were prospectively included (29 evaluable). A subsequent DTP FDG-PET/CT scan was conducted within 14 days, followed by a confirmatory FDG-PET/CT scan. Additionally, PERCIMT were applied. DTP FDG-PET/CT identified 24% with PsPD and 76% with PD. Applying PERCIMT criteria, 69% showed PsPD, while 31% had PD. On follow-up, 10 patients (34%) demonstrated confirmed PsPD, while 19 (66%) exhibited PD. The sensitivity and specificity of DTP FDG-PET/CT were 20% and 74%, respectively, and for PERCIMT this was 80% and 37%, respectively. Our findings suggest limited efficacy of DTP FDG-PET/CT in distinguishing PsPD from PD in ICI-treated patients with metastatic melanoma. The use of PERCIMT could complement clinical assessment and be incorporated in multidisciplinary team conferences for enhanced decision-making.
Collapse
Affiliation(s)
- Sif Homburg
- Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Charlotte Birk Christensen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Magnus Pedersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Simon Grund Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Helle Westergren Hendel
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev and Gentofte, 2730 Herlev, Denmark
| |
Collapse
|
43
|
Li Y, Wang P, Xu J, Shi X, Yin T, Teng F. Noninvasive radiomic biomarkers for predicting pseudoprogression and hyperprogression in patients with non-small cell lung cancer treated with immune checkpoint inhibition. Oncoimmunology 2024; 13:2312628. [PMID: 38343749 PMCID: PMC10857548 DOI: 10.1080/2162402x.2024.2312628] [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] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
This study aimed to develop a computed tomography (CT)-based radiomics model capable of precisely predicting hyperprogression and pseudoprogression (PP) in patients with non-small cell lung cancer (NSCLC) treated with immunotherapy. We retrospectively analyzed 105 patients with NSCLC, from three institutions, treated with immune checkpoint inhibitors (ICIs) and categorized them into training and independent testing set. Subsequently, we processed CT scans with a series of image-preprocessing techniques, and 6008 radiomic features capturing intra- and peritumoral texture patterns were extracted. We used the least absolute shrinkage and selection operator logistic regression model to select radiomic features and construct machine learning models. To further differentiate between progressive disease (PD) and hyperprogressive disease (HPD), we developed a new radiomics model. The logistic regression (LR) model showed optimal performance in distinguishing PP from HPD, with areas under the receiver operating characteristic curve (AUC) of 0.95 (95% confidence interval [CI]: 0.91-0.99) and 0.88 (95% CI: 0.66-1) in the training and testing sets, respectively. Additionally, the support vector machine model showed optimal performance in distinguishing PD from HPD, with AUC of 0.97 (95% CI: 0.93-1) and 0.87 (95% CI: 0.72-1) in the training and testing sets, respectively. Kaplan‒Meier survival curves showed clear stratification between PP predicted by the radiomics model and true progression (HPD and PD) (hazard ratio = 0.337, 95% CI: 0.200-0.568, p < 0.01) in overall survival. Our study demonstrates that radiomic features extracted from baseline CT scans are effective in predicting PP and HPD in patients with NSCLC treated with ICIs.
Collapse
Affiliation(s)
- Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Peiliang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Xiaonan Shi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Tianwen Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
44
|
Li L, Jiang H, Zeng B, Wang X, Bao Y, Chen C, Ma L, Yuan J. Liquid biopsy in lung cancer. Clin Chim Acta 2024; 554:117757. [PMID: 38184141 DOI: 10.1016/j.cca.2023.117757] [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: 10/23/2023] [Revised: 12/29/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Lung cancer is a highly prevalent malignancy worldwide and the primary cause of mortality. The absence of systematic and standardized diagnostic approaches for identifying potential pulmonary nodules, early-stage cancers, and indeterminate tumors has led clinicians to consider tissue biopsy and pathological sections as the preferred method for clinical diagnosis, often regarded as the gold standard. The conventional tissue biopsy is an invasive procedure that does not adequately capture the diverse characteristics and evolving nature of tumors. Recently, the concept of 'liquid biopsy' has gained considerable attention as a promising solution. Liquid biopsy is a non-invasive approach that facilitates repeated analysis, enabling real-time monitoring of tumor recurrence, metastasis, and response to treatment. Currently, liquid biopsy includes circulating tumor cells, circulating cell-free DNA, circulating tumor DNA, circulating cell-free RNA, extracellular vesicles, and other proteins and metabolites. With rapid progress in molecular technology, liquid biopsy has emerged as a highly promising and intriguing approach, yielding compelling results. This article critically examines the significant role and potential clinical implications of liquid biopsy in the diagnosis, treatment, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Lan Li
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Haixia Jiang
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Bingjie Zeng
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Xianzhao Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Yunxia Bao
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China
| | - Changqiang Chen
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China.
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China.
| | - Jin Yuan
- Department of Laboratory Medicine, Shanghai Chest Hospital Shanghai Jiao Tong University School of Medicine Shanghai China, Shanghai 200030, China; Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
45
|
Powles T, Assaf ZJ, Degaonkar V, Grivas P, Hussain M, Oudard S, Gschwend JE, Albers P, Castellano D, Nishiyama H, Daneshmand S, Sharma S, Sethi H, Aleshin A, Shi Y, Davarpanah N, Carter C, Bellmunt J, Mariathasan S. Updated Overall Survival by Circulating Tumor DNA Status from the Phase 3 IMvigor010 Trial: Adjuvant Atezolizumab Versus Observation in Muscle-invasive Urothelial Carcinoma. Eur Urol 2024; 85:114-122. [PMID: 37500339 DOI: 10.1016/j.eururo.2023.06.007] [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: 12/22/2022] [Revised: 05/16/2023] [Accepted: 06/13/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Interim results from IMvigor010 showed an overall survival (OS) benefit for adjuvant atezolizumab (anti-PD-L1) versus observation in patients with circulating tumor DNA (ctDNA)-positive muscle-invasive urothelial carcinoma (MIUC). OBJECTIVE To report updated OS and safety by ctDNA status. DESIGN, SETTING, AND PARTICIPANTS This ad hoc analysis from a global, open-label, randomized, phase 3 trial (NCT02450331) included intention-to-treat (ITT) population with evaluable cycle 1 day 1 (C1D1) ctDNA samples. INTERVENTION Atezolizumab (1200 mg every 3 wk) or observation for ≤1 yr. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS OS, relapse rates, and safety by ctDNA status were assessed. RESULTS AND LIMITATIONS Among 581 of 809 ITT patients included, 214 (37%) were ctDNA positive. Atezolizumab did not improve OS versus observation in ITT patients (hazard ratio [HR] 0.91 [95% confidence interval {CI} 0.73-1.13]; median follow-up 46.8 mo [interquartile range, 36.1-53.6]). In the observation arm, ctDNA positivity versus negativity was associated with shorter OS (HR 6.3 [95% CI 4.3-9.3]). The ctDNA positivity identified patients with an OS benefit favoring atezolizumab versus observation (HR 0.59 [95% CI 0.42-0.83]). A greater reduction in ctDNA levels with atezolizumab (C3D1) was associated with longer OS (100% clearance, 60.0 mo [95% CI 35.5-not estimable]; 50-99% reduction, 34.3 mo [95% CI 15.2-not estimable]; <50% reduction, 19.9 mo [95% CI 16.4-32.2]). The ctDNA positivity at C1D1 + C3D1 was associated with relapse with greater sensitivity than C1D1 alone (68% vs 57%). Adverse events were more frequent with atezolizumab than with observation, regardless of ctDNA status. A study limitation was its exploratory design. CONCLUSIONS Evidence suggests that ctDNA positivity in MIUC predicts a benefit with atezolizumab. An in-progress prospective study will further evaluate these findings. PATIENT SUMMARY Among patients with urothelial cancer after surgery, survival was poorer if tumor-derived DNA was detected in their bloodstream; these patients' survival was longer with atezolizumab versus observation. Bloodstream tumor-derived DNA may identify patients who benefit from atezolizumab.
Collapse
Affiliation(s)
- Thomas Powles
- Barts Cancer Institute, Queen Mary University of London ECMC, Barts Health, London, UK.
| | | | | | - Petros Grivas
- University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Stephane Oudard
- Georges Pompidou European Hospital, University of Paris, Paris, France
| | - Jürgen E Gschwend
- Department of Urology, Rechts der Isar Medical Center, Technical University Munich, Munich, Germany
| | - Peter Albers
- Department of Urology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Castellano
- Medical Oncology Department CIBER-ONC, University Hospital 12 de Octubre, Madrid, Spain
| | - Hiroyuki Nishiyama
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | | | | | | | | | - Yi Shi
- Roche/Genentech, South San Francisco, CA, USA
| | | | | | - Joaquim Bellmunt
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
46
|
Capuozzo M, Santorsola M, Ferrara F, Cinque C, Farace S, Patrone R, Granata V, Zovi A, Nasti G, Ottaiano A. Intrahepatic cholangiocarcinoma biomarkers: Towards early detection and personalized pharmacological treatments. Mol Cell Probes 2024; 73:101951. [PMID: 38244704 DOI: 10.1016/j.mcp.2024.101951] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
Cholangiocarcinoma (CCA) is a rare malignancy originating from the biliary tree and is anatomically categorized as intrahepatic (iCCA), perihilar, and extrahepatic or distal. iCCA, the second most prevalent hepatobiliary cancer following hepatocellular carcinoma (HCC), constitutes 5-20 % of all liver malignancies, with an increasing incidence. The challenging nature of iCCA, combined with nonspecific symptoms, often leads to late diagnoses, resulting in unfavorable outcomes. The advanced phase of this neoplasm is difficult to treat with dismal results. Early diagnosis could significantly reduce mortality attributed to iCCA but remains an elusive goal. The identification of biomarkers specific to iCCA and their translation into clinical practice could facilitate diagnosis, monitor therapy response, and potentially reveal novel interventions and personalized medicine. In this review, we present the current landscape of biomarkers in each of these contexts. In addition to CA19.9, a widely recognized biomarker for iCCA, others such as A1BG, CYFRA 21-1, FAM19A5, MMP-7, RBAK, SSP411, TuM2-PK, WFA, etc., as well as circulating tumor DNA, RNA, cells, and exosomes, are under investigation. Advancing our knowledge and monitoring of biomarkers may enable us to improve diagnosis, prognostication, and apply treatments dynamically and in a more personalized manner.
Collapse
Affiliation(s)
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131, Napoli, Italy
| | | | - Claudia Cinque
- Pharmaceutical Department, ASL-Naples-3, 80056, Ercolano, Italy
| | - Stefania Farace
- Pharmaceutical Department, ASL-Naples-3, 80056, Ercolano, Italy
| | - Renato Patrone
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131, Napoli, Italy
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131, Napoli, Italy
| | - Andrea Zovi
- Hospital Pharmacist, Ministry of Health, 00144, Roma, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131, Napoli, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via Mariano Semmola, 80131, Napoli, Italy.
| |
Collapse
|
47
|
Guler GD, Ning Y, Coruh C, Mognol GP, Phillips T, Nabiyouni M, Hazen K, Scott A, Volkmuth W, Levy S. Plasma cell-free DNA hydroxymethylation profiling reveals anti-PD-1 treatment response and resistance biology in non-small cell lung cancer. J Immunother Cancer 2024; 12:e008028. [PMID: 38212123 PMCID: PMC10806554 DOI: 10.1136/jitc-2023-008028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Treatment with immune checkpoint inhibitors (ICIs) targeting programmed death-1 (PD-1) can yield durable antitumor responses, yet not all patients respond to ICIs. Current approaches to select patients who may benefit from anti-PD-1 treatment are insufficient. 5-hydroxymethylation (5hmC) analysis of plasma-derived cell-free DNA (cfDNA) presents a novel non-invasive approach for identification of therapy response biomarkers which can tackle challenges associated with tumor biopsies such as tumor heterogeneity and serial sample collection. METHODS 151 blood samples were collected from 31 patients with non-small cell lung cancer (NSCLC) before therapy started and at multiple time points while on therapy. Blood samples were processed to obtain plasma-derived cfDNA, followed by enrichment of 5hmC-containing cfDNA fragments through biotinylation via a two-step chemistry and binding to streptavidin coated beads. 5hmC-enriched cfDNA and whole genome libraries were prepared in parallel and sequenced to obtain whole hydroxymethylome and whole genome plasma profiles, respectively. RESULTS Comparison of on-treatment time point to matched pretreatment samples from same patients revealed that anti-PD-1 treatment induced distinct changes in plasma cfDNA 5hmC profiles of responding patients, as judged by Response evaluation criteria in solid tumors, relative to non-responders. In responders, 5hmC accumulated over genes involved in immune activation such as inteferon (IFN)-γ and IFN-α response, inflammatory response and tumor necrosis factor (TNF)-α signaling, whereas in non-responders 5hmC increased over epithelial to mesenchymal transition genes. Molecular response to anti-PD-1 treatment, as measured by 5hmC changes in plasma cfDNA profiles were observed early on, starting with the first cycle of treatment. Comparison of pretreatment plasma samples revealed that anti-PD-1 treatment response and resistance associated genes can be captured by 5hmC profiling of plasma-derived cfDNA. Furthermore, 5hmC profiling of pretreatment plasma samples was able to distinguish responders from non-responders using T cell-inflamed gene expression profile, which was previously identified by tissue RNA analysis. CONCLUSIONS These results demonstrate that 5hmC profiling can identify response and resistance associated biological pathways in plasma-derived cfDNA, offering a novel approach for non-invasive prediction and monitoring of immunotherapy response in NSCLC.
Collapse
Affiliation(s)
| | - Yuhong Ning
- ClearNote Health Inc, San Diego, California, USA
| | - Ceyda Coruh
- ClearNote Health Inc, San Diego, California, USA
| | | | | | | | - Kyle Hazen
- ClearNote Health Inc, San Diego, California, USA
| | - Aaron Scott
- ClearNote Health Inc, San Diego, California, USA
| | | | - Samuel Levy
- ClearNote Health Inc, San Diego, California, USA
| |
Collapse
|
48
|
van der Leest P, Schuuring E. Critical Factors in the Analytical Work Flow of Circulating Tumor DNA-Based Molecular Profiling. Clin Chem 2024; 70:220-233. [PMID: 38175597 DOI: 10.1093/clinchem/hvad194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/30/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Liquid biopsy testing, especially molecular tumor profiling of circulating tumor DNA (ctDNA) in cell-free plasma, has received increasing interest in recent years as it serves as a reliable alternative for the detection of tumor-specific aberrations to guide treatment decision-making in oncology. Many (commercially available) applications have been developed, however, broad divergences in (pre)analytical work flows and lack of universally applied guidelines impede routine clinical implementation. In this review, critical factors in the blood-based ctDNA liquid biopsy work flow are evaluated. CONTENT In the preanalytical phase, several aspects (e.g., blood collection tubes [BCTs], plasma processing, and extraction method) affect the quantity and quality of the circulating cell-free DNA (ccfDNA) applicable for subsequent molecular analyses and should meet certain standards to be applied in diagnostic work flows. Analytical considerations, such as analytical input and choice of assay, might vary based on the clinical application (i.e., screening, primary diagnosis, minimal residual disease [MRD], response monitoring, and resistance identification). In addition to practical procedures, variant interpretation and reporting ctDNA results should be harmonized. Collaborative efforts in (inter)national consortia and societies are essential for the establishment of standard operating procedures (SOPs) in attempts to standardize the plasma-based ctDNA analysis work flow. SUMMARY Development of universally applicable guidelines regarding the critical factors in liquid biopsy testing are necessary to pave the way to clinical implementation for routine diagnostics.
Collapse
Affiliation(s)
- Paul van der Leest
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ed Schuuring
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
49
|
Qi J, Qi Q, Zhou Z, Wu Y, Cai A, Wu J, Chen B, Wang Q, Chen L, Wang F. PER-CRISPR/Cas14a system-based electrochemical biosensor for the detection of ctDNA EGFR L858R. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 16:51-61. [PMID: 38058174 DOI: 10.1039/d3ay01615c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
The detection of epidermal growth factor receptor (EGFR) mutation L858R in circulating tumor DNA (ctDNA) is beneficial for the clinical diagnosis and personalized therapy of non-small cell lung cancer (NSCLC). Herein, for the first time, the combination of the primer exchange reaction (PER) and clustered regularly interspaced short palindromic repeats (CRISPR) and its associated nucleases (Cas) 14a was used in electrochemical biosensor construction for the detection of ctDNA EGFR L858R. EGFR L858R, as the target, induced the isothermal amplification of the PER reaction, and then the CRISPR/Cas14a system was activated; subsequently, the substrate ssDNA-MB was cleaved and the electron on the surface of the gold electrode transferred, resulting in the fluctuation of the electrochemical redox signal on the electrode surface, whereas the electrochemical signal will be stable when EGFR L858R is absent. Therefore, the concentration of EGFR L858R can be quantified by electrochemical signal analysis. The low detection limit is 0.34 fM and the dynamic detection range is from 1 fM to 1 μM in this work. The PER-CRISPR/Cas14a electrochemical biosensor greatly improved the analytical sensitivity. In addition, this platform also exhibited excellent specificity, reproducibility, stability and good recovery. This study provides an efficient and novel strategy for the detection of ctDNA EGFR L858R, which has great potential for application in the diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Jing Qi
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Qianyi Qi
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Zhou Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yixuan Wu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Aiting Cai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jinran Wu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Bairong Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Qingxiang Wang
- College of Chemistry and Environment, Minnan Normal University, Zhangzhou 363000, China
- Nantong Institute of Liver Diseases, Nantong Third People's Hospital Affiliated Nantong Hospital 3 of Nantong University, Nantong 226006, China.
| | - Lin Chen
- Nantong Institute of Liver Diseases, Nantong Third People's Hospital Affiliated Nantong Hospital 3 of Nantong University, Nantong 226006, China.
| | - Feng Wang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
50
|
Spiliopoulou P, Holanda Lopes CD, Spreafico A. Promising and Minimally Invasive Biomarkers: Targeting Melanoma. Cells 2023; 13:19. [PMID: 38201222 PMCID: PMC10777980 DOI: 10.3390/cells13010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/29/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
The therapeutic landscape of malignant melanoma has been radically reformed in recent years, with novel treatments emerging in both the field of cancer immunotherapy and signalling pathway inhibition. Large-scale tumour genomic characterization has accurately classified malignant melanoma into four different genomic subtypes so far. Despite this, only somatic mutations in BRAF oncogene, as assessed in tumour biopsies, has so far become a validated predictive biomarker of treatment with small molecule inhibitors. The biology of tumour evolution and heterogeneity has uncovered the current limitations associated with decoding genomic drivers based only on a single-site tumour biopsy. There is an urgent need to develop minimally invasive biomarkers that accurately reflect the real-time evolution of melanoma and that allow for streamlined collection, analysis, and interpretation. These will enable us to face challenges with tumour tissue attainment and process and will fulfil the vision of utilizing "liquid biopsy" to guide clinical decisions, in a manner akin to how it is used in the management of haematological malignancies. In this review, we will summarize the most recent published evidence on the role of minimally invasive biomarkers in melanoma, commenting on their future potential to lead to practice-changing discoveries.
Collapse
Affiliation(s)
- Pavlina Spiliopoulou
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | | | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
| |
Collapse
|