201
|
The Role of Systems Biology in Deciphering Asthma Heterogeneity. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101562. [PMID: 36294997 PMCID: PMC9605413 DOI: 10.3390/life12101562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022]
Abstract
Asthma is one of the most common and lifelong and chronic inflammatory diseases characterized by inflammation, bronchial hyperresponsiveness, and airway obstruction episodes. It is a heterogeneous disease of varying and overlapping phenotypes with many confounding factors playing a role in disease susceptibility and management. Such multifactorial disorders will benefit from using systems biology as a strategy to elucidate molecular insights from complex, quantitative, massive clinical, and biological data that will help to understand the underlying disease mechanism, early detection, and treatment planning. Systems biology is an approach that uses the comprehensive understanding of living systems through bioinformatics, mathematical, and computational techniques to model diverse high-throughput molecular, cellular, and the physiologic profiling of healthy and diseased populations to define biological processes. The use of systems biology has helped understand and enrich our knowledge of asthma heterogeneity and molecular basis; however, such methods have their limitations. The translational benefits of these studies are few, and it is recommended to reanalyze the different studies and omics in conjugation with one another which may help understand the reasons for this variation and help overcome the limitations of understanding the heterogeneity in asthma pathology. In this review, we aim to show the different factors that play a role in asthma heterogeneity and how systems biology may aid in understanding and deciphering the molecular basis of asthma.
Collapse
|
202
|
Malapelle U, Pisapia P, Pepe F, Russo G, Buono M, Russo A, Gomez J, Khorshid O, Mack PC, Rolfo C, Troncone G. The evolving role of liquid biopsy in lung cancer. Lung Cancer 2022; 172:53-64. [PMID: 35998482 DOI: 10.1016/j.lungcan.2022.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 12/20/2022]
Abstract
Liquid biopsy has revolutionized the management of cancer patients. In particular, liquid biopsy-based testing has proven to be highly beneficial for identifying actionable cancer markers, especially when solid tissue biopsies are insufficient or unattainable. Beyond the predictive role, liquid biopsy may be a useful tool for comprehensive tumor genotyping, identification of emergent resistance mechanisms, monitoring of minimal residual disease, early detection, and cancer interception. The application of next generation sequencing to liquid biopsy has led to the "quantum leap" of predictive molecular pathology. Here, we review the evolving role of liquid biopsy in lung cancer.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mauro Buono
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Jorge Gomez
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Ola Khorshid
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Philip C Mack
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
203
|
Shah UJ, Alsulimani A, Ahmad F, Mathkor DM, Alsaieedi A, Harakeh S, Nasiruddin M, Haque S. Bioplatforms in liquid biopsy: advances in the techniques for isolation, characterization and clinical applications. Biotechnol Genet Eng Rev 2022; 38:339-383. [PMID: 35968863 DOI: 10.1080/02648725.2022.2108994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue biopsy analysis has conventionally been the gold standard for cancer prognosis, diagnosis and prediction of responses/resistances to treatments. The existing biopsy procedures used in clinical practice are, however, invasive, painful and often associated with pitfalls like poor recovery of tumor cells and infeasibility for repetition in single patients. To circumvent these limitations, alternative non-invasive, rapid and economical, yet sturdy, consistent and dependable, biopsy techniques are required. Liquid biopsy is an emerging technology that fulfills these criteria and potentially much more in terms of subject-specific real-time monitoring of cancer progression, determination of tumor heterogeneity and treatment responses, and specific identification of the type and stages of cancers. The present review first briefly revisits the state-of-the-art technique of liquid biopsy and then proceeds to address in detail, the advances in the potential clinical applications of four major biological agencies present in liquid biopsy samples (circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes and tumor-educated platelets (TEPs)). Finally, the authors conclude with the limitations that need to be addressed in order for liquid biopsy to effectively replace the conventional invasive biopsy methods in the clinical settings.
Collapse
Affiliation(s)
- Ushma Jaykamal Shah
- MedGenome Labs Ltd, Kailash Cancer Hospital and Research Center, Vadodara, India
| | - Ahmad Alsulimani
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore, India
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ahdab Alsaieedi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Steve Harakeh
- King Fahd Medical Research Center, and Yousef Abdullatif Jameel Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Nasiruddin
- MedGenome Labs Ltd, Narayana Health City, Bangalore, India.,Genomics Lab, Orbito Asia Diagnostics, Coimbatore, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
204
|
Tulpule V, Morrison GJ, Falcone M, Quinn DI, Goldkorn A. Integration of Liquid Biopsies in Clinical Management of Metastatic Prostate Cancer. Curr Oncol Rep 2022; 24:1287-1298. [PMID: 35575959 PMCID: PMC9474724 DOI: 10.1007/s11912-022-01278-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2022] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The field of liquid biopsies is constantly evolving through novel technologies. This review outlines current data on liquid biopsies and application to clinical management of metastatic prostate cancer. RECENT FINDINGS To date, there are three platforms with FDA approval for use in the setting of metastatic prostate cancer and others which have been clinically validated. There is substantial evidence supporting the use of circulating tumor cell (CTC) enumeration to guide prognosis in metastatic castration-resistant prostate cancer (mCRPC). Additional evidence supports targeted sequencing of CTC and cell-free DNA (cfDNA) to guide androgren-directed therapy, identify candidates for treatment with PARP inhibitors, and monitor development of resistance. As a real-time and minimally invasive approach, utilization of liquid biopsies has the potential to drastically impact the treatment of metastatic prostate cancer and improve overall survival. With further clinical validation, additional liquid biopsy is likely to enter standard clinical practice.
Collapse
Affiliation(s)
- Varsha Tulpule
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gareth J Morrison
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mary Falcone
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David I Quinn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
205
|
Galvano A, Castellana L, Gristina V, La Mantia M, Insalaco L, Barraco N, Perez A, Cutaia S, Calò V, Bazan Russo TD, Francini E, Incorvaia L, Mirisola MG, Vieni S, Rolfo C, Bazan V, Russo A. The diagnostic accuracy of PIK3CA mutations by circulating tumor DNA in breast cancer: an individual patient data meta-analysis. Ther Adv Med Oncol 2022; 14:17588359221110162. [PMID: 36188485 PMCID: PMC9516428 DOI: 10.1177/17588359221110162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Background The circulating tumor DNA (ctDNA) diagnostic accuracy for detecting phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations in breast cancer (BC) is under discussion. We aimed to compare plasma and tissue PIK3CA alterations, encompassing factors that could affect the results. Methods Two reviewers selected studies from different databases until December 2020. We considered BC patients with matched tumor tissue and plasma ctDNA. We performed meta-regression and subgroup analyses to explore sources of heterogeneity concerning tumor burden, diagnostic technique, sample size, sampling time, biological subtype, and hotspot mutation. Pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and the related area under the curve (AUC) were elaborated for the overall population and each subgroup. Results The pooled analysis was carried out on 25 cohorts for a total of 1966 patients. The overall ctDNA sensitivity and specificity were 0.73 (95% CI: 0.70-0.77) and 0.87 (95% CI: 0.85-0.89). The AUC was 0.93. Pooled concordance, negative predictive value and positive predictive value values were 0.87 (95% CI: 0.82-0.92), 0.86 (95% CI: 0.81-0.90), and 0.89 (95% CI: 0.81-0.95) with pooled PLR, NLR, and DOR of 7.94 (95% CI: 4.90-12.86), 0.33 (95% CI: 0.25-0.45), and 33.41 (95% CI: 17.23-64.79), respectively. The pooled results consistently favored next-generation sequencing (NGS)- over polymerase chain reaction-based methodologies. The best ctDNA performance in terms of sensitivity, specificity, and AUC (0.85, 0.99, and 0.94, respectively) was observed in the low-time sampling subgroup (⩽18 days between tissue and plasma collection). Meta-regression and subgroup analyses highlighted sampling time as a possible major cause of heterogeneity. Conclusions These findings reliably estimate the high ctDNA accuracy for the detection of PIK3CA mutations. A ctDNA-first approach for the assessment of PIK3CA mutational status by NGS may accurately replace tissue tumor sampling, representing the preferable strategy at diagnosis of metastatic BC in patients who present with visceral involvement and at least two metastatic lesions, primarily given low clinical compliance or inaccessible metastatic sites.
Collapse
Affiliation(s)
- Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Luisa Castellana
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Maria La Mantia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lavinia Insalaco
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Sofia Cutaia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Valentina Calò
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | | | - Edoardo Francini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Mario Giuseppe Mirisola
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Salvatore Vieni
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Bazan
- Department of Experimental Biomedicine and Clinical Neurosciences, School of Medicine, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, Palermo 90127, Italy
| |
Collapse
|
206
|
Nikanjam M, Kato S, Kurzrock R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol 2022; 15:131. [PMID: 36096847 PMCID: PMC9465933 DOI: 10.1186/s13045-022-01351-y] [Citation(s) in RCA: 348] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022] Open
Abstract
Liquid biopsies are increasingly used for cancer molecular profiling that enables a precision oncology approach. Circulating extracellular nucleic acids (cell-free DNA; cfDNA), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs) can be isolated from the blood and other body fluids. This review will focus on current technologies and clinical applications for liquid biopsies. ctDNA/cfDNA has been isolated and analyzed using many techniques, e.g., droplet digital polymerase chain reaction, beads, emulsion, amplification, and magnetics (BEAMing), tagged-amplicon deep sequencing (TAm-Seq), cancer personalized profiling by deep sequencing (CAPP-Seq), whole genome bisulfite sequencing (WGBS-Seq), whole exome sequencing (WES), and whole genome sequencing (WGS). CTCs have been isolated using biomarker-based cell capture, and positive or negative enrichment based on biophysical and other properties. ctDNA/cfDNA and CTCs are being exploited in a variety of clinical applications: differentiating unique immune checkpoint blockade response patterns using serial samples; predicting immune checkpoint blockade response based on baseline liquid biopsy characteristics; predicting response and resistance to targeted therapy and chemotherapy as well as immunotherapy, including CAR-T cells, based on serial sampling; assessing shed DNA from multiple metastatic sites; assessing potentially actionable alterations; analyzing prognosis and tumor burden, including after surgery; interrogating difficult-to biopsy tumors; and detecting cancer at early stages. The latter can be limited by the small amounts of tumor-derived components shed into the circulation; furthermore, cfDNA assessment in all cancers can be confounded by clonal hematopoeisis of indeterminate potential, especially in the elderly. CTCs can be technically more difficult to isolate that cfDNA, but permit functional assays, as well as evaluation of CTC-derived DNA, RNA and proteins, including single-cell analysis. Blood biopsies are less invasive than tissue biopsies and hence amenable to serial collection, which can provide critical molecular information in real time. In conclusion, liquid biopsy is a powerful tool, and remarkable advances in this technology have impacted multiple aspects of precision oncology, from early diagnosis to management of refractory metastatic disease. Future research may focus on fluids beyond blood, such as ascites, effusions, urine, and cerebrospinal fluid, as well as methylation patterns and elements such as exosomes.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, 1200 Garden View Road, Encinitas, CA, 92024, USA.
| | - Shumei Kato
- Division of Hematology-Oncology, University of California San Diego, La Jolla, 1200 Garden View Road, Encinitas, CA, 92024, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.,WIN Consortium, Paris, France
| |
Collapse
|
207
|
Madanat-Harjuoja LM, Renfro LA, Klega K, Tornwall B, Thorner AR, Nag A, Dix D, Dome JS, Diller LR, Fernandez CV, Mullen EA, Crompton BD. Circulating Tumor DNA as a Biomarker in Patients With Stage III and IV Wilms Tumor: Analysis From a Children's Oncology Group Trial, AREN0533. J Clin Oncol 2022; 40:3047-3056. [PMID: 35580298 PMCID: PMC9462535 DOI: 10.1200/jco.22.00098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/09/2022] [Accepted: 04/01/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The utility of circulating tumor DNA (ctDNA) analyses has not been established in the risk stratification of Wilms tumor (WT). We evaluated the detection of ctDNA and selected risk markers in the serum and urine of patients with WT and compared findings with those of matched diagnostic tumor samples. PATIENTS AND METHODS Fifty of 395 children with stage III or IV WT enrolled on Children's Oncology Group trial AREN0533 had banked pretreatment serum, urine, and tumor available. Next-generation sequencing was used to detect ctDNA. Copy-number changes in 1q, 16q, and 1p, and single-nucleotide variants in serum and urine were compared with tumor biopsy data. Event-free survival (EFS) was compared between patients with and without ctDNA detection. RESULTS ctDNA was detected in the serum of 41/50 (82%) and in the urine in 13/50 (26%) patients. Agreement between serum ctDNA detection and tumor sequencing results was as follows: 77% for 1q gain, 88% for 16q deletions, and 70% for 1p deletions, with ĸ-coefficients of 0.56, 0.74, and 0.29, respectively. Sequencing also demonstrated that single-nucleotide variants detected in tumors could be identified in the ctDNA. There was a trend toward worse EFS in patients with ctDNA detected in the serum (4-year EFS 80% v 100%, P = .14). CONCLUSION ctDNA demonstrates promise as an easily accessible prognostic biomarker with potential to detect tumor heterogeneity. The observed trend toward more favorable outcome in patients with undetectable ctDNA requires validation. ctDNA profiling should be further explored as a noninvasive diagnostic and prognostic tool in the risk-adapted treatment of patients with WT.
Collapse
Affiliation(s)
| | | | - Kelly Klega
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Brett Tornwall
- Children's Oncology Group Statistics and Data Center, Monrovia, CA
| | - Aaron R. Thorner
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA
| | - Anwesha Nag
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA
| | - David Dix
- BC Children's Hospital, Vancouver, BC, Canada
| | - Jeffrey S. Dome
- Children's National Hospital and the George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Lisa R. Diller
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | | | | | - Brian D. Crompton
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
208
|
Couto N, Elzanowska J, Maia J, Batista S, Pereira CE, Beck HC, Carvalho AS, Strano Moraes MC, Carvalho C, Oliveira M, Matthiesen R, Costa-Silva B. IgG+ Extracellular Vesicles Measure Therapeutic Response in Advanced Pancreatic Cancer. Cells 2022; 11:cells11182800. [PMID: 36139375 PMCID: PMC9496671 DOI: 10.3390/cells11182800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/06/2022] [Accepted: 08/18/2022] [Indexed: 12/05/2022] Open
Abstract
(1) Background: Pancreatic ductal adenocarcinoma (PDAC) is expected to be the second-leading cause of cancer deaths by 2030. Imaging techniques are the standard for monitoring the therapy response in PDAC, but these techniques have considerable limits, including delayed disease progression detection and difficulty in distinguishing benign from malignant lesions. Extracellular vesicle (EV) liquid biopsy is an emerging diagnosis modality. Nonetheless, the majority of research for EV-based diagnosis relies on point analyses of EVs at specified times, while longitudinal EV population studies before and during therapeutic interventions remain largely unexplored. (2) Methods: We analyzed plasma EV protein composition at diagnosis and throughout PDAC therapy. (3) Results: We found that IgG is linked with the diagnosis of PDAC and the patient’s response to therapy, and that the IgG+ EV population increases with disease progression and reduces with treatment response. Importantly, this covers PDAC patients devoid of the standard PDAC seric marker CA19.9 expression. We also observed that IgG is bound to EVs via the tumor antigen MAGE B1, and that this is independent of the patient’s inflammatory condition and IgG seric levels. (4) Conclusions: We here propose that a population analysis of IgG+ EVs in PDAC plasma represents a novel method to supplement the monitoring of the PDAC treatment response.
Collapse
Affiliation(s)
- Nuno Couto
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Digestive Unit, Champalimaud Clinical Centre, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Julia Elzanowska
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Joana Maia
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Silvia Batista
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Catarina Esteves Pereira
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Sdr. Boulevard 29, DK-5000 Odense, Denmark
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, iNOVA4Health, NOVA MedicalSchool|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-199 Lisbon, Portugal
| | | | - Carlos Carvalho
- Digestive Unit, Champalimaud Clinical Centre, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Manuela Oliveira
- Department of Mathematics and CIMA-Center for Research on Mathematics and Its Applications, University of Évora, 7004-516 Evora, Portugal
| | - Rune Matthiesen
- Computational and Experimental Biology Group, iNOVA4Health, NOVA MedicalSchool|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, 1150-199 Lisbon, Portugal
- Correspondence: (R.M.); (B.C.-S.); Tel.: +351-939-218-696 (R.M.); +351-210-480-134 (B.C.-S.)
| | - Bruno Costa-Silva
- Champalimaud Physiology and Cancer Programme, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Correspondence: (R.M.); (B.C.-S.); Tel.: +351-939-218-696 (R.M.); +351-210-480-134 (B.C.-S.)
| |
Collapse
|
209
|
Tabata M, Liu X, Khamhanglit C, Kotaki S, Miyahara Y. Detection of Epidermal Growth Factor Receptor Expression in Breast Cancer Cell Lines Using an Ion-Sensitive Field-Effect Transistor in Combination with Enzymatic Chemical Signal Amplification. J Am Chem Soc 2022; 144:16545-16552. [PMID: 36054724 DOI: 10.1021/jacs.2c06122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A novel strategy for epidermal growth factor receptor (EGFR) detection using a cell-based field-effect transistor (FET) with enzymatic chemical signal amplification is proposed. Four human breast cancer cell lines [BT474, MDA-MB-231 (MM231), MDA-MB-468 (MM468), and MDA-MB-453 (MM453)] were used to compare the expression levels of EGFR. The cells were non-specifically captured on the surface of the gate of the FET, irrespective of their surface antigens. With this configuration, the heterogeneity of the cells would be analyzed using secondary antibodies conjugated to different kinds of enzymes. Four breast cancer cell lines with different levels of EGFR expression were captured on the respective surfaces of the extracellular matrix (ECM) gel-coated gates of the FETs. Glucose oxidase (GOx) was conjugated to the secondary antibody, and the output signals of the cell-based FETs changed depending on the expression levels of EGFR upon addition of glucose. The order of the expression levels of EGFR among the four cell lines, determined with the cell-based FETs, was consistent with the results of fluorescence detection determined by fluorescence-activated cell sorting (FACS). The cell-based FETs are advantageous for miniaturization and in massive parallel analyses of target molecules expressed on the membranes of cells and EVs, and their small size and cost effectiveness for cancer testing could enable their realization in a future liquid biopsy.
Collapse
Affiliation(s)
- Miyuki Tabata
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062 Japan
| | - Xinyue Liu
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062 Japan
| | - Chattarika Khamhanglit
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062 Japan
| | - Sayo Kotaki
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062 Japan
| | - Yuji Miyahara
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062 Japan
| |
Collapse
|
210
|
Geng Y, Jin L, Tang G, Zhao Z, Gu Y, Yang D. LiqBioer: a manually curated database of cancer biomarkers in body fluid. Database (Oxford) 2022; 2022:6687198. [PMID: 36053554 PMCID: PMC9438745 DOI: 10.1093/database/baac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/15/2022] [Accepted: 08/27/2022] [Indexed: 11/14/2022]
Abstract
Cancer biomarkers are measurable indicators that play vital roles in clinical applications. Biomarkers in body fluids have gained considerable attention since the development of liquid biopsy, and their data volume is rapidly increasing. Nevertheless, current research lacks the compilation of published cancer body fluid biomarkers into a centralized and sustainable repository for researchers and clinicians, despite a handful of small-scale and specific data resources. To fulfill this purpose, we developed liquid biomarker (LiqBioer) containing 6231 manually curated records from 3447 studies, covering 3056 biomarkers and 74 types of cancer in 22 tissues. LiqBioer allows users to browse and download comprehensive information on body liquid biomarkers, including cancer types, source studies and clinical usage. As a comprehensive resource for body fluid biomarkers of cancer, LiqBioer is a powerful tool for researchers and clinicians to query and retrieve biomarkers in liquid biopsy.
Collapse
Affiliation(s)
- Yiding Geng
- Department of Biochemistry and Molecular Biology, Harbin Medical University , 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Lu Jin
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University , 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Guangjue Tang
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University , 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Zhangxiang Zhao
- The Sino-Russian Medical Research Centre, The Institute of Chronic Disease, The First Affiliated Hospital, Jinan University , Guangzhou, Guangdong 510630, China
| | - Yunyan Gu
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University , 157 Baojian Road, Nangang District, Harbin 150081, China
| | - Dan Yang
- Department of Biochemistry and Molecular Biology, Harbin Medical University , 157 Baojian Road, Nangang District, Harbin 150081, China
| |
Collapse
|
211
|
Hai L, Li L, Liu Z, Tong Z, Sun Y. Whole-genome circulating tumor DNA methylation landscape reveals sensitive biomarkers of breast cancer. MedComm (Beijing) 2022; 3:e134. [PMID: 35756163 PMCID: PMC9205580 DOI: 10.1002/mco2.134] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/12/2023] Open
Abstract
The changes in circulating tumor DNA (ctDNA) methylation are believed to be early events in breast cancer initiation, which makes them suitable as promising biomarkers for early diagnosis. However, applying ctDNA in breast cancer early diagnosis remains highly challenging due to the contamination of background DNA from blood and low DNA methylation signals. Here, we report an improved way to extract ctDNA, reduce background contamination, and build a whole-genome bisulfite sequencing (WGBS) library from different stages of breast cancer. We first compared the DNA methylation data of 74 breast cancer patients with those of seven normal controls to screen candidate methylation CpG site biomarkers for breast cancer diagnosis. The obtained 26 candidate ctDNA methylation biomarkers produced high accuracy in breast cancer patients (area under the curve [AUC] = 0.889; sensitivity: 100%; specificity: 75%). Furthermore, we revealed potential ctDNA methylated CpG sites for detecting early-stage breast cancer (AUC = 0.783; sensitivity: 93.44%; specificity: 50%). In addition, different subtypes of breast cancer could be well distinguished by the ctDNA methylome, which was obtained through our improved ctDNA-WGBS method. Overall, we identified high specificity and sensitivity breast cancer-specific methylation CpG site biomarkers, and they will be expected to have the potential to be translated to clinical practice.
Collapse
Affiliation(s)
- Luo Hai
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| | - Lingyu Li
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
| | - Zongzhi Liu
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Zhongsheng Tong
- Department of Breast OncologyTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Yingli Sun
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| |
Collapse
|
212
|
Liquid Biopsy as a Tool for the Diagnosis, Treatment, and Monitoring of Breast Cancer. Int J Mol Sci 2022; 23:ijms23179952. [PMID: 36077348 PMCID: PMC9456236 DOI: 10.3390/ijms23179952] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer (BC) is a highly heterogeneous disease. The treatment of BC is complicated owing to intratumoral complexity. Tissue biopsy and immunohistochemistry are the current gold standard techniques to guide breast cancer therapy; however, these techniques do not assess tumoral molecular heterogeneity. Personalized medicine aims to overcome these biological and clinical complexities. Advances in techniques and computational analyses have enabled increasingly sensitive, specific, and accurate application of liquid biopsy. Such progress has ushered in a new era in precision medicine, where the objective is personalized treatment of breast cancer, early screening, accurate diagnosis and prognosis, relapse detection, longitudinal monitoring, and drug selection. Liquid biopsy can be defined as the sampling of components of tumor cells that are released from a tumor and/or metastatic deposits into the blood, urine, feces, saliva, and other biological substances. Such components include circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) or circulating tumor RNA (ctRNA), platelets, and exosomes. This review aims to highlight the role of liquid biopsy in breast cancer and precision medicine.
Collapse
|
213
|
Pre-treatment strategies based on aqueous two-phase systems comprising ionic liquids to improve the adrenal cancer diagnosis. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
214
|
Exosomal lncRNA PCAT1 Promotes Tumor Circulating Cell-Mediated Colorectal Cancer Liver Metastasis by Regulating the Activity of the miR-329-3p/Netrin-1-CD146 Complex. J Immunol Res 2022; 2022:9916228. [PMID: 36093435 PMCID: PMC9453099 DOI: 10.1155/2022/9916228] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
Objective This study explored the colorectal cancer exosome lncRNA prostate cancer associated transcript 1– (PCAT1) mediated circulating tumors and the mechanism of cell colorectal cancer liver metastasis. Methods Exosomes were extracted from the primary colorectal cancer (CRC) cell lines HCT116 and SW480 and cultured with T84 and human umbilical vein endothelial (HUVE) cells. The expression of PCAT1 and miR-329-3p was detected by real-time quantitative polymerase chain reaction (RT-qPCR), the expression of Netrin-1, CD146, and epithelial mesenchymal transition (EMT) related proteins was detected by Western blot, the proliferation activity of T84 cells was detected by cell counting kit 8 (CCK-8), and cell migration was detected by Transwell. The expression of the F-actin signal was detected by immunofluorescence after coculture of exosomes with human umbilical vein endothelial cells (HUVECs). Changes in subcutaneous tumor and liver nodule size after PCAT1 deletion were observed in a mouse model of liver metastasis from rectal cancer. Results PCAT1 expression was upregulated in primary cell lines and their exosomes. After exosomes were cocultured with colorectal cancer tumor circulating T84 cells, the expression of Netrin-1 and CD146 was upregulated, the expression of miR-329-3p was downregulated, the proliferation and migration ability of T84 cells were enhanced, and EMT occurred. After knocking down PCAT1, the above phenomenon was reversed. Similarly, after exosomes were cocultured with HUVECs, the expression of the F-actin signal increased, and after PCAT1 was knocked down, the F-actin signal also decreased. PCAT1 regulates miR-329-3p/Netrin-1 and affects the biological behavior of T84 and F-actin signal expression in HUVECs. In a mouse model of colorectal cancer liver metastasis, knocking down PCAT1 significantly reduced the nodules formed by liver metastasis in mice. Conclusions LncRNA PCAT1 derived from colorectal cancer exosomes regulates the activity of the Netrin-1-CD146 complex in circulating tumor cells (CTCs) to promote the occurrence of colorectal cancer EMT and liver metastasis and provides new molecular targets for the treatment of colorectal cancer liver metastasis.
Collapse
|
215
|
Jayasinghe R, Jayarajah U, Seneviratne S. Circulating Biomarkers in the Management of Breast Cancer. Biomark Med 2022. [DOI: 10.2174/9789815040463122010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Circulating biomarkers have become a promising modality in the
management of many cancers. Similarly, in breast cancer, circulatory biomarkers are
useful, non-invasive methods in the diagnosis, prognostication, and evaluation of
response to treatment. Invasive surgical biopsies can be potentially replaced by “liquid
biopsy,” which involves analysing circulatory biomarkers that may reveal features of
primary and metastatic disease. Therefore, providing an insight into the cancer biology
can be utilised to monitor treatment response, treatment-induced adaptation and tumour
and disease progression through non-invasive means. The objective of this review is to
provide an overview of the current status of the circulating biomarkers highlighting
their promising impact on the management of patients with breast cancer.
Collapse
Affiliation(s)
- Ravindri Jayasinghe
- Faculty of Medicine, University of Colombo,Department of Surgery,Department of Surgery, Faculty of Medicine, University of Colombo, Colombo,Sri Lanka
| | - Umesh Jayarajah
- Faculty of Medicine, University of Colombo,Department of Surgery,Department of Surgery, Faculty of Medicine, University of Colombo, Colombo,Sri Lanka
| | - Sanjeewa Seneviratne
- Faculty of Medicine, University of Colombo,Department of Surgery,Department of Surgery, Faculty of Medicine, University of Colombo, Colombo,Sri Lanka
| |
Collapse
|
216
|
Guan S, Deng G, Sun J, Han Q, Lv Y, Xue T, Ding L, Yang T, Qian N, Dai G. Evaluation of circulating tumor DNA as a prognostic biomarker for metastatic pancreatic adenocarcinoma. Front Oncol 2022; 12:926260. [PMID: 36081557 PMCID: PMC9446234 DOI: 10.3389/fonc.2022.926260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
PurposePancreatic cancer is an aggressive solid tumor with a severe prognosis. Although tumor biomarkers are often used to identify advanced pancreatic cancer, this is not accurate, and the currently used biomarkers are not indicative of prognosis. The present study evaluated circulating tumor DNA (ctDNA) as a biomarker for prognosis prediction and disease monitoring in metastatic pancreatic adenocarcinoma (PAC).MethodsFrom 2017 to 2018, 40 patients with metastatic PAC were enrolled, and tumor tissue and blood samples were collected from 40 and 35 patients, respectively. CtDNA was sequenced by next-generation sequencing (NGS) with a 425-gene capture panel. The association of clinical characteristics, laboratory indicators, and dynamic ctDNA with patient outcomes was analyzed.ResultsMutations in KRAS (87.5%, N = 35) and TP53 (77.5%, N = 31) were most common in 40 tumor tissue. Patients’ ECOG score, CA19-9, CEA, neutrophil-lymphocyte ratio (NLR), platelet- lymphocyte ratio (PLR) levels and mutations in ≥ 3 driver genes were strongly correlated with patients’ overall survival (OS). Patients’ gender, ECOG score, CA19-9, and CEA levels were associated with progression-free survival (PFS) (P<0.05). In 35 blood samples, univariate analysis showed a significant association between ECOG score, CA19-9, KRAS or CDKN2A mutation in ctDNA and OS and between CA19-9, CDKN2A or SMAD4 mutation in ctDNA and PFS. Cox hazard proportion model showed that patients’ CDKN2A mutation in ctDNA (HR=16.1, 95% CI=4.4-59.1, P<0.001), ECOG score (HR=6.2, 95% CI=2.4-15.7, P<0.001) and tumor location (HR=0.4, 95% CI=0.1-0.9, P=0.027) were significantly associated with OS. Patients’ CDKN2A mutation in ctDNA (HR=6.8, 95% CI=2.3-19.9, P=0.001), SMAD4 mutation in ctDNA (HR=3.0, 95% CI=1.1-7.9, P=0.031) and metastatic organ (HR=0.4, 95% CI=0.2-1.0, P=0.046) were significantly associated with PFS. Longitudinal changes in gene mutation allelic frequency (MAF) value were evaluated in 24 patients. Detection of progression disease (PD) by ctDNA was 0.9 months earlier than by radiological imaging (mean PFS: 4.6m vs 5.5m, P=0.004, paired t-test).ConclusionsThe ctDNA has the potential as a specific survival predictive marker for metastatic PAC patients. Longitudinal ctDNA tracking could potentially help identify disease progression and be a valuable complement for routine clinical markers and imaging.
Collapse
Affiliation(s)
- Shasha Guan
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Guochao Deng
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Jingjie Sun
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Quanli Han
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yao Lv
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Tianhui Xue
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Lijuan Ding
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Tongxin Yang
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’ Liberation Army (PLA) General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Niansong Qian, ; Guanghai Dai,
| | - Guanghai Dai
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Niansong Qian, ; Guanghai Dai,
| |
Collapse
|
217
|
Liu Z, Han Y, Dang Q, Xu H, Zhang Y, Duo M, Lv J, Li H, Kong Y, Han X. Roles of circulating tumor DNA in PD-1/PD-L1 immune checkpoint Inhibitors: Current evidence and future directions. Int Immunopharmacol 2022; 111:109173. [PMID: 35998502 DOI: 10.1016/j.intimp.2022.109173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022]
Abstract
Circulating tumor DNA (ctDNA) sequencing holds considerable promise for early diagnosis and detection of surveillance and minimal residual disease. Blood ctDNA monitors specific cancers by detecting the alterations found in cancer cells, such as apoptosis and necrosis. Due to the short half-life, ctDNA reflects the actual burden of other treatments on tumors. In addition, ctDNA might be preferable to monitor tumor development and treatment compared with invasive tissue biopsy. ctDNA-based liquid biopsy brings remarkable strength to targeted therapy and precision medicine. Notably, multiple ctDNA analysis platforms have been broadly applied in clinical immunotherapy. Through targeted sequencing, early variations in ctDNA could predict response to immune checkpoint inhibitor (ICI). Several studies have demonstrated a correlation between ctDNA kinetics and anti-PD1 antibodies. The need for further research and development remains, although this biomarker holds significant prospects for early cancer detection. This review focuses on describing the basis of ctDNA and its current utilities in oncology and immunotherapy, either for clinical management or early detection, highlighting its advantages and inherent limitations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| | - Yilin Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mengjie Duo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinxiang Lv
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huanyun Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ying Kong
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
218
|
Malla M, Loree JM, Kasi PM, Parikh AR. Using Circulating Tumor DNA in Colorectal Cancer: Current and Evolving Practices. J Clin Oncol 2022; 40:2846-2857. [PMID: 35839443 PMCID: PMC9390824 DOI: 10.1200/jco.21.02615] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/04/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
There exists a tremendous opportunity in identifying and determining the appropriate predictive and prognostic biomarker(s) for risk stratification of patients with colorectal cancers (CRCs). Circulating tumor DNA (ctDNA) has emerged as a promising prognostic and possibly predictive biomarker in the personalized management of patients with CRCs. The disease is particularly suited to a liquid biopsy-based approach since there is a great deal of shedding of circulating tumor fragments (cells, DNA, methylation markers, etc). ctDNA has been shown to have several potential applications, including detecting minimal residual disease (MRD), monitoring for early recurrence, molecular profiling, and therapeutic response prediction. The utility of ctDNA has broadened from its initial use in the advanced/metastatic setting for molecular profiling and detection of acquired resistance mechanisms, toward identifying MRD, as well as early detection. Prospective studies such as CIRCULATE, COBRA, Dynamic II/III, and ACT3 are underway in the MRD setting to further understand how ctDNA may be used to inform clinical decision making using both tumor-informed and tumor-agnostic platforms. These prospective studies use ctDNA to guide management of patients with CRC and will be critical to help guide how and where ctDNA should or should not be used in clinical decision making. It is also important to understand that there are different types of ctDNA liquid biopsy platforms, each with advantages and disadvantages in different clinical indications. This review provides an overview of the current and evolving use of ctDNA in CRC.
Collapse
Affiliation(s)
- Midhun Malla
- West Virginia University School of Medicine, Section of Hematology/Oncology, Morgantown, WV
| | - Jonathan M Loree
- BC Cancer/The University of British Columbia, Division of Medical Oncology, Vancouver, BC, Canada
| | - Pashtoon Murtaza Kasi
- Weill Cornell Medicine, Meyer Cancer Center, Englander Institute of Precision Medicine, New York, NY
| | - Aparna Raj Parikh
- Harvard Medical School, Massachusetts General Hospital Cancer Center, Tucker Gosnell Center for Gastrointestinal Malignancies, Termeer Center for Targeted Therapies, Boston, MA
| |
Collapse
|
219
|
Zhuang Z, Zhang Y, Zhang X, Zhang M, Zou D, Zhang L, Jia C, Zhang W. Circulating cell-free DNA and IL-10 from cerebrospinal fluids aid primary vitreoretinal lymphoma diagnosis. Front Oncol 2022; 12:955080. [PMID: 36059608 PMCID: PMC9434796 DOI: 10.3389/fonc.2022.955080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
Primary vitreoretinal lymphoma (PVRL) is a rare variant of primary central nervous system lymphoma (PCNSL) that presents diagnostic challenges. Here, we focused on circulating cell-free DNA (cfDNA) and interleukin-10 (IL-10) isolated from cerebrospinal fluid. Twenty-three VRL patients (17 PVRL, 2 PCNSL/O, and 4 relapsed VRL, from 10/2018 to 12/2021) and 8 uveitis patients were included in this study. CSF samples from 19 vitreoretinal lymphoma patients had sufficient cfDNA for next-generation sequencing. Of these patients, 73.7% (14/19) had at least one meaningful non-Hodgkin lymphoma-related mutation. The characteristic MYD88 L265P mutation was detected in the CSF of 12 VRL patients, with a sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of 63.2%, 100%, 100%, and 46.2%, respectively. No meaningful lymphoma related mutations were found in CSF samples from uveitis controls with typical intraocular lesions. Meanwhile, CSF IL-10 levels were elevated in 95.7% of the VRL patients, with a sensitivity, specificity, PPV, and NPV of 95.7%, 100%, 100% and 88.9%, respectively. Key somatic mutations like MYD88 L265P and CD79B detected from CSF cfDNA and elevated CSF IL-10 levels can be promising adjuncts for primary vitreoretinal lymphoma diagnosis.
Collapse
Affiliation(s)
- Zhe Zhuang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiao Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meifen Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dongmei Zou
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China,*Correspondence: Wei Zhang,
| |
Collapse
|
220
|
Duffy MJ, Crown J. Circulating Tumor DNA as a Biomarker for Monitoring Patients with Solid Cancers: Comparison with Standard Protein Biomarkers. Clin Chem 2022; 68:1381-1390. [PMID: 35962648 DOI: 10.1093/clinchem/hvac121] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/21/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND Protein-based biomarkers are widely used in monitoring patients with diagnosed cancer. These biomarkers however, lack specificity for cancer and have poor sensitivity in detecting early recurrences and monitoring therapy effectiveness. Emerging data suggest that the use of circulating tumor DNA (ctDNA) has several advantages over standard biomarkers. CONTENT Following curative-intent surgery for cancer, the presence of ctDNA is highly predictive of early disease recurrence, while in metastatic cancer an early decline in ctDNA following the initiation of treatment is predictive of good outcome. Compared with protein biomarkers, ctDNA provides greater cancer specificity and sensitivity for detecting early recurrent/metastatic disease. Thus, in patients with surgically resected colorectal cancer, multiple studies have shown that ctDNA is superior to carcinoembryonic antigen (CEA) in detecting residual disease and early recurrence. Similarly, in breast cancer, ctDNA was shown to be more accurate than carbohydrate antigen 15-3 (CA 15-3) in detecting early recurrences. Other advantages of ctDNA over protein biomarkers in monitoring cancer patients include a shorter half-life in plasma and an ability to predict likely response to specific therapies and identify mechanisms of therapy resistance. However, in contrast to proteins, ctDNA biomarkers are more expensive to measure, less widely available, and have longer turnaround times for reporting. Furthermore, ctDNA assays are less well standardized. SUMMARY Because of their advantages, it is likely that ctDNA measurements will enter clinical use in the future, where they will complement existing biomarkers and imaging in managing patients with cancer. Hopefully, these combined approaches will lead to a better outcome for patients.
Collapse
Affiliation(s)
- Michael J Duffy
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin, Ireland
| | - John Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
221
|
Che H, Stanley K, Jatsenko T, Thienpont B, Vermeesch JR. Expanded knowledge of cell-free DNA biology: potential to broaden the clinical utility. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:216-234. [PMID: 39697489 PMCID: PMC11648412 DOI: 10.20517/evcna.2022.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 12/20/2024]
Abstract
Noninvasive sampling of an individual's body fluids is an easy means to capture circulating cell-free DNA (cfDNA). These small fragments of DNA carry information on the contributing cell's genome, epigenome, and nuclease content. Analysis of cfDNA for the assessment of genetic risk has already revolutionized clinical practice, and a compendium of increasingly higher-resolution approaches based on epigenetic and fragmentomic cfDNA signatures continues to expand. Profiling cfDNA has unlocked a wealth of molecular information that can be translated to the clinic. This review covers the biological characteristics of cfDNA, recent advances in liquid biopsy and the clinical utility of cfDNA.
Collapse
Affiliation(s)
- Huiwen Che
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven 3000, Belgium
| | - Kate Stanley
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven 3000, Belgium
| | - Tatjana Jatsenko
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven 3000, Belgium
| | - Bernard Thienpont
- Department of Human Genetics, Laboratory for Functional Epigenetics, KU Leuven, Leuven 3000, Belgium
| | - Joris Robert Vermeesch
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven 3000, Belgium
- Center for Human Genetics, University Hospitals Leuven, Leuven 3000, Belgium
| |
Collapse
|
222
|
Bocci M, Fabbri F, Neves RP, Rossi E. Editorial: Liquid biopsy: A tool for better understanding of the metastatic process ecosystem. Front Oncol 2022; 12:951158. [PMID: 36033488 PMCID: PMC9399825 DOI: 10.3389/fonc.2022.951158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Matteo Bocci
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, Lund, Sweden
- *Correspondence: Matteo Bocci,
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Rui P.L. Neves
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Elisabetta Rossi
- Department of Surgery, Oncology and Gastroenterology, Oncology Section, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
223
|
Eibl RH, Schneemann M. Cell-free DNA as a biomarker in cancer. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:195-215. [PMID: 39697490 PMCID: PMC11648514 DOI: 10.20517/evcna.2022.20] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/07/2022] [Accepted: 07/26/2022] [Indexed: 12/20/2024]
Abstract
Translational research of liquid biopsy is just at the edge of routine clinical application: an emerging validity of circulating tumor DNA (ctDNA) tests suggests its use for earlier cancer detection and better monitoring of minimal residual disease (MRD) and resistance development, thus offering earlier guidance for therapy choices with the intent to cure cancer. In this review, we focus on ctDNA as an advanced and standardized validated marker in liquid biopsy. We also discuss what will be needed to reach the new milestone of personalized (precision) medicine to be used as a common standard of care. We summarize recent developments of cell-free DNA (cfDNA) and its clinical use as a biomarker in cancer.
Collapse
Affiliation(s)
- Robert H. Eibl
- c/o M. Schneemann, Department of Internal Medicine, Hospitals of Schaffhausen, 8208 Schaffhausen, Switzerland
| | - Markus Schneemann
- Department of Internal Medicine, Hospitals of Schaffhausen, 8208 Schaffhausen, Switzerland
| |
Collapse
|
224
|
Monitoring Somatic Genetic Alterations in Circulating Cell-Free DNA/RNA of Patients with “Oncogene-Addicted” Advanced Lung Adenocarcinoma: A Real-World Clinical Study. Int J Mol Sci 2022; 23:ijms23158546. [PMID: 35955679 PMCID: PMC9369384 DOI: 10.3390/ijms23158546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Liquid biopsy has advantages over tissue biopsy, but also some technical limitations that hinder its wide use in clinical applications. In this study, we aimed to evaluate the usefulness of liquid biopsy for the clinical management of patients with advanced-stage oncogene-addicted non-small-cell lung adenocarcinomas. The investigation was conducted on a series of cases—641 plasma samples from 57 patients—collected in a prospective consecutive manner, which allowed us to assess the benefits and limitations of the approach in a real-world clinical context. Thirteen samples were collected at diagnosis, and the additional samples during the periodic follow-up visits. At diagnosis, we detected mutations in ctDNA in 10 of the 13 cases (77%). During follow-up, 36 patients progressed. In this subset of patients, molecular analyses of plasma DNA/RNA at progression revealed the appearance of mutations in 29 patients (80.6%). Mutations in ctDNA/RNA were typically detected an average of 80 days earlier than disease progression assessed by RECIST or clinical evaluations. Among the cases positive for mutations, we observed 13 de novo mutations, responsible for the development of resistance to therapy. This study allowed us to highlight the advantages and disadvantages of liquid biopsy, which led to suggesting algorithms for the use of liquid biopsy analyses at diagnosis and during monitoring of therapy response.
Collapse
|
225
|
Zhang L, Sun S, Zhao X, Liu J, Xu Y, Xu L, Song C, Li N, Yu J, Zhao S, Yu P, Fang F, Xie J, Ji X, Yu R, Ou Q, Zhao Z, Li M. Prognostic value of baseline genetic features and newly identified
TP53
mutations in advanced breast cancer. Mol Oncol 2022; 16:3689-3702. [PMID: 35971249 PMCID: PMC9580879 DOI: 10.1002/1878-0261.13297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/30/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Approximately 30% of breast cancer (BC) patients suffer from disease relapse after definitive treatment. Monitoring BC at baseline and disease progression using comprehensive genomic profiling would facilitate the prediction of prognosis. We retrospectively studied 101 BC patients ultimately experiencing relapse and/or metastases. The baseline and circulating tumor DNA‐monitoring cohorts included patients with baseline tumor tissue and serial plasma samples, respectively. Samples were analyzed with targeted next‐generation sequencing of 425 cancer‐relevant genes. Of 35 patients in the baseline cohort, patients with TP53 mutations (P < 0.01), or CTCF/GNAS mutations (P < 0.01) displayed inferior disease‐free survival, and patients harboring TP53 (P = 0.06) or NOTCH1 (P = 0.06) mutations showed relatively poor overall survival (OS), compared to patients with wild‐type counterparts. Of the 59 patients with serial plasma samples, 11 patients who were newly detected with TP53 mutations had worse OS than patients whose TP53 mutational status remained negative (P < 0.01). These results indicate that an inferior prognosis of advanced breast cancer was potentially associated with baseline TP53, CTCF, and NOTCH1 alterations. Newly identified TP53 mutations after relapse and/or metastasis was another potential prognostic biomarker of poor prognosis.
Collapse
Affiliation(s)
- Lanxin Zhang
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Siwen Sun
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Xiaotian Zhao
- Geneseeq Research Institute Nanjing Geneseeq Technology Inc Nanjing Jiangsu China
| | - Jingwen Liu
- Geneseeq Research Institute Nanjing Geneseeq Technology Inc Nanjing Jiangsu China
| | - Yang Xu
- Geneseeq Research Institute Nanjing Geneseeq Technology Inc Nanjing Jiangsu China
| | - Lingzhi Xu
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Chen Song
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Na Li
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Jing Yu
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Shanshan Zhao
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Peiyao Yu
- Department of Oncology First Affiliated Hospital of Dalian Medical University Dalian Liaoning China
| | - Fengqi Fang
- Department of Oncology First Affiliated Hospital of Dalian Medical University Dalian Liaoning China
| | - Jiping Xie
- Department of Breast and Thyroid Surgery Affiliated Zhongshan Hospital of Dalian University Dalian Liaoning China
| | - Xuening Ji
- Department of Oncology Affiliated Zhongshan Hospital of Dalian University Dalian Liaoning China
| | - Ruoying Yu
- Geneseeq Research Institute Nanjing Geneseeq Technology Inc Nanjing Jiangsu China
| | - Qiuxiang Ou
- Geneseeq Research Institute Nanjing Geneseeq Technology Inc Nanjing Jiangsu China
| | - Zuowei Zhao
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
- Department of Breast Surgery The Second Hospital of Dalian Medical University Dalian Liaoning China
| | - Man Li
- Department of Oncology The Second Hospital of Dalian Medical University Dalian Liaoning China
| |
Collapse
|
226
|
Paracchini L, Mannarino L, Beltrame L, Landoni F, Fruscio R, Grassi T, Dalessandro ML, D’Incalci M, Marchini S. Targeted Mutational Analysis of Circulating Tumor DNA to Decipher Temporal Heterogeneity of High-Grade Serous Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14153697. [PMID: 35954363 PMCID: PMC9367609 DOI: 10.3390/cancers14153697] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The issue of spatial and temporal heterogeneity of high-grade serous ovarian cancer (HGS-EOC) has hampered the possibility to shape the molecular portrait of relapsed disease, which ultimately impacts our ability to develop a more rational second-line treatment. Liquid biopsy offers the unique opportunity to track tumor evolution over time and infer the dynamic changes of tumor clonal architecture. Differently from other tumors, no actionable driving lesions characterize HGS-EOC, thus genome-scale analysis like whole-exome sequencing is not compatible with the clinical turnaround time. In the present work, we provided a novel framework based on the analysis of both qualitative and quantitative features of circulating tumor DNA (ctDNA) in order to identify, at the time of molecular relapse, the early genetic vulnerabilities that will characterize the clinical recurrence and thus be amenable of a more rational second-line treatment. Abstract We have previously demonstrated that longitudinal untargeted analysis of plasma samples withdrawn from patients with high-grade serous ovarian cancer (HGS-EOC) can intercept the presence of molecular recurrence (TRm) earlier than the diagnosis of clinical recurrence (TRc). This finding opens a clinical important temporal window to acquire through plasma sample analysis a real-time picture of those emerging molecular lesions that will drive and sustain the growth of relapsed disease and ultimately will confer resistance. In this proof of principle study, the same genomic libraries obtained at the diagnosis (T0), TRm and TRc were further analyzed by targeted resequencing approach to sequence the coding region of a panel of 65 genes to provide longitudinal analysis of clonal evolution as a novel strategy to support clinical decisions for the second-line treatment. Experiments were performed on plasma and tumor tissues withdrawn on a selection of previously analyzed cohorts of cases (i.e., 33 matched primary and synchronous lesions and 43 plasma samples from 18 patients). At T0, the median concordance of mutations shared by each tumor tissue biopsy and its matched plasma sample was 2.27%. This finding confirms the limit of a single tumor biopsy to be representative of the entire disease, while plasma analysis can recapitulate most of the main molecular lesions of the disease. A comparable scenario was observed during longitudinal analysis, where, with the exception of the TP53 gene and germline mutations in BRCA1/2 genes, no other gene shared the same locus specific gene mutation across T0, TRm and TRc time points. This high level of temporal heterogeneity has important implications for planning second-line treatment. For example, in three out of 13 cases, plasma ctDNA analysis at TRm or TRc reported acquired novel variants in the TP53BP1 gene not present at T0. In particular, patient 21564, potentially eligible for PARP-inhibitor (PARPi) treatment at the time of diagnosis (BRCA1 c.5182delA mutation), would unlikely respond to these drugs in second-line therapy due to the presence of eight distinct TP53BP1 variants in plasma samples collected TRc. This study demonstrates that liquid biopsy provides a real-time molecular picture to intercept those actionable genetic vulnerabilities or drug resistance mechanisms that could be used to plan a more rational second-line treatment.
Collapse
Affiliation(s)
- Lara Paracchini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (L.P.); (L.M.)
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.B.); (M.L.D.); (S.M.)
| | - Laura Mannarino
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (L.P.); (L.M.)
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.B.); (M.L.D.); (S.M.)
| | - Luca Beltrame
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.B.); (M.L.D.); (S.M.)
| | - Fabio Landoni
- Department of Obstetrics and Gynaecology, Università degli Studi Milano-Bicocca, San Gerardo Hospital, 20900 Monza, Italy; (F.L.); (R.F.); (T.G.)
| | - Robert Fruscio
- Department of Obstetrics and Gynaecology, Università degli Studi Milano-Bicocca, San Gerardo Hospital, 20900 Monza, Italy; (F.L.); (R.F.); (T.G.)
| | - Tommaso Grassi
- Department of Obstetrics and Gynaecology, Università degli Studi Milano-Bicocca, San Gerardo Hospital, 20900 Monza, Italy; (F.L.); (R.F.); (T.G.)
| | - Maria Luisa Dalessandro
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.B.); (M.L.D.); (S.M.)
| | - Maurizio D’Incalci
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy; (L.P.); (L.M.)
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.B.); (M.L.D.); (S.M.)
- Correspondence: ; Tel.: +39-02-8224-5259
| | - Sergio Marchini
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy; (L.B.); (M.L.D.); (S.M.)
| |
Collapse
|
227
|
Sattar RSA, Verma R, Nimisha, Kumar A, Dar GM, Apurva, Sharma AK, Kumari I, Ahmad E, Ali A, Mahajan B, Saluja SS. Diagnostic and prognostic biomarkers in colorectal cancer and the potential role of exosomes in drug delivery. Cell Signal 2022; 99:110413. [PMID: 35907519 DOI: 10.1016/j.cellsig.2022.110413] [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: 04/14/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/03/2022]
Abstract
Colorectal cancer (CRC) is third most common cancer with second most common cause of death worldwide. One fourth to one fifth of the CRC cases are detected at advance stage. Early detection of colorectal cancer might help in decreasing mortality and morbidity worldwide. CRC being a heterogeneous disease, new non-invasive approaches are needed to complement and improve the screening and management of CRC. Reliable and early detectable biomarkers would improve diagnosis, prognosis, therapeutic responses, and will enable the prediction of drug response and recurrence risk. Over the past decades molecular research has demonstrated the potentials of CTCs, ctDNAs, circulating mRNA, ncRNAs, and exosomes as tumor biomarkers. Non-invasive screening approaches using fecal samples for identification of altered gut microbes in CRC is also gaining attention. Exosomes can be potential candidates that can be employed in the drug delivery system. Further, the integration of in vitro, in vivo and in silico models that involve CRC biomarkers will help to understand the interactions occurring at the cellular level. This review summarizes recent update on CRC biomarkers and their application along with the nanoparticles followed by the application of organoid culture in CRC.
Collapse
Affiliation(s)
- Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Renu Verma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Indu Kumari
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Asgar Ali
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Bhawna Mahajan
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
228
|
Flach S, Kumbrink J, Walz C, Hess J, Drexler G, Belka C, Canis M, Jung A, Baumeister P. Analysis of genetic variants of frequently mutated genes in HPV-negative primary head and neck squamous cell carcinoma, resection margins, local recurrences and corresponding circulating cell-free DNA. J Oral Pathol Med 2022; 51:738-746. [PMID: 35895622 DOI: 10.1111/jop.13338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) remains a substantial burden to global health. Despite evolving therapies, 5-year survival is <50% and unlike in other cancers, reliable molecular biomarkers to guide treatment do not exist. METHODS We performed targeted panel Next Generation Sequencing to analyse somatic variants from primary and recurrent tumour tissue, corresponding resection margins and cell-free DNA from intraoperatively collected plasma samples from 8 patients with HPV-negative HNSCC. Patients were primarily treated with curative-intent surgery and received subsequent adjuvant treatment. RESULTS The most frequently mutated gene was TP53. Other mutated genes included NOTCH1, NF1 and CDKN2A among others. 20.8% of variants were shared between primary tumour and resection margin. Out of all the variants detected, 37.5% were shared between cell-free DNA and primary tumour, whereas 12.5% were commonly found in cell-free DNA, primary tumour and resection margin. Mutational profiling was able to distinguish between a locoregional recurrence and a second primary tumour by identifying a different TP53 mutation in the primary tumour compared to the recurrent tumour in addition to private FBXW7 and CTNNB1 mutations. We also identified identical TP53 and PIK3CA mutations in another primary tumour and corresponding recurrence. CONCLUSION Molecular profiling of cell-free DNA and resection margins has potential applications in clinical practice to guide future treatment decisions.
Collapse
Affiliation(s)
- Susanne Flach
- Department of Otorhinolaryngology, Head and Neck Surgery, LMU Klinikum, Marchioninistrasse 15, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Faculty of Medicine, LMU Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Walz
- Institute of Pathology, Faculty of Medicine, LMU Munich, Germany
| | - Julia Hess
- Clinical Cooperation Group "Personalised Radiotherapy in Head and Neck Cancer", German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Guido Drexler
- Clinical Cooperation Group "Personalised Radiotherapy in Head and Neck Cancer", German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Clinical Cooperation Group "Personalised Radiotherapy in Head and Neck Cancer", German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, Head and Neck Surgery, LMU Klinikum, Marchioninistrasse 15, Munich, Germany.,Clinical Cooperation Group "Personalised Radiotherapy in Head and Neck Cancer", German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Andreas Jung
- Institute of Pathology, Faculty of Medicine, LMU Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, Head and Neck Surgery, LMU Klinikum, Marchioninistrasse 15, Munich, Germany.,Clinical Cooperation Group "Personalised Radiotherapy in Head and Neck Cancer", German Research Center for Environmental Health GmbH, Neuherberg, Germany
| |
Collapse
|
229
|
Imai M, Nakamura Y, Sunami K, Kage H, Komine K, Koyama T, Amano T, Ennishi D, Kanai M, Kenmotsu H, Maeda T, Morita S, Sakai D, Bando H, Makiyama A, Suzuki T, Hirata M, Kohsaka S, Tsuchihara K, Naito Y, Yoshino T. Expert Panel Consensus Recommendations on the Use of Circulating Tumor DNA Assays for Patients with Advanced Solid Tumors. Cancer Sci 2022; 113:3646-3656. [PMID: 35876224 PMCID: PMC9633310 DOI: 10.1111/cas.15504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 12/01/2022] Open
Abstract
Comprehensive genomic profiling is increasingly used to facilitate precision oncology based on molecular stratification. In addition to conventional tissue comprehensive genomic profiling, comprehensive genomic profiling of circulating tumor DNA has become widely utilized in cancer care owing on its advantages, including less invasiveness, rapid turnaround time, and capturing heterogeneity. However, circulating tumor DNA comprehensive genomic profiling has some limitations, mainly false negatives due to low levels of plasma circulating tumor deoxyribonucleic acid and false positives caused by clonal hematopoiesis. Nevertheless, no guidelines and recommendations fully address these issues. Here, an expert panel committee involving representatives from 12 Designated Core Hospitals for Cancer Genomic Medicine in Japan was organized to develop expert consensus recommendations for the use of circulating tumor deoxyribonucleic acid‐based comprehensive genomic profiling. The aim was to generate guidelines for clinicians and allied healthcare professionals on the optimal use of the circulating tumor DNA assays in advanced solid tumors and to aid the design of future clinical trials that utilize and develop circulating tumor DNA assays to refine precision oncology. Fourteen clinical questions regarding circulating tumor deoxyribonucleic acid comprehensive genomic profiling including the timing of testing and considerations for interpreting results were established by searching and curating associated literatures, and corresponding recommendations were prepared based on the literature for each clinical question. Final consensus recommendations were developed by voting to determine the level of each recommendation by the Committee members.
Collapse
Affiliation(s)
- Mitsuho Imai
- Translational Research Support Section, National Cancer Center Hospital East.,Genomics Unit, Keio University School of Medicine
| | - Yoshiaki Nakamura
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital
| | - Hidenori Kage
- Department of Next-Generation Precision Medicine Development Laboratory, Graduate School of Medicine, The University of Tokyo
| | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital
| | - Toraji Amano
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University
| | | | - Takahiro Maeda
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences
| | - Sachi Morita
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital
| | - Daisuke Sakai
- Center for Cancer Genomics and Personalized Medicine, Osaka University Hospital
| | - Hideaki Bando
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| | | | - Tatsuya Suzuki
- Department of Hematology, National Cancer Center Hospital
| | - Makoto Hirata
- Department of Genetic Medicine and Services, National Cancer Center Hospital
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Japan
| | - Yoichi Naito
- Department of General Internal medicine/Experimental Therapeutics/Medical Oncology, National Cancer Center Hospital East
| | - Takayuki Yoshino
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| |
Collapse
|
230
|
Addanki S, Meas S, Sarli VN, Singh B, Lucci A. Applications of Circulating Tumor Cells and Circulating Tumor DNA in Precision Oncology for Breast Cancers. Int J Mol Sci 2022; 23:ijms23147843. [PMID: 35887191 PMCID: PMC9315812 DOI: 10.3390/ijms23147843] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022] Open
Abstract
Liquid biopsies allow for the detection of cancer biomarkers such as circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). Elevated levels of these biomarkers during cancer treatment could potentially serve as indicators of cancer progression and shed light on the mechanisms of metastasis and therapy resistance. Thus, liquid biopsies serve as tools for cancer detection and monitoring through a simple, non-invasive blood draw, allowing multiple longitudinal sampling. These circulating markers have significant prospects for use in assessing patients’ prognosis, monitoring response to therapy, and developing precision medicine. In addition, single-cell omics of these liquid biopsy markers can be potential tools for identifying tumor heterogeneity and plasticity as well as novel therapeutic targets. In this review, we focus on our current understanding of circulating tumor biomarkers, especially in breast cancer, and the scope of novel sequencing technologies and diagnostic methods for better prognostication and patient stratification to improve patient outcomes.
Collapse
Affiliation(s)
- Sridevi Addanki
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
| | - Salyna Meas
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vanessa Nicole Sarli
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Balraj Singh
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anthony Lucci
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.M.); (V.N.S.); (B.S.)
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
231
|
Gonçalves E, Gonçalves-Reis M, Pereira-Leal JB, Cardoso J. DNA methylation fingerprint of hepatocellular carcinoma from tissue and liquid biopsies. Sci Rep 2022; 12:11512. [PMID: 35798798 PMCID: PMC9262906 DOI: 10.1038/s41598-022-15058-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/17/2022] [Indexed: 11/09/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is amongst the cancers with highest mortality rates and is the most common malignancy of the liver. Early detection is vital to provide the best treatment possible and liquid biopsies combined with analysis of circulating tumour DNA methylation show great promise as a non-invasive approach for early cancer diagnosis and monitoring with low false negative rates. To identify reliable diagnostic biomarkers of early HCC, we performed a systematic analysis of multiple hepatocellular studies and datasets comprising > 1500 genome-wide DNA methylation arrays, to define a methylation signature predictive of HCC in both tissue and cell-free DNA liquid biopsy samples. Our machine learning pipeline identified differentially methylated regions in HCC, some associated with transcriptional repression of genes related with cancer progression, that benchmarked positively against independent methylation signatures. Combining our signature of 38 DNA methylation regions, we derived a HCC detection score which confirmed the utility of our approach by identifying in an independent dataset 96% of HCC tissue samples with a precision of 98%, and most importantly successfully separated cfDNA of tumour samples from healthy controls. Notably, our risk score could identify cell-free DNA samples from patients with other tumours, including colorectal cancer. Taken together, we propose a comprehensive HCC DNA methylation fingerprint and an associated risk score for detection of HCC from tissue and liquid biopsies.
Collapse
Affiliation(s)
- Emanuel Gonçalves
- Ophiomics, Pólo Tecnológico de 8, R. Cupertino de Miranda 9, 1600-513, Lisbon, Portugal.,INESC-ID, 1000-029, Lisbon, Portugal
| | - Maria Gonçalves-Reis
- Ophiomics, Pólo Tecnológico de 8, R. Cupertino de Miranda 9, 1600-513, Lisbon, Portugal
| | - José B Pereira-Leal
- Ophiomics, Pólo Tecnológico de 8, R. Cupertino de Miranda 9, 1600-513, Lisbon, Portugal
| | - Joana Cardoso
- Ophiomics, Pólo Tecnológico de 8, R. Cupertino de Miranda 9, 1600-513, Lisbon, Portugal.
| |
Collapse
|
232
|
Mjahed RB, Astaras C, Roth A, Koessler T. Where Are We Now and Where Might We Be Headed in Understanding and Managing Brain Metastases in Colorectal Cancer Patients? Curr Treat Options Oncol 2022; 23:980-1000. [PMID: 35482170 PMCID: PMC9174111 DOI: 10.1007/s11864-022-00982-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 02/01/2023]
Abstract
OPINION STATEMENT Compared to liver and lung metastases, brain metastases (BMs) from colorectal cancer (CRC) are rare and remain poorly investigated despite the anticipated rise in their incidence. CRC patients bearing BM have a dismal prognosis with a median survival of 3-6 months, significantly lower than that of patients with BM from other primary tumors, and of those with metastatic CRC manifesting extracranially. While liver and lung metastases from CRC have more codified treatment strategies, there is no consensus regarding the treatment of BM in CRC, and their management follows the approaches of BM from other solid tumors. Therapeutic strategies are driven by the number and localisation of the lesion, consisting in local treatments such as surgery, stereotactic radiosurgery, or whole-brain radiotherapy. Novel treatment modalities are slowly finding their way into this shy unconsented armatorium including immunotherapy, monoclonal antibodies, tyrosine kinase inhibitors, or a combination of those, among others.This article reviews the pioneering strategies aiming at understanding, diagnosing, and managing this disease, and discusses future directions, challenges, and potential innovations in each of these domains. HIGHLIGHTS • With the increasing survival in CRC, brain and other rare/late-onset metastases are rising. • Distal colon/rectal primary location, long-standing progressive lung metastases, and longer survival are risk factors for BM development in CRC. • Late diagnosis and lack of consensus treatment strategies make BM-CRC diagnosis very dismal. • Liquid biopsies using circulating tumor cells might offer excellent opportunities in the early diagnosis of BM-CRC and the search for therapeutic options. • Multi-modality treatment including surgical metastatic resection, postoperative SRS with/without WBRT, and chemotherapy is the best current treatment option. • Recent mid-sized clinical trials, case reports, and preclinical models show the potential of unconventional therapeutic approaches as monoclonal antibodies, targeted therapies, and immunotherapy. Graphical abstract.
Collapse
Affiliation(s)
- Ribal Bou Mjahed
- Department of Oncology, University hospital of Geneva (HUG), Geneva, Switzerland.
- Département de médecine interne - CHUV, Rue du Bugnon 21, CH-1011, Lausanne, Switzerland.
| | - Christoforos Astaras
- Department of Oncology, University hospital of Geneva (HUG), Geneva, Switzerland
| | - Arnaud Roth
- Department of Oncology, University hospital of Geneva (HUG), Geneva, Switzerland
| | - Thibaud Koessler
- Department of Oncology, University hospital of Geneva (HUG), Geneva, Switzerland
| |
Collapse
|
233
|
Igari F, Tanaka H, Giuliano AE. The applications of plasma cell-free DNA in cancer detection: Implications in the management of breast cancer patients. Crit Rev Oncol Hematol 2022; 175:103725. [PMID: 35618229 DOI: 10.1016/j.critrevonc.2022.103725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/28/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022] Open
Abstract
Liquid biopsy probes DNA, RNA, and proteins in body fluids for cancer detection and is one of the most rapidly developing areas in oncology. Tumor-derived DNA (circulating tumor DNA, ctDNA) in the context of cell-free DNA (cfDNA) in blood has been the main target for its potential utilities in cancer detection. Liquid biopsy can report tumor burden in real-time without invasive interventions, and would be feasible for screening tumor types that lack standard-of-care screening approaches. Two major approaches to interrogating ctDNA are genetic mutation and DNA methylation profiling. Mutation profiling can identify tumor driver mutations and guide precision therapy. Targeted genomic profiling of DNA methylation has become the main approach for cancer screening in the general population. Here we review the recent technological development and ongoing efforts in clinical applications. For clinical applications, we focus on breast cancer, in which subtype-specific biology demarcates the applications of ctDNA.
Collapse
Affiliation(s)
- Fumie Igari
- Department of Surgery, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA; Department of Breast Oncology, Juntendo University, Tokyo, Japan
| | - Hisashi Tanaka
- Department of Surgery, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute and Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA; Biomedical Sciences, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA.
| | - Armando E Giuliano
- Department of Surgery, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute and Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA; Biomedical Sciences, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA
| |
Collapse
|
234
|
Tutanov O, Tamkovich S. The Influence of Proteins on Fate and Biological Role of Circulating DNA. Int J Mol Sci 2022; 23:7224. [PMID: 35806228 PMCID: PMC9266439 DOI: 10.3390/ijms23137224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Circulating DNA has already proven itself as a valuable tool in translational medicine. However, one of the overlooked areas of circulating DNA research is its association with different proteins, despite considerable evidence that this association might impact DNA's fate in circulation and its biological role. In this review, we attempt to shed light on current ideas about circulating DNA origins and forms of circulation, known biological effects, and the clinical potential of circulating tumor deoxyribonucleoprotein complexes.
Collapse
Affiliation(s)
| | - Svetlana Tamkovich
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, 630090 Novosibirsk, Russia;
| |
Collapse
|
235
|
Pre-PCR Mutation-Enrichment Methods for Liquid Biopsy Applications. Cancers (Basel) 2022; 14:cancers14133143. [PMID: 35804916 PMCID: PMC9264780 DOI: 10.3390/cancers14133143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 01/25/2023] Open
Abstract
Liquid biopsy is having a remarkable impact on healthcare- and disease-management in the context of personalized medicine. Circulating free DNA (cfDNA) is one of the most instructive liquid-biopsy-based biomarkers and harbors valuable information for diagnostic, predictive, and prognostic purposes. When it comes to cancer, circulating DNA from the tumor (ctDNA) has a wide range of applications, from early cancer detection to the early detection of relapse or drug resistance, and the tracking of the dynamic genomic make-up of tumor cells. However, the detection of ctDNA remains technically challenging, due, in part, to the low frequency of ctDNA among excessive circulating cfDNA originating from normal tissues. During the past three decades, mutation-enrichment methods have emerged to boost sensitivity and enable facile detection of low-level mutations. Although most developed techniques apply mutation enrichment during or following initial PCR, there are a few techniques that allow mutation selection prior to PCR, which provides advantages. Pre-PCR enrichment techniques can be directly applied to genomic DNA and diminish the influence of PCR errors that can take place during amplification. Moreover, they have the capability for high multiplexity and can be followed by established mutation detection and enrichment technologies without changes to their established procedures. The first approaches for pre-PCR enrichment were developed by employing restriction endonucleases directly on genomic DNA in the early 1990s. However, newly developed pre-PCR enrichment methods provide higher sensitivity and versatility. This review describes the available pre-PCR enrichment methods and focuses on the most recently developed techniques (NaME-PrO, UVME, and DEASH/MAESTRO), emphasizing their applications in liquid biopsies.
Collapse
|
236
|
Lin RK, Su CM, Lin SY, Thi Anh Thu L, Liew PL, Chen JY, Tzeng HE, Liu YR, Chang TH, Lee CY, Hung CS. Hypermethylation of TMEM240 predicts poor hormone therapy response and disease progression in breast cancer. Mol Med 2022; 28:67. [PMID: 35715741 PMCID: PMC9204905 DOI: 10.1186/s10020-022-00474-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 12/16/2022] Open
Abstract
Background Approximately 25% of patients with early-stage breast cancer experience cancer progression throughout the disease course. Alterations in TMEM240 in breast cancer were identified and investigated to monitor treatment response and disease progression. Methods Circulating methylated TMEM240 in the plasma of breast cancer patients was used to monitor treatment response and disease progression. The Cancer Genome Atlas (TCGA) data in Western countries and Illumina methylation arrays in Taiwanese breast cancer patients were used to identify novel hypermethylated CpG sites and genes related to poor hormone therapy response. Quantitative methylation-specific PCR (QMSP), real-time reverse transcription PCR, and immunohistochemical analyses were performed to measure DNA methylation and mRNA and protein expression levels in 394 samples from Taiwanese and Korean breast cancer patients. TMEM240 gene manipulation, viability, migration assays, RNA-seq, and MetaCore were performed to determine its biological functions and relationship to hormone drug treatment response in breast cancer cells. Results Aberrant methylated TMEM240 was identified in breast cancer patients with poor hormone therapy response using genome-wide methylation analysis in the Taiwan and TCGA breast cancer cohorts. A cell model showed that TMEM240, which is localized to the cell membrane and cytoplasm, represses breast cancer cell proliferation and migration and regulates the expression levels of enzymes involved in estrone and estradiol metabolism. TMEM240 protein expression was observed in normal breast tissues but was not detected in 88.2% (67/76) of breast tumors and in 90.0% (9/10) of metastatic tumors from breast cancer patients. QMSP revealed that in 54.5% (55/101) of Taiwanese breast cancer patients, the methylation level of TMEM240 was at least twofold higher in tumor tissues than in matched normal breast tissues. Patients with hypermethylation of TMEM240 had poor 10-year overall survival (p = 0.003) and poor treatment response, especially hormone therapy response (p < 0.001). Circulating methylated TMEM240 dramatically and gradually decreased and then diminished in patients without disease progression, whereas it returned and its levels in plasma rose again in patients with disease progression. Prediction of disease progression based on circulating methylated TMEM240 was found to have 87.5% sensitivity, 93.1% specificity, and 90.2% accuracy. Conclusions Hypermethylation of TMEM240 is a potential biomarker for treatment response and disease progression monitoring in breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00474-9.
Collapse
Affiliation(s)
- Ruo-Kai Lin
- Program in Drug Discovery and Development Industry, Program in Clinical Drug Development of Herbal Medicine, Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Graduate Institute of Pharmacognosy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan.,Clinical Trial Center, Taipei Medical University Hospital, 252 Wu-Hsing Street, Taipei, Taiwan
| | - Chih-Ming Su
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Yun Lin
- Program in Drug Discovery and Development Industry, Program in Clinical Drug Development of Herbal Medicine, Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Graduate Institute of Pharmacognosy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Le Thi Anh Thu
- Quang Tri Medical College, Dien Bien Phu Str., Dong Luong District, Dong Ha City, Quang Tri, Vietnam
| | - Phui-Ly Liew
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jian-Yu Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Huey-En Tzeng
- Division of Hematology and Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Medical Research, Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan.,Program for Cancer Molecular Biology and Drug Discovery, and Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yang Lee
- Bioinformatics Center, Office of Data Science, Taipei Medical University, Taipei, Taiwan
| | - Chin-Sheng Hung
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan. .,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
237
|
Aulakh SS, Silverman DA, Young K, Dennis SK, Birkeland AC. The Promise of Circulating Tumor DNA in Head and Neck Cancer. Cancers (Basel) 2022; 14:2968. [PMID: 35740633 PMCID: PMC9221491 DOI: 10.3390/cancers14122968] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/17/2022] Open
Abstract
As the seventh most common cancer globally, head and neck cancers (HNC) exert considerable disease burden, with an estimated 277,597 deaths worldwide in 2020 alone. Traditional risk factors for HNC include tobacco, alcohol, and betel nut; more recently, human papillomavirus has emerged as a distinct driver of disease. Currently, limitations of cancer screening and surveillance methods often lead to identifying HNC in more advanced stages, with associated poor outcomes. Liquid biopsies, in particular circulating tumor DNA (ctDNA), offer the potential for enhancing screening, early diagnosis, and surveillance in HNC patients, with potential improvements in HNC patient outcomes. In this review, we examine current methodologies for detecting ctDNA and highlight current research illustrating viral and non-viral ctDNA biomarker utilities in HNC screening, diagnosis, treatment response, and prognosis. We also summarize current challenges and future directions for ctDNA testing in HNC patients.
Collapse
Affiliation(s)
| | - Dustin A. Silverman
- Department of Otolaryngology—Head and Neck Surgery, University of California, Davis, CA 95817, USA; (D.A.S.); (S.K.D.)
| | - Kurtis Young
- John A. Burns School of Medicine, Honolulu, HI 96813, USA;
| | - Steven K. Dennis
- Department of Otolaryngology—Head and Neck Surgery, University of California, Davis, CA 95817, USA; (D.A.S.); (S.K.D.)
| | - Andrew C. Birkeland
- Department of Otolaryngology—Head and Neck Surgery, University of California, Davis, CA 95817, USA; (D.A.S.); (S.K.D.)
| |
Collapse
|
238
|
Madanat-Harjuoja LM, Klega K, Lu Y, Shulman DS, Thorner AR, Nag A, Tap WD, Reinke DK, Diller L, Ballman KV, George S, Crompton BD. Circulating Tumor DNA Is Associated with Response and Survival in Patients with Advanced Leiomyosarcoma. Clin Cancer Res 2022; 28:2579-2586. [PMID: 35561344 PMCID: PMC9359745 DOI: 10.1158/1078-0432.ccr-21-3951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/21/2021] [Accepted: 02/17/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE We sought to determine whether the detection of circulating tumor DNA (ctDNA) in samples of patients undergoing chemotherapy for advanced leiomyosarcoma (LMS) is associated with objective response or survival. EXPERIMENTAL DESIGN Using ultra-low-passage whole-genome sequencing (ULP-WGS) of plasma cell-free DNA from patients treated on a prospective clinical trial, we tested whether detection of ctDNA evaluated prior to the start of therapy and after two cycles of chemotherapy was associated with treatment response and outcome. Associations between detection of ctDNA and pathologic measures of disease burden were evaluated. RESULTS We found that ctDNA was detectable by ULP-WGS in 49% patients prior to treatment and in 24.6% patients after two cycles of chemotherapy. Detection of pretreatment ctDNA was significantly associated with a lower overall survival [HR, 1.55; 95% confidence interval (CI), 1.03-2.31; P = 0.03] and a significantly lower likelihood of objective response [odds ratio (OR), 0.21; 95% CI, 0.06-0.59; P = 0.005]. After two cycles of chemotherapy, patients who continued to have detectable levels of ctDNA experienced a significantly worse overall survival (HR, 1.77; 95% CI, 1-3.14; P = 0.05) and were unlikely to experience an objective response (OR, 0.05; 95% CI, 0-0.39; P = 0.001). CONCLUSIONS Our results demonstrate that detection of ctDNA is associated with outcome and objective response to chemotherapy in patients with advanced LMS. These results suggest that liquid biopsy assays could be used to inform treatment decisions by recognizing patients who are likely and unlikely to benefit from chemotherapy. See related commentary by Kasper and Wilky, p. 2480.
Collapse
Affiliation(s)
| | - Kelly Klega
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Yao Lu
- Weill Cornell Medicine, New York, New York
| | - David S. Shulman
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Aaron R. Thorner
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anwesha Nag
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - William D. Tap
- Weill Cornell Medicine, New York, New York.,Memorial Sloan Kettering Cancer Center, New York, New York
| | - Denise K. Reinke
- University of Michigan, Department of Internal Medicine, Ann Arbor, Michigan
| | - Lisa Diller
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | | | - Suzanne George
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Brian D. Crompton
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Corresponding Author: Brian D. Crompton, Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, 450 Brookline Avenue, Boston, MA 02215. E-mail:
| |
Collapse
|
239
|
Hsiao YP, Mukundan A, Chen WC, Wu MT, Hsieh SC, Wang HC. Design of a Lab-On-Chip for Cancer Cell Detection through Impedance and Photoelectrochemical Response Analysis. BIOSENSORS 2022; 12:bios12060405. [PMID: 35735553 PMCID: PMC9221223 DOI: 10.3390/bios12060405] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 05/07/2023]
Abstract
In this study, a biochip was fabricated using a light-absorbing layer of a silicon solar element combined with serrated, interdigitated electrodes and used to identify four different types of cancer cells: CE81T esophageal cancer, OE21 esophageal cancer, A549 lung adenocarcinoma, and TSGH-8301 bladder cancer cells. A string of pearls was formed from dielectrophoretic aggregated cancer cells because of the serrated interdigitated electrodes. Thus, cancer cells were identified in different parts, and electron-hole pairs were separated by photo-excited carriers through the light-absorbing layer of the solar element. The concentration catalysis mechanism of GSH and GSSG was used to conduct photocurrent response and identification, which provides the fast, label-free measurement of cancer cells. The total time taken for this analysis was 13 min. Changes in the impedance value and photocurrent response of each cancer cell were linearly related to the number of cells, and the slope of the admittance value was used to distinguish the location of the cancerous lesion, the slope of the photocurrent response, and the severity of the cancerous lesion. The results show that the number of cancerous cells was directly proportional to the admittance value and the photocurrent response for all four different types of cancer cells. Additionally, different types of cancer cells could easily be differentiated using the slope value of the photocurrent response and the admittance value.
Collapse
Affiliation(s)
- Yu-Ping Hsiao
- Department of Dermatology, Chung Shan Medical University Hospital, No.110, Sec. 1, Jianguo N. Rd., South District, Taichung City 40201, Taiwan;
- Institute of Medicine, School of Medicine, Chung Shan Medical University, No.110, Sec. 1, Jianguo N. Rd., South District, Taichung City 40201, Taiwan
| | - Arvind Mukundan
- Department of Mechanical Engineering, Advanced Institute of Manufacturing with High Tech Innovations (AIM-HI), Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, 168, University Rd., Min Hsiung, Chia Yi 62102, Taiwan;
| | - Wei-Chung Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (W.-C.C.); (M.-T.W.)
| | - Ming-Tsang Wu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807377, Taiwan; (W.-C.C.); (M.-T.W.)
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
- Department of Public Health, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan
| | - Shang-Chin Hsieh
- Department of Plastic Surgery, Kaohsiung Armed Forces General Hospital, 2, Zhongzheng 1st Rd., Lingya District, Kaohsiung 80284, Taiwan
- Correspondence: (S.-C.H.); (H.-C.W.)
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, Advanced Institute of Manufacturing with High Tech Innovations (AIM-HI), Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, 168, University Rd., Min Hsiung, Chia Yi 62102, Taiwan;
- Correspondence: (S.-C.H.); (H.-C.W.)
| |
Collapse
|
240
|
Tabata M, Khamhanglit C, Kotaki S, Miyahara Y. Detection of cell membrane proteins using ion-sensitive field effect transistors combined with chemical signal amplification. Chem Commun (Camb) 2022; 58:7368-7371. [PMID: 35686960 DOI: 10.1039/d2cc02159e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The capture and detection of cells expressing a breast-cancer related membrane protein, namely a BT474 cell line expressing HER2, is demonstrated using ion-sensitive field effect transistors (ISFETs). BT474 cells were exposed to anti-HER2 antibodies and urease-conjugated secondary antibodies to induce chemical signal amplification by adding urea.
Collapse
Affiliation(s)
- Miyuki Tabata
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan.
| | - Chattarika Khamhanglit
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan.
| | - Sayo Kotaki
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan.
| | - Yuji Miyahara
- Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan.
| |
Collapse
|
241
|
Pascual J, Attard G, Bidard FC, Curigliano G, De Mattos-Arruda L, Diehn M, Italiano A, Lindberg J, Merker JD, Montagut C, Normanno N, Pantel K, Pentheroudakis G, Popat S, Reis-Filho JS, Tie J, Seoane J, Tarazona N, Yoshino T, Turner NC. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2022; 33:750-768. [PMID: 35809752 DOI: 10.1016/j.annonc.2022.05.520] [Citation(s) in RCA: 274] [Impact Index Per Article: 91.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 12/16/2022] Open
Abstract
Circulating tumour DNA (ctDNA) assays conducted on plasma are rapidly developing a strong evidence base for use in patients with cancer. The European Society for Medical Oncology convened an expert working group to review the analytical and clinical validity and utility of ctDNA assays. For patients with advanced cancer, validated and adequately sensitive ctDNA assays have utility in identifying actionable mutations to direct targeted therapy, and may be used in routine clinical practice, provided the limitations of the assays are taken into account. Tissue based testing remains the preferred test for many cancer patients, due to limitations of ctDNA assays detecting fusion events and copy number changes, although ctDNA assays may be routinely used when faster results will be clinically important, or when tissue biopsies are not possible or inappropriate. Reflex tumour testing should be considered following a non-informative ctDNA result, due to false negative results with ctDNA testing. In patients treated for early-stage cancers, detection of molecular residual disease (MRD) or molecular relapse (MR), has high evidence of clinical validity in anticipating future relapse in many cancers. MRD/MR detection cannot be recommended in routine clinical practice, as currently there is no evidence for clinical utility in directing treatment. Additional potential applications of ctDNA assays, under research development and not recommended for routine practice, include identifying patients not responding to therapy with early dynamic changes in ctDNA levels, monitoring therapy for the development of resistance mutations prior to clinical progression, and in screening asymptomatic people for cancer. Recommendation for reporting of results, future development of ctDNA assays, and future clinical research are made.
Collapse
Affiliation(s)
- Javier Pascual
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA, Malaga, Spain
| | - Gerhardt Attard
- Urological Cancer Research, University College London, London, UK
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, Paris, France; University of Versailles Saint-Quentin-en-Yvelines (UVSQ)/Paris-Saclay University, Saint Cloud, France
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Milano, Italy
| | - Leticia De Mattos-Arruda
- IrsiCaixa, Hospital Universitari Trias i Pujol, Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, US
| | - Antoine Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonie, Bordeaux, France; DITEP, Gustave Roussy, Villejuif, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Jason D Merker
- Departments of Pathology and Laboratory Medicine & Genetics, UNC School of Medicine, Chapel Hill, NC, US
| | - Clara Montagut
- Medical Oncology Department, Hospital del Mar-IMIM, CIBERONC, Universitat Pompeu Fabra, Barcelona, Spain
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, 'Fondazione G. Pascale' - IRCCS, Naples, Italy
| | - Klaus Pantel
- Institute for Tumour Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - George Pentheroudakis
- Scientific and Medical Division, European Society for Medical Oncology, Lugano, Switzerland
| | - Sanjay Popat
- Royal Marsden Hospital, London, UK; Institute of Cancer Research, London, UK
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, US
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Division of Personalised Oncology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Joan Seoane
- Preclinical and Translational Research Programme, Vall d'Hebron Institute of Oncology (VHIO), ICREA, CIBERONC, Barcelona, Spain,; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Noelia Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Nicholas C Turner
- Royal Marsden Hospital, London, UK; Institute of Cancer Research, London, UK
| |
Collapse
|
242
|
Woof VG, Lee RJ, Lorigan P, French DP. Circulating tumour DNA monitoring and early treatment for relapse: views from patients with early-stage melanoma. Br J Cancer 2022; 126:1450-1456. [PMID: 35301436 PMCID: PMC8927744 DOI: 10.1038/s41416-022-01766-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Circulating tumour DNA (ctDNA) monitoring is a new technology that detects cancer DNA fragments in blood circulation. Regular monitoring with ctDNA has the potential to detect and treat cancer relapse earlier, but there is little evidence on patient acceptability. This study examines the views of patients with early-stage melanoma on the acceptability of the test and early treatment. METHODS A qualitative cross-sectional design using one-to-one semi-structured telephone interviews was employed. Twenty-five patients diagnosed with early-stage melanoma (Stage IA-IIC) were asked for their views on ctDNA monitoring and early treatment for relapse. Interviews were analysed using reflexive thematic analysis. RESULTS Two themes were generated: ctDNA monitoring would add service value where participants described regular ctDNA monitoring in follow-up care as more reassuring, more "scientific" than skin checks and preferable to scans. Test results provide opportunity and knowledge focuses on how participants wanted to know when to expect results to manage anxiety, with a positive result seen as an opportunity to receive treatment early. CONCLUSIONS Participants were positive about ctDNA monitoring and early treatment and would welcome extra surveillance, as well as trust ctDNA tests. This indicates the feasibility of the wider implementation of ctDNA tests, which have applicability for many tumour types and disease stages.
Collapse
Affiliation(s)
- Victoria G Woof
- Manchester Centre of Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| | - Rebecca J Lee
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, Manchester, UK
- Faculty of Biology Medicine and Health, The University of Manchester, Oxford Road, M13 9PL, Manchester, UK
| | - Paul Lorigan
- The Christie NHS Foundation Trust, Wilmslow Road, M20 4BX, Manchester, UK
- Faculty of Biology Medicine and Health, The University of Manchester, Oxford Road, M13 9PL, Manchester, UK
| | - David P French
- Manchester Centre of Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| |
Collapse
|
243
|
Wang Y, Li Y, Liang X, Xin S, Yang L, Cao P, Jiang M, Xin Y, Zhang S, Yang Y, Lu J. The implications of cell-free DNAs derived from tumor viruses as biomarkers of associated cancers. J Med Virol 2022; 94:4677-4688. [PMID: 35652186 DOI: 10.1002/jmv.27903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022]
Abstract
Cancer is still ranked as a leading cause of death according to estimates from the World Health Organization (WHO) and the strong link between tumor viruses and human cancers have been proved for almost six decades. Cell-free DNA (cfDNA) has drawn enormous attention for its dynamic, instant, and noninvasive advantages as one popular type of cancer biomarker. cfDNAs are mainly released from apoptotic cells and exosomes released from cancer cells, including those infected with viruses. Although cfDNAs are present at low concentrations in peripheral blood, they can reflect tumor load with high sensitivity. Considering the relevance of the tumor viruses to the associated cancers, cfDNAs derived from viruses may serve as good biomarkers for the early screening, diagnosis, and treatment monitoring. In this review, we summarize the methods and newly developed analytic techniques for the detection of cfDNAs from different body fluids, and discuss the implications of cfDNAs derived from different tumor viruses in the detection and treatment monitoring of virus-associated cancers. A better understanding of cfDNAs derived from tumor viruses may help formulate novel anti-tumoral strategies to decrease the burden of cancers that attributed to viruses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yiwei Wang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Yanling Li
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Xinyu Liang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China
| | - Shuyu Xin
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Li Yang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Pengfei Cao
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China
| | - Mingjuan Jiang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Yujie Xin
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Senmiao Zhang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Yang Yang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Jianhong Lu
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China.,NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China.,Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| |
Collapse
|
244
|
Nkosi D, Miller CA, Jajosky AN, Oltvai ZN. Incidental discovery of acute myeloid leukemia during liquid biopsy of a lung cancer patient. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006201. [PMID: 35732498 PMCID: PMC9235846 DOI: 10.1101/mcs.a006201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/25/2022] [Indexed: 11/25/2022] Open
Abstract
Liquid biopsy is considered an alternative to standard next-generation sequencing (NGS) of solid tumor samples when biopsy tissue is inadequate for testing or when testing of a peripheral blood sample is preferred. A common assumption of liquid biopsies is that the NGS data obtained on circulating cell-free DNA is a high-fidelity reflection of what would be found by solid tumor testing. Here, we describe a case that challenges this widely held assumption. A patient diagnosed with lung carcinoma showed pathogenic IDH1 and TP53 mutations by liquid biopsy NGS at an outside laboratory. Subsequent in-house NGS of a metastatic lymph node fine-needle aspiration (FNA) sample revealed two pathogenic EGFR mutations. Morphologic and immunophenotypic assessment of the patient's blood sample identified acute myeloid leukemia, with in-house NGS confirming and identifying pathogenic IDH1, TP53, and BCOR mutations, respectively. This case, together with a few similar reports, demonstrates that caution is needed when interpreting liquid biopsy NGS results, especially if they are inconsistent with the presumptive diagnosis. Our case suggests that routine parallel sequencing of peripheral white blood cells would substantially increase the fidelity of the obtained liquid biopsy results.
Collapse
Affiliation(s)
- Dingani Nkosi
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Caroline A Miller
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Audrey N Jajosky
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Zoltán N Oltvai
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| |
Collapse
|
245
|
Li CL, Yeh SH, Chen PJ. Circulating Virus–Host Chimera DNAs in the Clinical Monitoring of Virus-Related Cancers. Cancers (Basel) 2022; 14:cancers14102531. [PMID: 35626135 PMCID: PMC9139492 DOI: 10.3390/cancers14102531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cell-free tumor DNA (ctDNA), the DNA released into circulation from tumors, is a promising tumor marker with versatile applications. The associations of the amount, somatic mutation frequency, and epigenetic modifications of ctDNA with the tumor burden, tumor behavior, and prognosis have been widely investigated in different types of tumors. However, there are still some challenging issues to be resolved before ctDNA can complement or even replace current serum tumor markers. We propose employing exogenous viral DNA integration that produces unique virus–host chimera DNA (vh-DNA) at junction sites. Cell-free vh-DNA may become a new biomarker because it overcomes background interference detection problems, takes advantage of virus tropism to localize the tumor, and acts as a universal marker for monitoring clonal expansion or tumor loads in tumors related to oncogenic viruses. Abstract The idea of using tumor-specific cell-free DNA (ctDNA) as a tumor biomarker has been widely tested and validated in various types of human cancers and different clinical settings. ctDNA can reflect the presence or size of tumors in a real-time manner and can enable longitudinal monitoring with minimal invasiveness, allowing it to be applied in treatment response assessment and recurrence monitoring for cancer therapies. However, tumor detection by ctDNA remains a great challenge due to the difficulty in enriching ctDNA from a large amount of homologous non-tumor cell-free DNA (cfDNA). Only ctDNA with nonhuman sequences (or rearrangements) can be selected from the background of cfDNA from nontumor DNAs. This is possible for several virus-related cancers, such as hepatitis B virus (HBV)-related HCC or human papillomavirus (HPV)-related cervical or head and neck cancers, which frequently harbor randomly integrated viral DNA. The junction fragments of the integrations, namely virus–host chimera DNA (vh-DNA), can represent the signatures of individual tumors and are released into the blood. Such ctDNA can be enriched by capture with virus-specific probes and therefore exploited as a circulating biomarker to track virus-related cancers in clinical settings. Here, we review virus integrations in virus-related cancers to evaluate the feasibility of vh-DNA as a cell-free tumor marker and update studies on the development of detection and applications. vh-DNA may be a solution to the development of specific markers to manage virus-related cancers in the future.
Collapse
Affiliation(s)
- Chiao-Ling Li
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Shiou-Hwei Yeh
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Correspondence: (S.-H.Y.); (P.-J.C.)
| | - Pei-Jer Chen
- Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Correspondence: (S.-H.Y.); (P.-J.C.)
| |
Collapse
|
246
|
Yaung SJ, Woestmann C, Ju C, Ma XM, Gattam S, Zhou Y, Xi L, Pal S, Balasubramanyam A, Tikoo N, Heussel CP, Thomas M, Kriegsmann M, Meister M, Schneider MA, Herth FJ, Wehnl B, Diehn M, Alizadeh AA, Palma JF, Muley T. Early Assessment of Chemotherapy Response in Advanced Non-Small Cell Lung Cancer with Circulating Tumor DNA. Cancers (Basel) 2022; 14:cancers14102479. [PMID: 35626082 PMCID: PMC9139958 DOI: 10.3390/cancers14102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 11/16/2022] Open
Abstract
Monitoring treatment efficacy early during therapy could enable a change in treatment to improve patient outcomes. We report an early assessment of response to treatment in advanced NSCLC using a plasma-only strategy to measure changes in ctDNA levels after one cycle of chemotherapy. Plasma samples were collected from 92 patients with Stage IIIB-IV NSCLC treated with first-line chemo- or chemoradiation therapies in an observational, prospective study. Retrospective ctDNA analysis was performed using next-generation sequencing with a targeted 198-kb panel designed for lung cancer surveillance and monitoring. We assessed whether changes in ctDNA levels after one or two cycles of treatment were associated with clinical outcomes. Subjects with ≤50% decrease in ctDNA level after one cycle of chemotherapy had a lower 6-month progression-free survival rate (33% vs. 58%, HR 2.3, 95% CI 1.2 to 4.2, log-rank p = 0.009) and a lower 12-month overall survival rate (25% vs. 70%, HR 4.3, 95% CI 2.2 to 9.7, log-rank p < 0.001). Subjects with ≤50% decrease in ctDNA level after two cycles of chemotherapy also had shorter survival. Using non-invasive liquid biopsies to measure early changes in ctDNA levels in response to chemotherapy may help identify non-responders before standard-of-care imaging in advanced NSCLC.
Collapse
Affiliation(s)
- Stephanie J. Yaung
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
- Correspondence: ; Tel.: +1-925-523-8824
| | | | - Christine Ju
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Xiaoju Max Ma
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
| | - Sandeep Gattam
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Yiyong Zhou
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Liu Xi
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
| | - Subrata Pal
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Aarthi Balasubramanyam
- Roche Molecular Systems, Inc., Pleasanton, CA 94588, USA; (C.J.); (S.G.); (Y.Z.); (S.P.); (A.B.)
| | - Nalin Tikoo
- Alector, Inc., South San Francisco, CA 94080, USA;
| | - Claus Peter Heussel
- Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik, University Hospital, 69126 Heidelberg, Germany;
- Diagnostic and Interventional Radiology, University Hospital, 69120 Heidelberg, Germany
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
| | - Michael Thomas
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Michael Meister
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Marc A. Schneider
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Felix J. Herth
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| | - Birgit Wehnl
- Roche Diagnostics GmbH, 82377 Penzberg, Germany;
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.A.A.)
| | - Ash A. Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; (M.D.); (A.A.A.)
| | - John F. Palma
- Roche Sequencing Solutions, Inc., Pleasanton, CA 94588, USA; (X.M.M.); (L.X.); (J.F.P.)
| | - Thomas Muley
- Translational Lung Research Centre (TLRC) Heidelberg, Member of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany; (M.T.); (M.M.); (M.A.S.); (F.J.H.); (T.M.)
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany
| |
Collapse
|
247
|
Yan X, Liu C. Application of Non-Blood-Derived Fluid Biopsy in Monitoring Minimal Residual Diseases of Lung Cancer. Front Surg 2022; 9:865040. [PMID: 35651679 PMCID: PMC9149287 DOI: 10.3389/fsurg.2022.865040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/28/2022] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is one of the most fatal malignant tumors in the world. Overcoming this disease is difficult due to its late diagnosis and relapse after treatment. Minimal residual disease (MRD) is described as the presence of free circulating tumor cells or other tumor cell derivatives in the biological fluid of patients without any clinical symptoms of cancer and negative imaging examination after the treatment of primary tumors. It has been widely discussed in the medical community as a bridge to solid tumor recurrence. Radiology, serology (carcinoembryonic antigen), and other clinical diagnosis and treatment methods widely used to monitor the progression of disease recurrence have obvious time-limited and -specific defects. Furthermore, as most samples of traditional liquid biopsies come from patients’ blood (including plasma and serum), the low concentration of tumor markers in blood samples limits the ability of these liquid biopsies in the early detection of cancer recurrence. The use of non-blood-derived fluid biopsy in monitoring the status of MRD and further improving the postoperative individualized treatment of patients with lung cancer is gradually ushering in the dawn of hope. This paper reviews the progress of several non-blood-derived fluid samples (urine, saliva, sputum, and pleural effusion) in detecting MRD in lung cancer as well as selecting the accurate treatment for it.
Collapse
Affiliation(s)
- Xing Yan
- Thoracic Surgery Department, The Second Affiliated Hospital of Dalian Medical University Thoracic surgery, DaLian, China
| | - Changhong Liu
- Thoracic Surgery Department, The Second Affiliated Hospital of Dalian Medical University Thoracic surgery, DaLian, China
- Correspondence: Changhong Liu
| |
Collapse
|
248
|
Merz V, Mangiameli D, Zecchetto C, Quinzii A, Pietrobono S, Messina C, Casalino S, Gaule M, Pesoni C, Vitale P, Trentin C, Frisinghelli M, Caffo O, Melisi D. Predictive Biomarkers for a Personalized Approach in Resectable Pancreatic Cancer. Front Surg 2022; 9:866173. [PMID: 35599791 PMCID: PMC9114435 DOI: 10.3389/fsurg.2022.866173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022] Open
Abstract
The mainstay treatment for patients with immediate resectable pancreatic cancer remains upfront surgery, which represents the only potentially curative strategy. Nevertheless, the majority of patients surgically resected for pancreatic cancer experiences disease relapse, even when a combination adjuvant therapy is offered. Therefore, aiming at improving disease free survival and overall survival of these patients, there is an increasing interest in evaluating the activity and efficacy of neoadjuvant and perioperative treatments. In this view, it is of utmost importance to find biomarkers able to select patients who may benefit from a preoperative therapy rather than upfront surgical resection. Defined genomic alterations and a dynamic inflammatory microenvironment are the major culprits for disease recurrence and resistance to chemotherapeutic treatments in pancreatic cancer patients. Signal transduction pathways or tumor immune microenvironment could predict early recurrence and response to chemotherapy. In the last decade, distinct molecular subtypes of pancreatic cancer have been described, laying the bases to a tailored therapeutic approach, started firstly in the treatment of advanced disease. Patients with homologous repair deficiency, in particular with mutant germline BRCA genes, represent the first subgroup demonstrating to benefit from specific therapies. A fraction of patients with pancreatic cancer could take advantage of genome sequencing with the aim of identifying possible targetable mutations. These genomic driven strategies could be even more relevant in a potentially curative setting. In this review, we outline putative predictive markers that could help in the next future in tailoring the best therapeutic strategy for pancreatic cancer patients with a potentially curable disease.
Collapse
Affiliation(s)
- Valeria Merz
- Medical Oncology Unit, Santa Chiara Hospital, Trento, Italy
- Digestive Molecular Clinical Oncology Research Unit, Università degli Studi di Verona, Verona, Italy
| | - Domenico Mangiameli
- Digestive Molecular Clinical Oncology Research Unit, Università degli Studi di Verona, Verona, Italy
| | - Camilla Zecchetto
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Alberto Quinzii
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Silvia Pietrobono
- Digestive Molecular Clinical Oncology Research Unit, Università degli Studi di Verona, Verona, Italy
| | | | - Simona Casalino
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Marina Gaule
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Camilla Pesoni
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | - Chiara Trentin
- Medical Oncology Unit, Santa Chiara Hospital, Trento, Italy
| | | | - Orazio Caffo
- Medical Oncology Unit, Santa Chiara Hospital, Trento, Italy
| | - Davide Melisi
- Digestive Molecular Clinical Oncology Research Unit, Università degli Studi di Verona, Verona, Italy
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
249
|
Ho HL, Jiang Y, Chiang CL, Karwowska S, Yerram R, Sharma K, Scudder S, Chiu CH, Tsai CM, Palma JF, Sharma A, Chou TY. Efficacy of liquid biopsy for disease monitoring and early prediction of tumor progression in EGFR mutation-positive non-small cell lung cancer. PLoS One 2022; 17:e0267362. [PMID: 35482671 PMCID: PMC9049536 DOI: 10.1371/journal.pone.0267362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/06/2022] [Indexed: 11/18/2022] Open
Abstract
15–40% of non-small cell lung cancer (NSCLC) patients harbor epidermal growth factor receptor (EGFR)-sensitizing mutations. Tyrosine kinase inhibitors (TKIs) provide significant clinical benefit in this population, yet all patients will ultimately progress. Liquid biopsy can reliably identify somatic tumor-associated EGFR mutations in plasma. This study aimed to assess the feasibility and value of the quantitative assessment of EGFR driver mutations in plasma in EGFR-mutated NSCLC patients treated with EGFR-TKIs as a tool to evaluate therapeutic response to TKIs and monitor for disease progression. The study included 136 patients with tissue biopsy-confirmed EGFR-sensitizing, mutation-positive lung adenocarcinoma with plasma collected prior to TKI treatment and at least two post-initiation TKI treatment/follow-up blood samples. Plasma samples were tested with the cobas® EGFR Mutation Test v2 (cobas EGFR Test), and semi-quantitative index (SQI) values for each identified mutation were reported by the assay software. The most common baseline EGFR mutations detected in tissue were L858R (53.7%) and exon 19 deletion (39.7%). Plasma cell-free DNA analysis detected EGFR mutations in 74% of the baseline samples. Objective response rate by RECIST 1.1 was achieved in 72% of patients, while 93% had a molecular response (defined as disappearance of the EGFR mutation from plasma). 83% of patients had molecular progression (MP; 1.5X SQI increase or new T790M mutation), and 82% who had a clinical response had clinical progression. On average, MP occurred 42 days prior to clinical progression. Patients who progressed while on first-line TKI showed MP of the original EGFR-sensitizing mutations prior to the emergence of a T790M mutation, which was detected in 27% of the EGFR plasma-positive patients. Longitudinal monitoring of EGFR mutational load in plasma is feasible and can predict both response and clinical progression in EGFR-mutated NSCLC patients treated with EGFR-TKIs, as well as detect treatment-emergent EGFR mutations.
Collapse
Affiliation(s)
- Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yuqiu Jiang
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
| | - Chi-Lu Chiang
- Division of Thoracic Oncology, Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sylwia Karwowska
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
| | - Ranga Yerram
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
| | - Keerti Sharma
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
| | - Sidney Scudder
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
| | - Chao-Hua Chiu
- Division of Thoracic Oncology, Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Ming Tsai
- Division of Thoracic Oncology, Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - John F. Palma
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
| | - Abha Sharma
- Clinical Development and Medical Affairs, Roche Diagnostic Solutions, Pleasanton, California, United States of America
- * E-mail: (AS); (TYC)
| | - Teh-Ying Chou
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- * E-mail: (AS); (TYC)
| |
Collapse
|
250
|
Arnolda R, Howlett K, Chan T, Raleigh J, Hatzimihalis A, Bell A, Fellowes A, Sandhu S, McArthur GA, Fox SB, Dawson SJ, Hewitt C, Jones K, Wong SQ. Clinical validation and implementation of droplet digital PCR for the detection of BRAF mutations from cell-free DNA. Pathology 2022; 54:772-778. [DOI: 10.1016/j.pathol.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/03/2022] [Accepted: 02/17/2022] [Indexed: 10/18/2022]
|