1
|
Cercek A, Foote MB, Rousseau B, Smith JJ, Shia J, Sinopoli J, Weiss J, Lumish M, Temple L, Patel M, Wilde C, Saltz LB, Argiles G, Stadler Z, Artz O, Maron S, Ku G, Gu P, Janjigian YY, Molena D, Iyer G, Coleman J, Abida W, Cohen S, Soares K, Schattner M, Strong VE, Yaeger R, Paty P, Shcherba M, Sugarman R, Romesser PB, Zervoudakis A, Desai A, Segal NH, El Dika I, Widmar M, Wei I, Pappou E, Fumo G, Aparo S, Gonen M, Gollub M, Jayaprakasham VS, Kim TH, Garcia Aguilar J, Weiser M, Diaz LA. Nonoperative Management of Mismatch Repair-Deficient Tumors. N Engl J Med 2025. [PMID: 40293177 DOI: 10.1056/nejmoa2404512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
BACKGROUND Among patients with mismatch repair-deficient (dMMR), locally advanced rectal cancer, neoadjuvant checkpoint blockade eliminated the need for surgery in a high proportion of patients. Whether this approach can be extended to all early-stage dMMR solid tumors, regardless of tumor site, is unknown. METHODS We conducted a phase 2 study in which patients with stage I, II, or III dMMR solid tumors that were amenable to curative-intent surgery were treated with neoadjuvant dostarlimab, a programmed cell death 1 (PD-1) blocking agent, for 6 months. The response to treatment was assessed in two cohorts: patients in cohort 1 had dMMR, locally advanced rectal cancer, and patients in cohort 2 had dMMR nonrectal solid tumors. Patients with a clinical complete response could elect to proceed with nonoperative management; those with residual disease were to undergo resection. In this analysis, the primary end point, assessed in cohort 1, was a sustained clinical complete response at 12 months. Recurrence-free survival and safety were evaluated. RESULTS A total of 117 patients were included in the analysis. In cohort 1, all 49 patients who completed treatment had a clinical complete response and elected to proceed with nonoperative management. A total of 37 patients had a sustained clinical complete response at 12 months, a finding that met the criterion for efficacy. In cohort 2, a total of 35 of 54 patients who completed treatment had a clinical complete response, and 33 elected to proceed with nonoperative management. Among the 103 patients who completed treatment across both cohorts, 84 had a clinical complete response, and 82 did not undergo surgery. Among the 117 total patients, recurrence-free survival at 2 years was 92% (95% confidence interval, 86 to 99); the median follow-up for recurrence was 20.0 months (range, 0 to 60.8). The majority of patients (95%) had reversible, grade 1 or 2 adverse events (60%) or had no adverse events (35%). The option for curative resection was not compromised during or after treatment in any of the patients. CONCLUSIONS Among patients with early-stage dMMR solid tumors that were amenable to curative-intent surgery, neoadjuvant PD-1 blockade led to organ preservation in a high proportion of patients. (Funded by Swim Across America and others; ClinicalTrials.gov number, NCT04165772.).
Collapse
Affiliation(s)
- Andrea Cercek
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Michael B Foote
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Benoit Rousseau
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - J Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - Jenna Sinopoli
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Jill Weiss
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Melissa Lumish
- Department of Oncology, Case Comprehensive Cancer Center, Cleveland
| | - Lindsay Temple
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Miteshkumar Patel
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Callahan Wilde
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Leonard B Saltz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Guillem Argiles
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Zsofia Stadler
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Oliver Artz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Steven Maron
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Geoffrey Ku
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Ping Gu
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Yelena Y Janjigian
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Daniela Molena
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Gopa Iyer
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Jonathan Coleman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Wassim Abida
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Seth Cohen
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Kevin Soares
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Mark Schattner
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Vivian E Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Rona Yaeger
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Philip Paty
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Marina Shcherba
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Ryan Sugarman
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Alice Zervoudakis
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Avni Desai
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Neil H Segal
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Imane El Dika
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| | - Maria Widmar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Iris Wei
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Emmanouil Pappou
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Gerard Fumo
- Department of Oncology, Hartford HealthCare, Hartford, CT
| | - Santiago Aparo
- Department of Oncology, Baptist Health Miami Cancer Institute, Miami
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York
| | - Marc Gollub
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York
| | | | - Tae-Hyung Kim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York
| | | | - Martin Weiser
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York
| | - Luis A Diaz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York
| |
Collapse
|
2
|
Bartolomucci A, Nobrega M, Ferrier T, Dickinson K, Kaorey N, Nadeau A, Castillo A, Burnier JV. Circulating tumor DNA to monitor treatment response in solid tumors and advance precision oncology. NPJ Precis Oncol 2025; 9:84. [PMID: 40122951 PMCID: PMC11930993 DOI: 10.1038/s41698-025-00876-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a dynamic biomarker in cancer, as evidenced by its increasing integration into clinical practice. Carrying tumor specific characteristics, ctDNA can be used to inform treatment selection, monitor response, and identify drug resistance. In this review, we provide a comprehensive, up-to-date summary of ctDNA in monitoring treatment response with a focus on lung, colorectal, and breast cancers, and discuss current challenges and future directions.
Collapse
Affiliation(s)
- Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Monyse Nobrega
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nivedita Kaorey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Alberto Castillo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Murciano-Goroff YR, Hui ABY, Araujo Filho JA, Hamilton EG, Chabon JJ, Moding EJ, Bonilla RF, Lebow ES, Gomez D, Rimner A, Ginsberg MS, Offin M, Kundra R, Allaj V, Norton L, Reis-Filho JS, Razavi P, Drilon A, Jones DR, Isbell JM, Lai WV, Rudin CM, Alizadeh AA, Li BT, Diehn M. Early Circulating Tumor DNA Shedding Kinetics for Prediction of Platinum Sensitivity in Patients With Small Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2400216. [PMID: 39231375 PMCID: PMC11376985 DOI: 10.1200/po.24.00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/06/2024] [Accepted: 07/18/2024] [Indexed: 09/06/2024] Open
Abstract
PURPOSE Small cell lung cancer (SCLC) is characterized by rapid progression after platinum resistance. Circulating tumor (ctDNA) dynamics early in treatment may help determine platinum sensitivity. MATERIALS AND METHODS Serial plasma samples were collected from patients receiving platinum-based chemotherapy for SCLC on the first 3 days of cycle one and on the first days of subsequent cycles with paired samples collected both before and again after infusions. Tumor-informed plasma analysis was carried out using CAncer Personalized Profiling by deep Sequencing (CAPP-Seq). The mean variant allele frequency (VAF) of all pretreatment mutations was tracked in subsequent blood draws and correlated with radiologic response. RESULTS ctDNA kinetics were assessed in 122 samples from 21 patients. Pretreatment VAF did not differ significantly between patients who did and did not respond to chemotherapy (mean 22.5% v 4.6%, P = .17). A slight increase in ctDNA on cycle 1, day 1 immediately post-treatment was seen in six of the seven patients with available draws (fold change from baseline: 1.01-1.44), half of whom achieved a response. All patients who responded had a >2-fold decrease in mean VAF on cycle 2 day 1 (C2D1). Progression-free survival (PFS) and overall survival (OS) were significantly longer in patients with a >2-fold decrease in mean VAF after one treatment cycle (6.8 v 2.6 months, log-rank P = .0004 and 21.7 v 6.4 months, log rank P = .04, respectively). CONCLUSION A >2-fold decrease in ctDNA concentration was observed by C2D1 in all patients who were sensitive to platinum-based therapy and was associated with longer PFS and OS.
Collapse
Affiliation(s)
- Yonina R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Angela B-Y Hui
- Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Jose A Araujo Filho
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jacob J Chabon
- Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Rene F Bonilla
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Emily S Lebow
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michelle S Ginsberg
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ritika Kundra
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Viola Allaj
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - David R Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - James M Isbell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - W Victoria Lai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA
| | - Bob T Li
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Department of Radiation Oncology, Stanford University, Stanford, CA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA
| |
Collapse
|
4
|
Annapragada AV, Niknafs N, White JR, Bruhm DC, Cherry C, Medina JE, Adleff V, Hruban C, Mathios D, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genome-wide repeat landscapes in cancer and cell-free DNA. Sci Transl Med 2024; 16:eadj9283. [PMID: 38478628 PMCID: PMC11323656 DOI: 10.1126/scitranslmed.adj9283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/16/2024] [Indexed: 03/22/2024]
Abstract
Genetic changes in repetitive sequences are a hallmark of cancer and other diseases, but characterizing these has been challenging using standard sequencing approaches. We developed a de novo kmer finding approach, called ARTEMIS (Analysis of RepeaT EleMents in dISease), to identify repeat elements from whole-genome sequencing. Using this method, we analyzed 1.2 billion kmers in 2837 tissue and plasma samples from 1975 patients, including those with lung, breast, colorectal, ovarian, liver, gastric, head and neck, bladder, cervical, thyroid, or prostate cancer. We identified tumor-specific changes in these patients in 1280 repeat element types from the LINE, SINE, LTR, transposable element, and human satellite families. These included changes to known repeats and 820 elements that were not previously known to be altered in human cancer. Repeat elements were enriched in regions of driver genes, and their representation was altered by structural changes and epigenetic states. Machine learning analyses of genome-wide repeat landscapes and fragmentation profiles in cfDNA detected patients with early-stage lung or liver cancer in cross-validated and externally validated cohorts. In addition, these repeat landscapes could be used to noninvasively identify the tissue of origin of tumors. These analyses reveal widespread changes in repeat landscapes of human cancers and provide an approach for their detection and characterization that could benefit early detection and disease monitoring of patients with cancer.
Collapse
Affiliation(s)
- Akshaya V. Annapragada
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Noushin Niknafs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - James R. White
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daniel C. Bruhm
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Christopher Cherry
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jamie E. Medina
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Vilmos Adleff
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Carolyn Hruban
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dimitrios Mathios
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zachariah H. Foda
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jillian Phallen
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert B. Scharpf
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Victor E. Velculescu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
5
|
Peng WW, Liu Y, Sha HH, Wen SD, Fang Y, Zhou GR. Relationship between plasma circulating cell-free DNA concentration and treatment outcomes including prognosis in patients with advanced non-small cell lung cancer. BMC Pulm Med 2023; 23:348. [PMID: 37710221 PMCID: PMC10503004 DOI: 10.1186/s12890-023-02586-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Some research found that elevated plasma cell-free DNA (cfDNA) concentrations and poor prognosis are associated in non-small cell lung cancer (NSCLC). However, more studies need to be carried out to verify this conclusion. Therefore, this study investigated the relationship between cfDNA concentration and treatment outcomes including prognosis in patients with advanced NSCLC. METHODS We retrospectively collected medical records and cfDNA data from 160 patients with advanced NSCLC. Progression-free survival (PFS) were calculated using the Kaplan-Meier method and were compared between groups using the log rank test. Cox regression analysis was used for estimating the independent predictors of PFS. And we used logistic regression to evaluate the relationship between baseline biomarkers and efficacy. In our study, BT1 cfDNA, BT2 cfDNA, and BT3 cfDNA were defined as cfDNA concentration before the first treatment (baseline cfDNA concentration), cfDNA concentration before the second treatment, and cfDNA concentration before the third treatment, respectively. RESULTS Patients with low cfDNA (BT1 cfDNA < 15 (ng/mL)) were reported a significantly prolonged median progression-free survival (mPFS) compared with patients with patients with high cfDNA (BT1 cfDNA ≥ 15(ng/mL)) (mPFS: 14.6 vs. 8.3 months, P = 0.002), as well as patients with neutrophil/lymphocyte ratio (NLR)<2.98 (mPFS: 13.1 vs. 7.9 months, P = 0.023). In addition, Cox proportional hazards regression analysis identified independent indicators associated with PFS including BT1 cfDNA ≥ 15 (ng/mL), NLR ≥ 2.98 and extrapulmonary metastasis. The best cut-off value for BT3 cfDNA for predicting disease progression is 41.46 (ng/mL) (Area Under the Curve (AUC): 0.652, 95%CI: 0.516-0.788), achieving 90.7% sensitivity and 37.5% specificity for the prediction of disease progression. BT3 cfDNA (OR = 6.08, 95% CI: 1.94-19.57, P = 0.002) was an independent factor for disease progression in patients with advanced NSCLC. CONCLUSIONS BT1 cfDNA may be a biomarker to assess the prognosis of advanced NSCLC. Patients with advanced NSCLC with lower cfDNA and NLR before treatment had a better prognosis. Increased BT3 cfDNA concentration was an independent factor of disease progression in advanced NSCLC patients. These findings may assist in identifying high-risk patients and guiding treatment strategies.
Collapse
Affiliation(s)
- Wei-Wei Peng
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing, 210009, Jiangsu, China
| | - Ying Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing, 210009, Jiangsu, China
| | - Huan-Huan Sha
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing, 210009, Jiangsu, China
| | - Shao-Di Wen
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing, 210009, Jiangsu, China
| | - Ying Fang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing, 210009, Jiangsu, China.
| | - Guo-Ren Zhou
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
6
|
Ferrier ST, Tsering T, Sadeghi N, Zeitouni A, Burnier JV. Blood and saliva-derived ctDNA is a marker of residual disease after treatment and correlates with recurrence in human papillomavirus-associated head and neck cancer. Cancer Med 2023; 12:15777-15787. [PMID: 37526056 PMCID: PMC10469655 DOI: 10.1002/cam4.6191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/15/2023] [Accepted: 05/21/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND There is an alarming increase in human papillomavirus-associated head and neck cancer (HNC), reaching epidemic levels. While patient prognosis is generally good, off-target treatment effects are associated with decreased quality of life. Thus, non-invasive strategies to predict treatment response and risk of recurrence could help de-escalate treatment. In this study, we tested circulating tumor (ct)DNA in liquid biopsies (blood/saliva) of HPV-positive HNC patients to assess treatment response and disease progression. METHODS A total of 235 blood and saliva samples were collected from 60 HPV-positive and 17 HPV-negative HNC patients (control group) before and/or after treatment. Samples were analyzed using ddPCR for HPV16/18/31/33/35/45 and correlated with imaging and pathological examination. RESULTS HPV-ctDNA detection was significantly higher prior to treatment (91%) than after treatment (8.0%) (χ2 p < 0.00001), with high concordance between saliva and blood (93%). In matched samples, all patients positive for ctDNA before treatment showed significant reductions in ctDNA levels post treatment (p < 0.0001). All but one patient with persistent ctDNA after treatment showed residual tumor and subsequent recurrence. Finally, fragmentomic analysis revealed shifts in cell-free DNA fragment size after treatment, suggesting a complementary biomarker for treatment response. CONCLUSIONS Blood and saliva were found to be good sources of HPV-ctDNA. The presence of ctDNA strongly correlated with treatment response, demonstrating clinical utility as a non-invasive biomarker to monitor tumor progression in HPV-positive HNC. Liquid biopsy based ctDNA testing could be an effective approach to predict recurrence and stratify patients for de-escalation of treatment, thereby improving quality of life.
Collapse
Affiliation(s)
- Sarah Tadhg Ferrier
- Cancer Research ProgramResearch Institute of the McGill University Health CentreMontrealCanada
- Department of PathologyMcGill UniversityMontrealCanada
| | - Thupten Tsering
- Cancer Research ProgramResearch Institute of the McGill University Health CentreMontrealCanada
- Department of PathologyMcGill UniversityMontrealCanada
| | - Nader Sadeghi
- Cancer Research ProgramResearch Institute of the McGill University Health CentreMontrealCanada
- Department of Otolaryngology – Head and Neck SurgeryMcGill UniversityMontrealCanada
- Gerald Bronfman Department of OncologyMcGill UniversityMontrealCanada
| | - Anthony Zeitouni
- Department of Otolaryngology – Head and Neck SurgeryMcGill UniversityMontrealCanada
| | - Julia V. Burnier
- Cancer Research ProgramResearch Institute of the McGill University Health CentreMontrealCanada
- Department of PathologyMcGill UniversityMontrealCanada
- Gerald Bronfman Department of OncologyMcGill UniversityMontrealCanada
| |
Collapse
|
7
|
Diaz IM, Nocon A, Held SAE, Kobilay M, Skowasch D, Bronkhorst AJ, Ungerer V, Fredebohm J, Diehl F, Holdenrieder S, Holtrup F. Pre-Analytical Evaluation of Streck Cell-Free DNA Blood Collection Tubes for Liquid Profiling in Oncology. Diagnostics (Basel) 2023; 13:diagnostics13071288. [PMID: 37046506 PMCID: PMC10093569 DOI: 10.3390/diagnostics13071288] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Excellent pre-analytical stability is an essential precondition for reliable molecular profiling of circulating tumor DNA (ctDNA) in oncological diagnostics. Therefore, in vitro degradation of ctDNA and the additional release of contaminating genomic DNA from lysed blood cells must be prevented. Streck Cell-Free DNA blood collection tubes (cfDNA BCTs) have proposed advantages over standard K2EDTA tubes, but mainly have been tested in healthy individuals. Blood was collected from cancer patients (n = 53) suffering from colorectal (n = 21), pancreatic (n = 11), and non-small-cell lung cancer (n = 21) using cfDNA BCT tubes and K2EDTA tubes that were processed immediately or after 3 days (BCTs) or 6 hours (K2EDTA) at room temperature. The cfDNA isolated from these samples was characterized in terms of yield using LINE-1 qPCR; the level of gDNA contamination; and the mutation status of KRAS, NRAS, and EGFR genes using BEAMing ddPCR. CfDNA yield and gDNA levels were comparable in both tube types and were not affected by prolonged storage of blood samples for at least 3 days in cfDNA BCTs or 6 hours in K2EDTA tubes. In addition, biospecimens collected in K2EDTA tubes and cfDNA BCTs stored for up to 3 days demonstrated highly comparable levels of mutational load across all respective cancer patient cohorts and a wide range of concentrations. Our data support the applicability of clinical oncology specimens collected and stored in cfDNA BCTs for up to 3 days for reliable cfDNA and mutation analyses.
Collapse
|
8
|
Koval AP, Khromova AS, Blagodatskikh KA, Zhitnyuk YV, Shtykova YA, Alferov AA, Kushlinskii NE, Shcherbo DS. Application of PCR-based approaches for evaluation of cell-free DNA fragmentation in colorectal cancer. Front Mol Biosci 2023; 10:1101179. [PMID: 37051326 PMCID: PMC10083340 DOI: 10.3389/fmolb.2023.1101179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Cell-free DNA (cfDNA) testing is the core of most liquid biopsy assays. In particular, cfDNA fragmentation features could facilitate non-invasive cancer detection due to their interconnection with tumor-specific epigenetic alterations. However, the final cfDNA fragmentation profile in a purified sample is the result of a complex interplay between informative biological and artificial technical factors. In this work, we use ddPCR to study cfDNA lengths in colorectal cancer patients and observe shorter and more variable cfDNA fragments in accessible chromatin loci compared to the densely packed pericentromeric region. We also report a convenient qPCR system suitable for screening cfDNA samples for artificial high molecular weight DNA contamination.
Collapse
Affiliation(s)
- Anastasia P. Koval
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexandra S. Khromova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Konstantin A. Blagodatskikh
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Center of Genetics and Reproductive Medicine “Genetico”, Moscow, Russia
| | - Yulia V. Zhitnyuk
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Aleksandr A. Alferov
- Laboratory of Clinical Biochemistry, N. N. Blokhin Cancer Research Medical Center of Oncology, Moscow, Russia
| | - Nikolay E. Kushlinskii
- Laboratory of Clinical Biochemistry, N. N. Blokhin Cancer Research Medical Center of Oncology, Moscow, Russia
| | - Dmitry S. Shcherbo
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- *Correspondence: Dmitry S. Shcherbo,
| |
Collapse
|
9
|
Conde AJ, Keraite I, Leslie NR, Kersaudy-Kerhoas M. Microfluidic Acoustic Method for High Yield Extraction of Cell-Free DNA in Low-Volume Plasma Samples. Methods Mol Biol 2023; 2679:163-180. [PMID: 37300615 DOI: 10.1007/978-1-0716-3271-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell-free DNA has many applications in clinical medicine, in particular in cancer diagnosis and cancer treatment monitoring. Microfluidic-based solutions could provide solutions for rapid, cheaper, decentralized detection of cell-free tumoral DNA from a simple blood draw, or liquid biopsies, replacing invasive procedures or expensive scans. In this method, we present a simple microfluidic system for the extraction of cell-free DNA from low volume of plasma samples (≤500 μL). The technique is suitable for either static or continuous flow systems and can be used as a stand-alone module or integrated within a lab-on-chip system. The system relies on a simple yet highly versatile bubble-based micromixer module whose custom components can be fabricated with a combination of low-cost rapid prototyping techniques or ordered via widely available 3D-printing services. This system is capable of performing cell-free DNA extractions from small volumes of blood plasma with up to a tenfold increase in capture efficiency when compared to control methods.
Collapse
Affiliation(s)
- Alvaro J Conde
- Micronit B.V., Enschede, Netherlands
- Heriot-Watt University, Edinburgh, UK
| | - Ieva Keraite
- Heriot-Watt University, Edinburgh, UK
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | | | - Maïwenn Kersaudy-Kerhoas
- Heriot-Watt University, Edinburgh, UK.
- University of Edinburgh, Infection Medicine, Edinburgh, UK.
| |
Collapse
|
10
|
Brooks PJ, Malkin EZ, De Michino S, Bratman SV. Isolation of salivary cell-free DNA for cancer detection. PLoS One 2023; 18:e0285214. [PMID: 37130100 PMCID: PMC10153704 DOI: 10.1371/journal.pone.0285214] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023] Open
Abstract
Saliva is an emerging source of disease biomarkers, particularly for cancers of the head and neck. Although analysis of cell-free DNA (cfDNA) in saliva holds promise as a liquid biopsy for cancer detection, currently there are no standardized methodologies for the collection and isolation of saliva for the purposes of studying DNA. Here, we evaluated various saliva collection receptacles and DNA purification techniques, comparing DNA quantity, fragment size, source, and stability. Then, using our optimized techniques, we tested the ability to detect human papillomavirus (HPV) DNA- a bona fide cancer biomarker in a subset of head and neck cancers- from patient saliva samples. For saliva collection, we found that the Oragene OG-600 receptacle yielded the highest concentration of total salivary DNA as well as short fragments <300 bp corresponding to mononucleosomal cell-free DNA. Moreover, these short fragments were stabilized beyond 48 hours after collection in contrast to other saliva collection receptacles. For DNA purification from saliva, the QIAamp Circulating Nucleic Acid kit yielded the highest concentration of mononucleosome-sized DNA fragments. Freeze-thaw of saliva samples did not affect DNA yield or fragment size distribution. Salivary DNA isolated from the OG-600 receptacle was found to be composed of both single and double-stranded DNA, including mitochondrial and microbial sources. While levels of nuclear DNA were consistent over time, levels of mitochondrial and microbial DNA were more variable and increased 48 hours after collection. Finally, we found that HPV DNA was stable in OG-600 receptacles, was reliably detected within the saliva of patients with HPV-positive head and neck cancer, and was abundant among mononucleosome-sized cell-free DNA fragments. Our studies have defined optimal techniques for isolating DNA from saliva that will contribute to future applications in liquid biopsy-based cancer detection.
Collapse
Affiliation(s)
- Patricia J Brooks
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ethan Z Malkin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Steven De Michino
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Ciriaco N, Zamora E, Escrivá-de-Romaní S, Miranda Gómez I, Jiménez Flores J, Saura C, Sloane H, Starus A, Fredebohm J, Georgieva L, Speight G, Jones F, Ramón y Cajal S, Espinosa-Bravo M, Peg V. Clearance of ctDNA in triple-negative and HER2-positive breast cancer patients during neoadjuvant treatment is correlated with pathologic complete response. Ther Adv Med Oncol 2022; 14:17588359221139601. [PMID: 36479470 PMCID: PMC9720791 DOI: 10.1177/17588359221139601] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/31/2022] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Although the standard of care is to perform surgery of primary breast cancer (BC) after neoadjuvant chemotherapy (NAC), for certain patients achieving clinical complete response (cCR) and pathologic complete response (pCR), omission of surgical treatment may be an option. Levels of circulating tumor DNA (ctDNA) during and after therapy could identify patients achieving minimal residual disease. In this study, we evaluated whether ctDNA clearance during NAC could be a correlate to effective response in human epidermal growth factor receptor 2 positive (HER2+) and triple-negative (TN) BC patients. METHODS A prospective study was conducted to identify patient-specific PIK3CA and TP53 mutations in tissue using next-generation sequencing, which could then be used to track the presence/absence of mutations prior to, during, and following NAC using Sysmex SafeSEQ technology. All patients underwent a surgical excision after NAC, and pCR was assessed. RESULTS A total of 29 TN and HER2+ BC patients were examined and 20 that carried mutations in the PIK3CA and/or TP53 genes were recruited. Overall, 19 of these 20 patients harbored at least one tumor-specific mutation in their plasma at baseline. After NAC, 15 patients (75.0%) achieved pCR according to the histopathologic evaluation of the surgical specimen, and 15 patients (75.0%) had a cCR; 18 of 20 patients (90.0%) had concordant pCR and cCR. The status of 'no mutation detected' (NMD) following NAC in cCR patients correctly identified the pCR in 14 of 15 patients (93.33%), as well as correctly ruled out pCR in three patients, with an accuracy of 89.47%. During the 12-month follow-up after surgery, 40 plasma samples collected from 15 patients all showed no detectable ctDNA (NMD), and no patient recurred. CONCLUSION These findings prompt further research of the value of ctDNA for non-invasive prediction of clinical/pathological response, raising the possibility of sparing surgery following NAC in selected BC patients.
Collapse
Affiliation(s)
- Nikaoly Ciriaco
- Pathology Department, Hospital del Mar, Barcelona, Spain
- Universidad Autónoma de Barcelona, Barcelona, Spain
| | - Esther Zamora
- Universidad Autónoma de Barcelona, Barcelona, Spain
- Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Santiago Escrivá-de-Romaní
- Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | | | - José Jiménez Flores
- Molecular Oncology Lab. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Cristina Saura
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Hillary Sloane
- Sysmex Inostics, Inc., Baltimore, MD, USA
- Sysmex Inostics GmbH, Hamburg, Germany
| | - Anna Starus
- Sysmex Inostics, Inc., Baltimore, MD, USA
- Sysmex Inostics GmbH, Hamburg, Germany
| | - Johannes Fredebohm
- Sysmex Inostics, Inc., Baltimore, MD, USA
- Sysmex Inostics GmbH, Hamburg, Germany
| | | | | | - Frederick Jones
- Sysmex Inostics, Inc., Baltimore, MD, USA
- Sysmex Inostics GmbH, Hamburg, Germany
| | - Santiago Ramón y Cajal
- Universidad Autónoma de Barcelona, Barcelona, Spain
- Pathology Department, Vall d’Hebron University Hospital, Barcelona, Spain
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain
| | | | - Vicente Peg
- Universidad Autónoma de Barcelona, Barcelona, Spain
- Pathology Department, Vall d’Hebron University Hospital, Paseo Vall d’Hebron 119-129, Barcelona 08035, Spain
- Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain
| |
Collapse
|
12
|
Sauer CM, Heider K, Belic J, Boyle SE, Hall JA, Couturier D, An A, Vijayaraghavan A, Reinius MAV, Hosking K, Vias M, Rosenfeld N, Brenton JD. Longitudinal monitoring of disease burden and response using ctDNA from dried blood spots in xenograft models. EMBO Mol Med 2022; 14:e15729. [PMID: 35694774 PMCID: PMC9358392 DOI: 10.15252/emmm.202215729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 12/29/2022] Open
Abstract
Whole-genome sequencing (WGS) of circulating tumour DNA (ctDNA) is now a clinically important biomarker for predicting therapy response, disease burden and disease progression. However, the translation of ctDNA monitoring into vital preclinical PDX models has not been possible owing to low circulating blood volumes in small rodents. Here, we describe the longitudinal detection and monitoring of ctDNA from minute volumes of blood in PDX mice. We developed a xenograft Tumour Fraction (xTF) metric using shallow WGS of dried blood spots (DBS), and demonstrate its application to quantify disease burden, monitor treatment response and predict disease outcome in a preclinical study of PDX mice. Further, we show how our DBS-based ctDNA assay can be used to detect gene-specific copy number changes and examine the copy number landscape over time. Use of sequential DBS ctDNA assays could transform future trial designs in both mice and patients by enabling increased sampling and molecular monitoring.
Collapse
Affiliation(s)
- Carolin M Sauer
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Katrin Heider
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Jelena Belic
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Samantha E Boyle
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - James A Hall
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Dominique‐Laurent Couturier
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Medical Research Council Biostatistics UnitUniversity of CambridgeCambridgeUK
| | - Angela An
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Aadhitthya Vijayaraghavan
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Marika AV Reinius
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Karen Hosking
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Maria Vias
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
| | - James D Brenton
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
- Cancer Research UK Major Centre–CambridgeUniversity of CambridgeCambridgeUK
- Cambridge University Hospitals NHS Foundation Trust and National Institute for Health Research Cambridge Biomedical Research CentreAddenbrooke's HospitalCambridgeUK
- Department of OncologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
13
|
Liu AP, Smith KS, Kumar R, Robinson GW, Northcott PA. Low-coverage whole-genome sequencing of cerebrospinal-fluid-derived cell-free DNA in brain tumor patients. STAR Protoc 2022; 3:101292. [PMID: 35463474 PMCID: PMC9026582 DOI: 10.1016/j.xpro.2022.101292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This protocol summarizes the pipeline for analysis of tumor-derived cell-free DNA (cfDNA) from cerebrospinal fluid (CSF) using low-coverage whole-genome sequencing (lcWGS). This approach enables resolution of chromosomal and focal copy-number variations (CNVs) as oncologic signatures, particularly for patients with central nervous system tumors. Our strategy tolerates sub-nanogram cfDNA input and is thus optimized for CSF samples where cfDNA yields are typically low. Overall, the detection of tumor-specific signatures in CSF-derived cfDNA is a promising biomarker for personalization of brain-tumor therapy. For complete details on the use and execution of this protocol, please refer to Liu et al. (2021).
Collapse
Affiliation(s)
- Anthony P.Y. Liu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children’s Hospital, Hong Kong SAR, China
- Division of Brain Tumor Research, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Kyle S. Smith
- Division of Brain Tumor Research, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Rahul Kumar
- Division of Brain Tumor Research, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Giles W. Robinson
- Division of Neuro-Oncology, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Paul A. Northcott
- Division of Brain Tumor Research, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
14
|
Basic Science with Preclinical Models to Investigate and Develop Liquid Biopsy: What Are the Available Data and Is It a Fruitful Approach? Int J Mol Sci 2022; 23:ijms23105343. [PMID: 35628154 PMCID: PMC9141279 DOI: 10.3390/ijms23105343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
The molecular analysis of circulating analytes (circulating tumor-DNA (ctDNA), -cells (CTCs) and -RNA (ctRNA)/exosomes) deriving from solid tumors and detected in the bloodstream—referred as liquid biopsy—has emerged as one of the most promising concepts in cancer management. Compelling data have evidenced its pivotal contribution and unique polyvalence through multiple applications. These data essentially derived from translational research. Therewith, data on liquid biopsy in basic research with preclinical models are scarce, a concerning lack that has been widely acknowledged in the field. This report aimed to comprehensively review the available data on the topic, for each analyte. Only 17, 17 and 2 studies in basic research investigated ctDNA, CTCs and ctRNA/exosomes, respectively. Albeit rare, these studies displayed noteworthy relevance, demonstrating the capacity to investigate questions related to the biology underlying analytes release that could not be explored via translational research with human samples. Translational, clinical and technological sectors of liquid biopsy may benefit from basic research and should take note of some important findings generated by these studies. Overall, results underscored the need to intensify the efforts to conduct future studies on liquid biopsy in basic research with new preclinical models.
Collapse
|
15
|
Jeffers M, Kappeler C, Kuss I, Beckmann G, Mehnert DH, Fredebohm J, Teufel M. Broad spectrum of regorafenib activity on mutant KIT and absence of clonal selection in gastrointestinal stromal tumor (GIST): correlative analysis from the GRID trial. Gastric Cancer 2022; 25:598-608. [PMID: 35050442 PMCID: PMC9013336 DOI: 10.1007/s10120-021-01274-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND In the phase 3 GRID trial, regorafenib improved progression-free survival (PFS) independent of KIT mutations in exons 9 and 11. In this retrospective, exploratory analysis of the GRID trial, we investigated whether a more comprehensive KIT mutation analysis could identify mutations that impact treatment outcome with regorafenib and a regorafenib-induced mutation pattern. METHODS Archived tumor samples, collected at any time prior to enrollment in GRID, were analyzed by Sanger sequencing (n = 102) and next-generation sequencing (FoundationONE; n = 47). Plasma samples collected at baseline were analyzed by BEAMing (n = 163) and SafeSEQ (n = 96). RESULTS In archived tumor samples, 67% (68/102) had a KIT mutation; 61% (62/102) had primary KIT mutations (exons 9 and 11) and 12% (12/102) had secondary mutations (exons 13, 14, 17, and 18). At baseline, 81% of samples (78/96) had KIT mutations by SafeSEQ, including the M541L polymorphism (sole event in 6 patients). Coexisting mutations in other oncogenes were rare, as were mutations in PDGFR, KRAS, and BRAF. Regorafenib showed PFS benefit across all primary and secondary KIT mutational subgroups examined. Available patient-matched samples taken at baseline and end of treatment (n = 41; SafeSEQ), revealed heterogeneous KIT mutational changes with no specific mutation pattern emerging upon regorafenib treatment. CONCLUSION These data support the results of the GRID trial, and suggest that patients may benefit from regorafenib in the presence of KIT mutations and without the selection of particular mutation patterns that confer resistance. The study was not powered to address biomarker-related questions, and the results are exploratory and hypothesis-generating.
Collapse
Affiliation(s)
- Michael Jeffers
- Bayer HealthCare Pharmaceuticals, 100 Bayer Blvd, Whippany, NJ, 07981, USA.
| | | | | | | | | | | | - Michael Teufel
- Bayer HealthCare Pharmaceuticals, 100 Bayer Blvd, Whippany, NJ, 07981, USA
- Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| |
Collapse
|
16
|
Chakraborty AR, Vassilev A, Jaiswal SK, O'Connell CE, Ahrens JF, Mallon BS, Pera MF, DePamphilis ML. Selective elimination of pluripotent stem cells by PIKfyve specific inhibitors. Stem Cell Reports 2022; 17:397-412. [PMID: 35063131 PMCID: PMC8828683 DOI: 10.1016/j.stemcr.2021.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 01/05/2023] Open
Abstract
Inhibition of PIKfyve phosphoinositide kinase selectively kills autophagy-dependent cancer cells by disrupting lysosome homeostasis. Here, we show that PIKfyve inhibitors can also selectively eliminate pluripotent embryonal carcinoma cells (ECCs), embryonic stem cells, and induced pluripotent stem cells under conditions where differentiated cells remain viable. PIKfyve inhibitors prevented lysosome fission, induced autophagosome accumulation, and reduced cell proliferation in both pluripotent and differentiated cells, but they induced death only in pluripotent cells. The ability of PIKfyve inhibitors to distinguish between pluripotent and differentiated cells was confirmed with xenografts derived from ECCs. Pretreatment of ECCs with the PIKfyve specific inhibitor WX8 suppressed their ability to form teratocarcinomas in mice, and intraperitoneal injections of WX8 into mice harboring teratocarcinoma xenografts selectively eliminated pluripotent cells. Differentiated cells continued to proliferate, but at a reduced rate. These results provide a proof of principle that PIKfyve specific inhibitors can selectively eliminate pluripotent stem cells in vivo as well as in vitro.
Collapse
Affiliation(s)
- Arup R Chakraborty
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Alex Vassilev
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Sushil K Jaiswal
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Constandina E O'Connell
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - John F Ahrens
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA
| | - Barbara S Mallon
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Melvin L DePamphilis
- National Institute of Child Health & Human Development, National Institutes of Health, Bldg. 6A/3A15, 6 Center Drive, Bethesda, MD 20892-2790, USA.
| |
Collapse
|
17
|
Wan JCM, Mughal TI, Razavi P, Dawson SJ, Moss EL, Govindan R, Tan IB, Yap YS, Robinson WA, Morris CD, Besse B, Bardelli A, Tie J, Kopetz S, Rosenfeld N. Liquid biopsies for residual disease and recurrence. MED 2021; 2:1292-1313. [PMID: 35590147 DOI: 10.1016/j.medj.2021.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/27/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023]
Abstract
Detection of minimal residual disease in patients with cancer, who are in complete remission with no cancer cells detectable, has the potential to improve recurrence-free survival through treatment selection. Studies analyzing circulating tumor DNA (ctDNA) in patients with solid tumors suggest the potential to accurately predict and detect relapse, enabling treatment strategies that may improve clinical outcomes. Over the past decade, assays for ctDNA detection in plasma samples have steadily increased in sensitivity and specificity. These are applied for the detection of residual disease after treatment and for earlier detection of recurrence. Novel clinical trials are now assessing how assays for "residual disease and recurrence" (RDR) may influence current treatment paradigms and potentially change the landscape of risk classification for cancer recurrence. In this review, we appraise the progress of RDR detection using ctDNA and consider the emerging role of liquid biopsy in the monitoring and management of solid tumors.
Collapse
Affiliation(s)
| | - Tariq Imdadali Mughal
- Tufts University School of Medicine, Boston, MA 02111, USA; University of Buckingham, Buckingham MK18 1EG, UK
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Esther Louise Moss
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK; Department of Gynaecological Oncology, University Hospitals of Leicester NHS Trust, Leicester General Hospital, Leicester LE5 4PW, UK
| | | | - Iain Beehuat Tan
- Division of Medical Oncology, National Cancer Centre Singapore, 169610 Singapore, Singapore
| | - Yoon-Sim Yap
- Division of Medical Oncology, National Cancer Centre Singapore, 169610 Singapore, Singapore
| | | | | | - Benjamin Besse
- Department of Cancer Medicine, Institut Gustave Roussy Cancer Center, 94805 Villejuif, France
| | - Alberto Bardelli
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo TO, Italy; Department of Oncology, University of Turin, 10060 Candiolo TO, Italy
| | - Jeanne Tie
- Peter MacCallum Cancer Center, Melbourne, VIC 3000, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Scott Kopetz
- MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nitzan Rosenfeld
- Inivata, Cambridge CB22 3FH, UK; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; Cancer Research UK Cambridge Centre, Cambridge CB2 0RE, UK.
| |
Collapse
|
18
|
Bortolini Silveira A, Bidard FC, Tanguy ML, Girard E, Trédan O, Dubot C, Jacot W, Goncalves A, Debled M, Levy C, Ferrero JM, Jouannaud C, Rios M, Mouret-Reynier MA, Dalenc F, Hego C, Rampanou A, Albaud B, Baulande S, Berger F, Lemonnier J, Renault S, Desmoulins I, Proudhon C, Pierga JY. Multimodal liquid biopsy for early monitoring and outcome prediction of chemotherapy in metastatic breast cancer. NPJ Breast Cancer 2021; 7:115. [PMID: 34504096 PMCID: PMC8429692 DOI: 10.1038/s41523-021-00319-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) are two cancer-derived blood biomarkers that inform on patient prognosis and treatment efficacy in breast cancer. We prospectively evaluated the clinical validity of quantifying both CTCs (CellSearch) and ctDNA (targeted next-generation sequencing). Their combined value as prognostic and early monitoring markers was assessed in 198 HER2-negative metastatic breast cancer patients. All patients were included in the prospective multicenter UCBG study COMET (NCT01745757) and treated by first-line chemotherapy with weekly paclitaxel and bevacizumab. Blood samples were obtained at baseline and before the second cycle of chemotherapy. At baseline, CTCs and ctDNA were respectively detected in 72 and 74% of patients and were moderately correlated (Kendall’s τ = 0.3). Only 26 (13%) patients had neither detectable ctDNA nor CTCs. Variants were most frequently observed in TP53 and PIK3CA genes. KMT2C/MLL3 variants detected in ctDNA were significantly associated with a lower CTC count, while the opposite trend was seen with GATA3 alterations. Both CTC and ctDNA levels at baseline and after four weeks of treatment were correlated with survival. For progression-free and overall survival, the best multivariate prognostic model included tumor subtype (triple negative vs other), grade (grade 3 vs other), ctDNA variant allele frequency (VAF) at baseline (per 10% increase), and CTC count at four weeks (≥5CTC/7.5 mL). Overall, this study demonstrates that CTCs and ctDNA have nonoverlapping detection profiles and complementary prognostic values in metastatic breast cancer patients. A comprehensive liquid-biopsy approach may involve simultaneous detection of ctDNA and CTCs.
Collapse
Affiliation(s)
| | - François-Clément Bidard
- Circulating Tumor Biomarkers laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France.,Department of Medical Oncology, Institut Curie, Paris and Saint Cloud, Paris, France.,UVSQ, Université Paris-Saclay, Paris, France
| | | | - Elodie Girard
- INSERM U900, Institut Curie, Mines ParisTech, PSL Research University, Paris, France
| | - Olivier Trédan
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Coraline Dubot
- Department of Medical Oncology, Institut Curie, Paris and Saint Cloud, Paris, France
| | - William Jacot
- Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier University, IRCM INSERM, Montpellier, France
| | - Anthony Goncalves
- Department of Medical Oncology, Aix-Marseille Univ, INSERM U1068, CNRS UMR7258, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Marc Debled
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Jean-Marc Ferrero
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | | | - Maria Rios
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-Lès-Nancy, France
| | | | - Florence Dalenc
- Department of Medical Oncology, Institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | - Caroline Hego
- Circulating Tumor Biomarkers laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Aurore Rampanou
- Circulating Tumor Biomarkers laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Benoit Albaud
- ICGex Next-Generation Sequencing Platform, Institut Curie, Paris, France
| | - Sylvain Baulande
- ICGex Next-Generation Sequencing Platform, Institut Curie, Paris, France
| | | | | | - Shufang Renault
- Circulating Tumor Biomarkers laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France
| | - Isabelle Desmoulins
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Charlotte Proudhon
- Circulating Tumor Biomarkers laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France.,INSERM U934 CNRS UMR3215, Paris, France
| | - Jean-Yves Pierga
- Circulating Tumor Biomarkers laboratory, INSERM CIC BT-1428, Institut Curie, Paris, France. .,Department of Medical Oncology, Institut Curie, Paris and Saint Cloud, Paris, France. .,Université de Paris, Paris, France.
| |
Collapse
|
19
|
Krasic J, Abramovic I, Vrtaric A, Nikolac Gabaj N, Kralik-Oguic S, Katusic Bojanac A, Jezek D, Sincic N. Impact of Preanalytical and Analytical Methods on Cell-Free DNA Diagnostics. Front Cell Dev Biol 2021; 9:686149. [PMID: 34552921 PMCID: PMC8451956 DOI: 10.3389/fcell.2021.686149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/13/2021] [Indexed: 01/18/2023] Open
Abstract
While tissue biopsy has for the longest time been the gold-standard in biomedicine, precision/personalized medicine is making the shift toward liquid biopsies. Cell-free DNA (cfDNA) based genetic and epigenetic biomarkers reflect the molecular status of its tissue-of-origin allowing for early and non-invasive diagnostics of different pathologies. However, selection of preanalytical procedures (including cfDNA isolation) as well as analytical methods are known to impact the downstream results. Calls for greater standardization are made continuously, yet comprehensive assessments of the impact on diagnostic parameters are lacking. This study aims to evaluate the preanalytic and analytic factors that influence cfDNA diagnostic parameters in blood and semen. Text mining analysis has been performed to assess cfDNA research trends, and identify studies on isolation methods, preanalytical and analytical impact. Seminal and blood plasma were tested as liquid biopsy sources. Traditional methods of cfDNA isolation, commercial kits (CKs), and an in-house developed protocol were tested, as well as the impact of dithiothreitol (DTT) on cfDNA isolation performance. Fluorimetry, qPCR, digital droplet PCR (ddPCR), and bioanalyzer were compared as cfDNA quantification methods. Fragment analysis was performed by qPCR and bioanalyzer while the downstream application (cfDNA methylation) was analyzed by pyrosequencing. In contrast to blood, semen as a liquid biopsy source has only recently begun to be reported as a liquid biopsy source, with almost half of all publications on it being review articles. Experimental data revealed that cfDNA isolation protocols give a wide range of cfDNA yields, both from blood and seminal plasma. The addition of DTT to CKs has improved yields in seminal plasma and had a neutral/negative impact in blood plasma. Capillary electrophoresis and fluorometry reported much higher yields than PCR methods. While cfDNA yield and integrity were highly impacted, cfDNA methylation was not affected by isolation methodology or DTT. In conclusion, NucleoSnap was recognized as the kit with the best overall performance. DTT improved CK yields in seminal plasma. The in-house developed protocol has shown near-kit isolation performance. ddPCR LINE-1 assay for absolute detection of minute amounts of cfDNA was established and allowed for quantification of samples inhibited in qPCR. cfDNA methylation was recognized as a stable biomarker unimpacted by cfDNA isolation method. Finally, semen was found to be an abundant source of cfDNA offering potential research opportunities and benefits for cfDNA based biomarkers development related to male reproductive health.
Collapse
Affiliation(s)
- Jure Krasic
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Irena Abramovic
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Alen Vrtaric
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Nora Nikolac Gabaj
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Sasa Kralik-Oguic
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
- Clinical Institute of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ana Katusic Bojanac
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Davor Jezek
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nino Sincic
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
20
|
Takai E, Omata W, Totoki Y, Nakamura H, Shiba S, Takahashi A, Namikawa K, Mori T, Yamazaki N, Shibata T, Yachida S. Clonal dynamics of circulating tumor DNA during immune checkpoint blockade therapy for melanoma. Cancer Sci 2021; 112:4748-4757. [PMID: 34477284 PMCID: PMC8586661 DOI: 10.1111/cas.15088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/10/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022] Open
Abstract
Assessment of treatment efficacy of immune checkpoint inhibitors in melanoma patients is difficult as the response to these therapies varies among patients or lesions. The clonal evolution of cancer during immune checkpoint blockade therapy could cause treatment resistance. We investigated the potential of liquid biopsy in monitoring the mutational profiles of metastatic melanoma during immunotherapy. Plasma samples collected from 21 Japanese metastatic melanoma patients before immune checkpoint blockade therapy were subjected to whole‐exome sequencing (WES). Furthermore, 14 Japanese patients with melanoma were enrolled for longitudinal analysis of circulating tumor DNA (ctDNA). Plasma samples were collected prospectively before and during therapy and sequenced. WES of the pretreatment plasma from Japanese melanoma patients showed detectable ctDNA levels with wide ranges of variant allele frequencies within a sample, suggesting clonal and subclonal mutations in ctDNA. In targeted sequencing using longitudinal samples, ctDNA levels correlated with increased tumor size, while ctDNA content immediately decreased after a surge in a patient exhibiting pseudo‐progression, suggesting the potential of ctDNA analysis in discriminating between pseudo‐ and true progression. Mutant ctDNA levels showed different patterns within the clinical course of specific patients, suggesting that these mutations were derived from different tumor clones with distinct therapeutic responses. During further investigation, WES of plasma samples from 1 patient showed marked differences in the mutational profiles of ctDNA, including expansive tumor evolution during an acute exacerbation. Immunotherapy may induce characteristic clonal evolutions of tumors; longitudinal analysis of ctDNA has the potential of determining these tumor evolution patterns and therapeutic responses.
Collapse
Affiliation(s)
- Erina Takai
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Wataru Omata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.,Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Totoki
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiromi Nakamura
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Satoshi Shiba
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Takahashi
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Taisuke Mori
- Department of Diagnostic Pathology, National Cancer Canter Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.,Laboratory of Molecular Medicine, The Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.,Division of Genomic Medicine, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
21
|
Mishra A, Zennami K, Velarde E, Thorek DLJ, Yegnasubramanian S, DeWeese TL, Lupold SE. Longitudinal measurement of subcutaneous and intratibial human prostate cancer xenograft growth and response to ionizing radiation by plasma Alu and LINE-1 ctDNA: A comparison to standard methods. Prostate 2021; 81:745-753. [PMID: 34032307 DOI: 10.1002/pros.24171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Current preclinical models of metastatic prostate cancer (PCa) require sophisticated technologies and/or genetically engineered cells for the noninvasive monitoring of tumors in remote sites, such as bone. Recent developments in circulating tumor DNA (ctDNA) analysis provide an alternative method for noninvasive tumor monitoring at a low cost. Here, we sought to evaluate human Alu and LINE-1 ctDNA for the longitudinal measurement of subcutaneous and intratibial human PCa xenograft growth and response to ionizing radiation (IR) through comparison with standard slide caliper and bioluminescence measurements. MATERIAL AND METHODS Intratibial and subcutaneous xenografts were established in male athymic nude mice using LNCaP cells that stably express firefly luciferase. A subset of tumors was treated with a single dose of IR (CT-guided focal IR, 6 Gy). Tumor measurements were simultaneously taken by slide caliper (subcutaneous only), in vivo bioluminescence imaging, and quantitative real-time PCR (qPCR) of human-specific Alu and LINE-1 ctDNA for several weeks. RESULTS Levels of ctDNA and bioluminescence increased concordantly with subcutaneous and intratibial tumor growth. A statistically significant correlation (Spearman) was observed between ctDNA and subcutaneous tumor volume (LINE-1, r = .94 and Alu, r = .95, p < .0001), ctDNA and bioluminescence (LINE-1, r = .66 and Alu, r = .60, p < .002), and bioluminescence and tumor volume (r = .66, p = .0003). Bioluminescence and ctDNA were also significantly correlated in intratibial tumors (LINE-1, r = .82 and Alu, r = .81, p < .0001). Following external beam IR, the tumor responses varied briefly by method of measurement, but followed a similar trend. Statistically significant correlations were maintained between ctDNA and slide caliper measurement in irradiated subcutaneous tumors (LINE-1, r = .64 and Alu, r = .44, p < .02), and ctDNA and bioluminescence in intratibial tumors (LINE-1, r = .55, p = .018). CONCLUSIONS Real-time qPCR of circulating human Alu and LINE-1 DNA provides an accurate measurement of subcutaneous and intratibial xenograft burden that is comparable with conventional bioluminescence imaging and slide caliper measurement. Transient differences in measurements were observed following tumor-targeted IR, but overall all measurements mirrored tumor growth and response.
Collapse
Affiliation(s)
- Alok Mishra
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenji Zennami
- Department of Urology, The James Buchanan Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel L J Thorek
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Radiology and Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Srinivasan Yegnasubramanian
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Theodore L DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, The James Buchanan Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shawn E Lupold
- Department of Urology, The James Buchanan Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Vangala D, Ladigan S, Liffers ST, Noseir S, Maghnouj A, Götze TM, Verdoodt B, Klein-Scory S, Godfrey L, Zowada MK, Huerta M, Edelstein DL, de Villarreal JM, Marqués M, Kumbrink J, Jung A, Schiergens T, Werner J, Heinemann V, Stintzing S, Lindoerfer D, Mansmann U, Pohl M, Teschendorf C, Bernhardt C, Wolters H, Stern J, Usta S, Viebahn R, Admard J, Casadei N, Fröhling S, Ball CR, Siveke JT, Glimm H, Tannapfel A, Schmiegel W, Hahn SA. Secondary resistance to anti-EGFR therapy by transcriptional reprogramming in patient-derived colorectal cancer models. Genome Med 2021; 13:116. [PMID: 34271981 PMCID: PMC8283888 DOI: 10.1186/s13073-021-00926-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The development of secondary resistance (SR) in metastatic colorectal cancer (mCRC) treated with anti-epidermal growth factor receptor (anti-EGFR) antibodies is not fully understood at the molecular level. Here we tested in vivo selection of anti-EGFR SR tumors in CRC patient-derived xenograft (PDX) models as a strategy for a molecular dissection of SR mechanisms. METHODS We analyzed 21 KRAS, NRAS, BRAF, and PI3K wildtype CRC patient-derived xenograft (PDX) models for their anti-EGFR sensitivity. Furthermore, 31 anti-EGFR SR tumors were generated via chronic in vivo treatment with cetuximab. A multi-omics approach was employed to address molecular primary and secondary resistance mechanisms. Gene set enrichment analyses were used to uncover SR pathways. Targeted therapy of SR PDX models was applied to validate selected SR pathways. RESULTS In vivo anti-EGFR SR could be established with high efficiency. Chronic anti-EGFR treatment of CRC PDX tumors induced parallel evolution of multiple resistant lesions with independent molecular SR mechanisms. Mutations in driver genes explained SR development in a subgroup of CRC PDX models, only. Transcriptional reprogramming inducing anti-EGFR SR was discovered as a common mechanism in CRC PDX models frequently leading to RAS signaling pathway activation. We identified cAMP and STAT3 signaling activation, as well as paracrine and autocrine signaling via growth factors as novel anti-EGFR secondary resistance mechanisms. Secondary resistant xenograft tumors could successfully be treated by addressing identified transcriptional changes by tailored targeted therapies. CONCLUSIONS Our study demonstrates that SR PDX tumors provide a unique platform to study molecular SR mechanisms and allow testing of multiple treatments for efficient targeting of SR mechanisms, not possible in the patient. Importantly, it suggests that the development of anti-EGFR tolerant cells via transcriptional reprogramming as a cause of anti-EGFR SR in CRC is likely more prevalent than previously anticipated. It emphasizes the need for analyses of SR tumor tissues at a multi-omics level for a comprehensive molecular understanding of anti-EGFR SR in CRC.
Collapse
Affiliation(s)
- Deepak Vangala
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780, Bochum, Germany
- Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany
| | - Swetlana Ladigan
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780, Bochum, Germany
- Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany
| | - Sven T Liffers
- Institute of Pathology, Ruhr University of Bochum, Bochum, Germany
- Present Address Division of Solid Tumor Translational Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Soha Noseir
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780, Bochum, Germany
| | - Abdelouahid Maghnouj
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780, Bochum, Germany
| | - Tina-Maria Götze
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780, Bochum, Germany
| | | | - Susanne Klein-Scory
- Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany
| | - Laura Godfrey
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Martina K Zowada
- Translational Functional Cancer Genomics, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Mario Huerta
- Translational Functional Cancer Genomics, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), Dresden, and German Cancer Research Center (DKFZ), Dresden, Germany
| | | | | | - Miriam Marqués
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Jörg Kumbrink
- Institute of Pathology, Ludwig Maximilian University (LMU), Munich, Germany
- German Cancer Consortium (DKTK, partner site Munich), Munich, Germany
| | - Andreas Jung
- Institute of Pathology, Ludwig Maximilian University (LMU), Munich, Germany
- German Cancer Consortium (DKTK, partner site Munich), Munich, Germany
| | - Tobias Schiergens
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology, and Tumor Immunology (CCM) Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Doris Lindoerfer
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ulrich Mansmann
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Pohl
- Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany
| | | | | | - Heiner Wolters
- Department of Visceral and General Surgery, St. Josef Hospital, Dortmund, Germany
| | - Josef Stern
- Department of Visceral and General Surgery, St. Josef Hospital, Dortmund, Germany
| | - Selami Usta
- Department of Visceral and General Surgery, St. Josef Hospital, Dortmund, Germany
| | - Richard Viebahn
- Department of Surgery, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany
| | - Jacob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Stefan Fröhling
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Deptartment of Translational Medical Oncology, NCT Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Claudia R Ball
- Translational Functional Cancer Genomics, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), Dresden, and German Cancer Research Center (DKFZ), Dresden, Germany
- Center for Personalized Oncology, NCT Dresden and University Hospital Carl Gustav Carus Dresden at TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Hanno Glimm
- Translational Functional Cancer Genomics, NCT Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT), Dresden, and German Cancer Research Center (DKFZ), Dresden, Germany
- Center for Personalized Oncology, NCT Dresden and University Hospital Carl Gustav Carus Dresden at TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
| | - Andrea Tannapfel
- Institute of Pathology, Ruhr University of Bochum, Bochum, Germany
| | - Wolff Schmiegel
- Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany
| | - Stephan A Hahn
- Department of Molecular GI Oncology, Faculty of Medicine, Ruhr University Bochum, 44780, Bochum, Germany.
- Department of Internal Medicine, Ruhr University Bochum, Knappschaftskrankenhaus, Bochum, Germany.
| |
Collapse
|
23
|
Labgaa I, von Felden J, Craig AJ, Martins-Filho SN, Villacorta-Martin C, Demartines N, Dormond O, D'Avola D, Villanueva A. Experimental Models of Liquid Biopsy in Hepatocellular Carcinoma Reveal Clone-Dependent Release of Circulating Tumor DNA. Hepatol Commun 2021; 5:1095-1105. [PMID: 34141992 PMCID: PMC8183169 DOI: 10.1002/hep4.1692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/05/2021] [Accepted: 01/24/2021] [Indexed: 01/12/2023] Open
Abstract
Liquid biopsy, the molecular analysis of tumor components released into the bloodstream, has emerged as a noninvasive and resourceful means to access genomic information from cancers. Most data derived from translational studies showcase its numerous potential clinical applications. However, data from experimental models are scarce, and little is known about the underlying mechanisms and factors controlling the release of circulating tumor DNA (ctDNA) and cells (CTCs). This study aimed to model liquid biopsy in hepatocellular carcinoma xenografts and to study the dynamics of release of ctDNA and CTCs; this included models of intratumoral heterogeneity (ITH) and metastatic disease. We quantified ctDNA by quantitative polymerase chain reaction (PCR) targeting human long interspersed nuclear element group 1; targeted mutation analysis was performed with digital droplet PCR. CTCs were traced by flow cytometry. Results demonstrated the feasibility of detecting ctDNA, including clone-specific mutations, as well as CTCs in blood samples of mice. In addition, the concentration of ctDNA and presence of tumor-specific mutations reflected tumor progression, and detection of CTCs was associated with metastases. Our ITH model suggested differences in the release of DNA fragments impacted by the cell-clone origin and the treatment. Conclusion: These data present new models to study liquid biopsy and its underlying mechanisms and highlighted a clone-dependent release of ctDNA into the bloodstream.
Collapse
Affiliation(s)
- Ismail Labgaa
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,Department of Visceral SurgeryLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Johann von Felden
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,1. Department of Internal MedicineUniversity Medical Center Hamburg EppendorfHamburgGermany
| | - Amanda J Craig
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,Liver Cancer ProgramCenter for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Sebastiao N Martins-Filho
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,Department of Pathology and Laboratory MedicineUniversity Health NetworkUniversity of TorontoTorontoONCanada.,Departamento de PatologiaFaculdade de Medicina da Universidade de São PauloUniversidade de São PauloSão PauloBrazil
| | - Carlos Villacorta-Martin
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,Center for Regenerative MedicineBoston University and Boston Medical CenterBostonMAUSA
| | - Nicolas Demartines
- Department of Visceral SurgeryLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Olivier Dormond
- Department of Visceral SurgeryLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Delia D'Avola
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,Liver UnitClinica Universidad de NavarraPamplonaSpain.,Liver UnitClinica Universidad de NavarraMadridSpain.,Centro de Investigacion Biomédica en Red de Enfermedades Hepáticas y DigestivasPamplonaSpain
| | - Augusto Villanueva
- Division of Liver DiseasesLiver Cancer ProgramTisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA.,Division of Hematology/OncologyDepartment of MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
24
|
Noh JM, Kim YJ, Lee HY, Choi C, Ahn WG, Lee T, Pyo H, Park JH, Park D, Park WY. Targeted Liquid Biopsy Using Irradiation to Facilitate the Release of Cell-Free DNA from a Spatially Aimed Tumor Tissue. Cancer Res Treat 2021; 54:40-53. [PMID: 34044476 PMCID: PMC8756125 DOI: 10.4143/crt.2021.151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/24/2021] [Indexed: 11/22/2022] Open
Abstract
Purpose We investigated the feasibility of using an anatomically localized, target-enriched liquid biopsy (TLB) in mouse models of lung cancer. Materials and Methods After irradiating xenograft mouse with human lung cancer cell lines, H1299 (NRAS proto-oncogene, GTPase [NRAS] Q61K) and HCC827 (epidermal growth factor receptor [EGFR] E746–750del), circulating (cell-free) tumor DNA (ctDNA) levels were monitored with quantitative polymerase chain reaction on human long interspersed nuclear element-1 and cell line-specific mutations. We checked dose-dependency at 6, 12, or 18 Gy to each tumor-bearing mouse leg using 6-MV photon beams. We also analyzed ctDNA of lung cancer patients by LiquidSCAN, a targeted deep sequencing to validated the clinical performances of TLB method. Results Irradiation could enhance the detection sensitivity of NRAS Q61K in the plasma sample of H1299-xenograft mouse to 4.5-fold. While cell-free DNA (cfDNA) level was not changed at 6 Gy, ctDNA level was increased upon irradiation. Using double-xenograft mouse with H1299 and HCC827, ctDNA polymerase chain reaction analysis with local irradiation in each region could specify mutation type matched to transplanted cell types, proposing an anatomically localized, TLB. Furthermore, when we performed targeted deep sequencing of cfDNA to monitor ctDNA level in 11 patients with lung cancer who underwent radiotherapy, the average ctDNA level was increased within a week after the start of radiotherapy. Conclusion TLB using irradiation could temporarily amplify ctDNA release in xenograft mouse and lung cancer patients, which enables us to develop theragnostic method for cancer patients with accurate ctDNA detection.
Collapse
Affiliation(s)
- Jae Myoung Noh
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeon Jeong Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Changhoon Choi
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won-Gyun Ahn
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Taeseob Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,GENINUS Inc, Seoul, Korea
| | - Hongryull Pyo
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jee Hyun Park
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Donghyun Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,GENINUS Inc, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
25
|
Detection of low-frequency DNA variants by targeted sequencing of the Watson and Crick strands. Nat Biotechnol 2021; 39:1220-1227. [PMID: 33941929 DOI: 10.1038/s41587-021-00900-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Identification and quantification of low-frequency mutations remain challenging despite improvements in the baseline error rate of next-generation sequencing technologies. Here, we describe a method, termed SaferSeqS, that addresses these challenges by (1) efficiently introducing identical molecular barcodes in the Watson and Crick strands of template molecules and (2) enriching target sequences with strand-specific PCR. The method achieves high sensitivity and specificity and detects variants at frequencies below 1 in 100,000 DNA template molecules with a background mutation rate of <5 × 10-7 mutants per base pair (bp). We demonstrate that it can evaluate mutations in a single amplicon or simultaneously in multiple amplicons, assess limited quantities of cell-free DNA with high recovery of both strands and reduce the error rate of existing PCR-based molecular barcoding approaches by >100-fold.
Collapse
|
26
|
Nakatsuka T, Nakagawa H, Hayata Y, Wake T, Yamada T, Nishibatake Kinoshita M, Nakagomi R, Sato M, Minami T, Uchino K, Enooku K, Kudo Y, Tanaka Y, Kishikawa T, Otsuka M, Tateishi R, Koike K. Post-treatment cell-free DNA as a predictive biomarker in molecular-targeted therapy of hepatocellular carcinoma. J Gastroenterol 2021; 56:456-469. [PMID: 33712873 DOI: 10.1007/s00535-021-01773-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/27/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Liquid biopsies, particularly those involving circulating tumor DNA (ctDNA), are rapidly emerging as a non-invasive alternative to tumor biopsies. However, clinical applications of ctDNA analysis in hepatocellular carcinoma (HCC) have not been fully elucidated. METHODS We measured the amount of plasma-derived cell-free DNA (cfDNA) in HCC patients before (n = 100) and a few days after treatment (n = 87), including radiofrequency ablation, transarterial chemoembolization, and molecular-targeted agents (MTAs), and prospectively analyzed their associations with clinical parameters and prognosis. TERT promoter mutations in cfDNA were analyzed using droplet digital PCR. Furthermore, we performed a comprehensive mutational analysis of post-treatment cfDNA via targeted ultra-deep sequencing (22,000× coverage) in a panel of 275 cancer-related genes in selected patients. RESULTS Plasma cfDNA levels increased significantly according to HCC clinical stage, and a high cfDNA level was independently associated with a poor prognosis. TERT promoter mutations were detected in 45% of all cases but were not associated with any clinical characteristics. cfDNA levels increased significantly a few days after treatment, and a greater increase in post-treatment cfDNA levels was associated with a greater therapeutic response to MTAs. The detection rate of TERT mutations increased to 57% using post-treatment cfDNA, suggesting that the ctDNA was enriched. Targeted ultra-deep sequencing using post-treatment cfDNA after administering lenvatinib successfully detected various gene mutations and obtained promising results in lenvatinib-responsive cases. CONCLUSIONS Post-treatment cfDNA analysis may facilitate the construction of biomarkers for predicting MTA treatment effects.
Collapse
Affiliation(s)
- Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yuki Hayata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taijiro Wake
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoharu Yamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mizuki Nishibatake Kinoshita
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Ryo Nakagomi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Clinical Laboratory Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tatsuya Minami
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Koji Uchino
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kenichiro Enooku
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yotaro Kudo
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasuo Tanaka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takahiro Kishikawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
27
|
Rostami A, Lambie M, Yu CW, Stambolic V, Waldron JN, Bratman SV. Senescence, Necrosis, and Apoptosis Govern Circulating Cell-free DNA Release Kinetics. Cell Rep 2021; 31:107830. [PMID: 32610131 DOI: 10.1016/j.celrep.2020.107830] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 02/22/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022] Open
Abstract
The kinetics of circulating cell-free DNA (cfDNA) release may provide a real-time assessment of induced cell death. However, there is a limited understanding of the underlying biological rationale for cfDNA release following distinct treatments and cell death mechanisms. Here, we uncover a complex interplay between apoptosis, necrosis, and senescence in determining cfDNA release kinetics. Utilizing multiple in vitro and in vivo preclinical models, we show how cfDNA release is modulated through a combination of apoptotic and senescent triggers and inhibitors. Interestingly, we identify treatment-induced senescence as a previously unrecognized determinant of cfDNA kinetics that can counteract its release. Necrosis is the predominant cell death mechanism that consistently contributes to cfDNA release in response to ionizing radiation, and, surprisingly, apoptosis plays a comparatively minor role in some tumors. Based on our results, we propose a model to explain cfDNA release from cells over time, with important implications for future studies.
Collapse
Affiliation(s)
- Ariana Rostami
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Meghan Lambie
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Caberry W Yu
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Vuk Stambolic
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - John N Waldron
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Radiation Oncology, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Radiation Oncology, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada.
| |
Collapse
|
28
|
Perillo A, Agbaje Olufemi MV, De Robbio J, Mancuso RM, Roscigno A, Tirozzi M, Scognamiglio IR. Liquid biopsy in NSCLC: a new challenge in radiation therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:156-173. [PMID: 36046142 PMCID: PMC9400754 DOI: 10.37349/etat.2021.00038] [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: 05/24/2020] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the most common cancer and the leading cause of cancer mortality worldwide. To date, tissue biopsy has been the gold standard for the diagnosis and the identification of specific molecular mutations, to guide choice of therapy. However, this procedure has several limitations. Liquid biopsy could represent a solution to the intrinsic limits of traditional biopsy. It can detect cancer markers such as circulating tumor DNA or RNA (ctDNA, ctRNA), and circulating tumor cells, in plasma, serum or other biological fluids. This procedure is minimally invasive, reproducible and can be used repeatedly. The main clinical applications of liquid biopsy in non-small cell lung cancer (NSCLC) patients are the early diagnosis, stratification of the risk of relapse, identification of mutations to guide application of targeted therapy and the evaluation of the minimum residual disease. In this review, the current role of liquid biopsy and associated markers in the management of NSCLC patients was analyzed, with emphasis on ctDNA and CTCs, and radiotherapy.
Collapse
Affiliation(s)
- Annarita Perillo
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Mohamed Vincenzo Agbaje Olufemi
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Jacopo De Robbio
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Rossella Margherita Mancuso
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Anna Roscigno
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Maddalena Tirozzi
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| | - Ida Rosalia Scognamiglio
- Department of Advanced Biomedical Sciences, University “Federico II” School of Medicine, Via Sergio Pansini 5, 80131 Napoli, Italy
| |
Collapse
|
29
|
Rostami A, Yu C, Bratman SV. Serial Cell-free DNA Assessments in Preclinical Models. STAR Protoc 2020; 1:100145. [PMID: 33377039 PMCID: PMC7757313 DOI: 10.1016/j.xpro.2020.100145] [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] [Indexed: 11/09/2022] Open
Abstract
Studying circulating cell-free DNA (cfDNA) release within preclinical model systems provides opportunities to investigate the mechanisms and kinetics underlying this process under various conditions. We present a detailed protocol for longitudinal evaluation of cfDNA release through (1) seeding of cancer cell lines and establishment of xenograft tumors, (2) treatment of cancer cells and xenograft tumors, (3) serial collection of cell line media and xenograft blood, and (4) processing and isolation of cfDNA for (5) quantification of cfDNA by quantitative PCR. For complete details on the use and execution of this protocol please refer to Rostami et al. (2020). Cell line media and xenograft blood can be serially collected after irradiation Small plasma volumes can be simultaneously purified for cell-free DNA (cfDNA) Cell line media can be diluted for quantification of cfDNA using quantitative PCR Human LINE-1 can be used for ultrasensitive detection of cfDNA
Collapse
Affiliation(s)
- Ariana Rostami
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Caberry Yu
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Center, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada.,Department of Radiation Oncology, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
30
|
Gardner L, Warrington J, Rogan J, Rothwell DG, Brady G, Dive C, Kostarelos K, Hadjidemetriou M. The biomolecule corona of lipid nanoparticles contains circulating cell-free DNA. NANOSCALE HORIZONS 2020; 5:1476-1486. [PMID: 32853302 DOI: 10.1039/d0nh00333f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The spontaneous adsorption of biomolecules onto the surface of nanoparticles (NPs) in complex physiological biofluids has been widely investigated over the last decade. Characterisation of the protein composition of the 'biomolecule corona' has dominated research efforts, whereas other classes of biomolecules, such as nucleic acids, have received no interest. Scarce, speculative statements exist in the literature about the presence of nucleic acids in the biomolecule corona, with no previous studies attempting to describe the contribution of genomic content to the blood-derived NP corona. Herein, we provide the first experimental evidence of the interaction of circulating cell-free DNA (cfDNA) with lipid-based NPs upon their incubation with human plasma samples, obtained from healthy volunteers and ovarian carcinoma patients. Our results also demonstrate an increased amount of detectable cfDNA in patients with cancer. Proteomic analysis of the same biomolecule coronas revealed the presence of histone proteins, suggesting an indirect, nucleosome-mediated NP-cfDNA interaction. The finding of cfDNA as part of the NP corona, offers a previously unreported new scope regarding the chemical composition of the 'biomolecule corona' and opens up new possibilities for the potential exploitation of the biomolecule corona for the enrichment and analysis of blood-circulating nucleic acids.
Collapse
Affiliation(s)
- Lois Gardner
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, The University of Manchester, AV Hill Building, Manchester, UK.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Loree JM, Dowers A, Tu D, Jonker DJ, Edelstein DL, Quinn H, Holtrup F, Price T, Zalcberg JR, Moore MJ, Karapetis CS, O'Callaghan CJ, Waring P, Kennecke HF, Hamilton SR, Kopetz S. Expanded Low Allele Frequency RAS and BRAF V600E Testing in Metastatic Colorectal Cancer as Predictive Biomarkers for Cetuximab in the Randomized CO.17 Trial. Clin Cancer Res 2020; 27:52-59. [PMID: 33087330 DOI: 10.1158/1078-0432.ccr-20-2710] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/31/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Expanded RAS/BRAF mutations have not been assessed as predictive for single-agent cetuximab in metastatic colorectal cancer (mCRC), and low mutant allele frequency (MAF) mutations are of unclear significance. We aimed to establish cetuximab efficacy in optimally selected patients using highly sensitive beads, emulsion, amplification, and magnetics (BEAMing) analysis, capable of detecting alterations below standard clinical assays. PATIENTS AND METHODS CO.17 trial compared cetuximab versus best supportive care (BSC) in RAS/BRAF-unselected mCRC. We performed RAS/BRAF analysis on microdissected tissue of 242 patients in CO.17 trial using BEAMing for KRAS/NRAS (codons 12/13/59/61/117/146) and BRAF V600E. Patients without BEAMing but with previous Sanger sequencing-detected mutations were included. RESULTS KRAS, NRAS, and BRAF mutations were present in 53%, 4%, and 3% of tumors, respectively. Cetuximab improved overall survival [OS; HR, 0.51; 95% confidence interval (CI), 0.32-0.81; P = 0.004] and progression-free survival (PFS; HR, 0.25; 95% CI, 0.15-0.41; P < 0.0001) compared with BSC in RAS/BRAF wild-type patients. Cetuximab did not improve OS/PFS for KRAS-, NRAS-, or BRAF-mutated tumors, and tests of interaction confirmed expanded KRAS (P = 0.0002) and NRAS (P = 0.006) as predictive, while BRAF mutations were not (P = 0.089). BEAMing identified 14% more tumors as RAS mutant than Sanger sequencing, and cetuximab lacked activity in these patients. Mutations at MAF < 5% were noted in 6 of 242 patients (2%). One patient with a KRAS A59T mutation (MAF = 2%) responded to cetuximab. More NRAS than KRAS mutations were low MAF (OR, 20.50; 95% CI, 3.88-96.85; P = 0.0038). CONCLUSIONS We establish single-agent cetuximab efficacy in optimally selected patients and show that subclonal RAS/BRAF alterations are uncommon and remain of indeterminate significance.
Collapse
Affiliation(s)
- Jonathan M Loree
- BC Cancer, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anthony Dowers
- The University of Melbourne, Melbourne, Victoria, Australia
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | - Derek J Jonker
- The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | - Timothy Price
- Queen Elizabeth Hospital, Adelaide, South Australia, Australia
| | | | - Malcolm J Moore
- BC Cancer, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Paul Waring
- The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Scott Kopetz
- University of Texas, MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
32
|
Liu HE, Vuppalapaty M, Wilkerson C, Renier C, Chiu M, Lemaire C, Che J, Matsumoto M, Carroll J, Crouse S, Hanft VR, Jeffrey SS, Di Carlo D, Garon EB, Goldman J, Sollier E. Detection of EGFR Mutations in cfDNA and CTCs, and Comparison to Tumor Tissue in Non-Small-Cell-Lung-Cancer (NSCLC) Patients. Front Oncol 2020; 10:572895. [PMID: 33117705 PMCID: PMC7578230 DOI: 10.3389/fonc.2020.572895] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) therapies, based on the evaluation of EGFR mutations, have shown dramatic clinical benefits. EGFR mutation assays are mainly performed on tumor biopsies, which carry risks, are not always successful and give results relevant to the timepoint of the assay. To detect secondary EGFR mutations, which cause resistance to 1st and 2nd generation TKIs and lead to the administration of a 3rd generation drug, effective and non-invasive monitoring of EGFR mutation status is needed. Liquid biopsy analytes, such as circulating tumor cells (CTCs) and circulating tumor DNA (cfDNA), allow such monitoring over the course of the therapy. The aim of this study was to develop and optimize a workflow for the evaluation of cfDNA and CTCs in NSCLC patients all from one blood sample. Using Vortex technology and EntroGen ctEGFR assay, EGFR mutations were identified at 0.5 ng of DNA (∼83 cells), with a sensitivity ranging from 0.1 to 2.0% for a total DNA varying from 25 ng (∼4 CTCs among 4000 white blood cells, WBCs) to 1 ng (∼4 CTCs among 200 WBCs). The processing of plasma-depleted-blood provided comparable capture recovery as whole blood, confirming the possibility of a multimodality liquid biopsy analysis (cfDNA and CTC DNA) from a single tube of blood. Different anticoagulants were evaluated and compared in terms of respective performance. Blood samples from 24 NSCLC patients and 6 age-matched healthy donors were analyzed with this combined workflow to minimize blood volume needed and sample-to-sample bias, and the EGFR mutation profile detected from CTCs and cfDNA was compared to matched tumor tissues. Despite the limited size of the patient cohort, results from this non-invasive EGFR mutation analysis are encouraging and this combined workflow represents a valuable means for informing therapy selection and for monitoring treatment of patients with NSCLC.
Collapse
Affiliation(s)
- Haiyan E Liu
- Vortex Biosciences, Inc., Pleasanton, CA, United States
| | | | | | | | - Michael Chiu
- Vortex Biosciences, Inc., Pleasanton, CA, United States
| | | | - James Che
- Vortex Biosciences, Inc., Pleasanton, CA, United States
| | - Melissa Matsumoto
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - James Carroll
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Steve Crouse
- Vortex Biosciences, Inc., Pleasanton, CA, United States
| | - Violet R Hanft
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,California NanoSystems Institute, Los Angeles, CA, United States.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Edward B Garon
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jonathan Goldman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States
| | | |
Collapse
|
33
|
Diao F. Targeted Sequencing Analysis of Matched Cell-Free DNA and White Blood Cells: A Facile Method for Detection of Residual Disease in Gastric Cancer. Glob Med Genet 2020; 7:27-29. [PMID: 32939511 PMCID: PMC7490118 DOI: 10.1055/s-0040-1716333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Feiyu Diao
- Department of General Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
34
|
Yates JWT, Byrne H, Chapman SC, Chen T, Cucurull-Sanchez L, Delgado-SanMartin J, Di Veroli G, Dovedi SJ, Dunlop C, Jena R, Jodrell D, Martin E, Mercier F, Ramos-Montoya A, Struemper H, Vicini P. Opportunities for Quantitative Translational Modeling in Oncology. Clin Pharmacol Ther 2020; 108:447-457. [PMID: 32569424 DOI: 10.1002/cpt.1963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022]
Abstract
A 2-day meeting was held by members of the UK Quantitative Systems Pharmacology Network () in November 2018 on the topic of Translational Challenges in Oncology. Participants from a wide range of backgrounds were invited to discuss current and emerging modeling applications in nonclinical and clinical drug development, and to identify areas for improvement. This resulting perspective explores opportunities for impactful quantitative pharmacology approaches. Four key themes arose from the presentations and discussions that were held, leading to the following recommendations: Evaluate the predictivity and reproducibility of animal cancer models through precompetitive collaboration. Apply mechanism of action (MoA) based mechanistic models derived from nonclinical data to clinical trial data. Apply MoA reflective models across trial data sets to more robustly quantify the natural history of disease and response to differing interventions. Quantify more robustly the dose and concentration dependence of adverse events through mathematical modelling techniques and modified trial design.
Collapse
Affiliation(s)
| | | | | | - Tao Chen
- University of Surrey, Surrey, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Heider K, Wan JCM, Hall J, Belic J, Boyle S, Hudecova I, Gale D, Cooper WN, Corrie PG, Brenton JD, Smith CG, Rosenfeld N. Detection of ctDNA from Dried Blood Spots after DNA Size Selection. Clin Chem 2020; 66:697-705. [PMID: 32268361 DOI: 10.1093/clinchem/hvaa050] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/07/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent advances in the study and clinical applications of circulating tumor DNA (ctDNA) are limited by practical considerations of sample collection. Whole-genome sequencing (WGS) is increasingly used for analysis of ctDNA, identifying copy-number alterations and fragmentation patterns. We hypothesized that low-depth/shallow WGS (sWGS) data may be generated from minute amounts of cell-free DNA, and that fragment-size selection may remove contaminating genomic DNA from small blood volumes. Dried blood spots have practical advantages for sample collection, may facilitate serial sampling, and could support novel study designs in humans and animal models. METHODS We developed a protocol for the isolation and analysis of cell-free DNA from dried blood spots using filter paper cards and bead-based size selection. DNA extracted and size-selected from dried spots was analyzed using sWGS and polymerase chain reaction (PCR). RESULTS Analyzing a 50 μL dried blood spot from frozen whole blood of a patient with melanoma, we identified ctDNA based on the presence of tumor-specific somatic copy-number alterations, and found a fragment-size profile similar to that observed in plasma DNA. We found alterations in different chromosomes in blood spots from 2 patients with high-grade serous ovarian carcinoma. Extending this approach to serial dried blood spots from mouse xenograft models, we detect tumor-derived cell-free DNA and identified ctDNA from the originally grafted ascites. CONCLUSION Our data suggest that ctDNA can be detected and monitored in dried blood spots from archived and fresh blood samples, enabling new approaches for sample collection and novel study/trial designs for both patients and in vivo models.
Collapse
Affiliation(s)
- Katrin Heider
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Jonathan C M Wan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - James Hall
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Jelena Belic
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Samantha Boyle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Irena Hudecova
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Davina Gale
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Wendy N Cooper
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Pippa G Corrie
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Christopher G Smith
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cancer Research UK Major Centre-Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| |
Collapse
|
36
|
Ma G, Wang J, Huang H, Han X, Xu J, Veeramootoo JS, Xia T, Wang S. Identification of the plasma total cfDNA level before and after chemotherapy as an indicator of the neoadjuvant chemotherapy response in locally advanced breast cancer. Cancer Med 2020; 9:2271-2282. [PMID: 32017472 PMCID: PMC7131846 DOI: 10.1002/cam4.2906] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/03/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023] Open
Abstract
This study aimed to retrospectively evaluate the circulating free DNA (cfDNA) level in patients with locally advanced breast cancer (LABC) having different neoadjuvant chemotherapy (NCT) responses and to investigate whether dynamic changes in cfDNA level could predict the effectiveness of NCT in patients with LABC. Data on 61 patients with LABC were included. NCT responses were evaluated using the response evaluation criteria. Blood samples were collected for cfDNA detection before treatment and after the first and eighth courses of chemotherapy. The Alu 111‐bp and 260‐bp fragment levels were evaluated by polymerase chain reaction, and the predictive value of the cfDNA level in the NCT response was determined. In vitro, the MCF‐7 and MCF‐7/ADR cell lines were applied to simulate the phenomenon of drug resistance and explain the underlying mechanism. The Alu 111‐bp level increased after the first NCT course (P = .014) and then remained high after NCT in the high‐R group (P = .047), but it remained steady in the low‐R group during NCT. A similar tendency in the Alu 260‐bp level was revealed in different groups. The ∆∆Ct value of Alu 260‐bp had good diagnostic efficiency in assessing predictive ability. The area under the curve for the ∆∆Ct1 and ∆∆Ct2 of Alu 260‐bp was 0.697 and 0.647, respectively. The cfDNA level was closely related to epirubicin‐induced apoptosis and changes in the Ki‐67 index in vitro. The elevation of cfDNA after one chemotherapy cycle was mediated by the apoptosis of tumor cells and related to the improved chemotherapy response.
Collapse
Affiliation(s)
- Ge Ma
- Department of Breast Surgery, The Firsft Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingyi Wang
- Department of Breast Surgery, The Firsft Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Huaxing Huang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Xu Han
- Department of Breast Surgery, The Firsft Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jin Xu
- Department of Breast and Thyroid Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | | | - Tiansong Xia
- Department of Breast Surgery, The Firsft Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shui Wang
- Department of Breast Surgery, The Firsft Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Lenaerts L, Tuveri S, Jatsenko T, Amant F, Vermeesch JR. Detection of incipient tumours by screening of circulating plasma DNA: hype or hope? Acta Clin Belg 2020; 75:9-18. [PMID: 31578135 DOI: 10.1080/17843286.2019.1671653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background: The last half-decade has been marked by a rapid expansion of research efforts in the field of so-called liquid biopsies, thereby investigating the potential of blood-derived cell-free tumour DNA (ctDNA) markers for application in clinical oncological management. The analysis of cfDNA appears to be particularly attractive for therapy monitoring purposes, while in terms of early cancer diagnosis and screening the potentials are just starting to be explored. Challenges, both of biological and technical nature, need to be addressed. One such challenge is to overcome the low levels of ctDNA in the circulation, intrinsic to many early-stage cancers. Methods: Here, we give an overview of the features of ctDNA and the approaches that are currently being applied with the ultimate aim to detect tumours in a presymptomatic stage. Conclusion: Although many studies report encouraging results, further technical development and larger studies are warranted before application of ctDNA analysis may find its place in clinic.
Collapse
Affiliation(s)
| | | | | | - Frédéric Amant
- Department of Oncology, KU Leuven, Leuven, Belgium
- Gynecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium
- Center for Gynecological Oncology Amsterdam, Academic Medical Centre Amsterdam-University of Amsterdam and The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Joris Robert Vermeesch
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Centre of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
38
|
White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun 2020; 11:525. [PMID: 31988276 PMCID: PMC6985115 DOI: 10.1038/s41467-020-14310-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023] Open
Abstract
Liquid biopsies are providing new opportunities for detection of residual disease in cell-free DNA (cfDNA) after surgery but may be confounded through identification of alterations arising from clonal hematopoiesis. Here, we identify circulating tumor-derived DNA (ctDNA) alterations through ultrasensitive targeted sequencing analyses of matched cfDNA and white blood cells from the same patient. We apply this approach to analyze samples from patients in the CRITICS trial, a phase III randomized controlled study of perioperative treatment in patients with operable gastric cancer. After filtering alterations from matched white blood cells, the presence of ctDNA predicts recurrence when analyzed within nine weeks after preoperative treatment and after surgery in patients eligible for multimodal treatment. These analyses provide a facile method for distinguishing ctDNA from other cfDNA alterations and highlight the utility of ctDNA as a predictive biomarker of patient outcome to perioperative cancer therapy and surgical resection in patients with gastric cancer.
Collapse
|
39
|
Alghofaili L, Almubarak H, Gassem K, Islam SS, Coskun S, Kaya N, Karakas B. Cell-free DNA levels of twins and sibling pairs indicate individuality and possible use as a personalized biomarker. PLoS One 2019; 14:e0223470. [PMID: 31600277 PMCID: PMC6786590 DOI: 10.1371/journal.pone.0223470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/22/2019] [Indexed: 01/01/2023] Open
Abstract
Cell-free DNA (cfDNA) in the human blood circulation has been under investigation since its initial observation in 1948. Plasma cfDNA is known to be significantly elevated in diseased people. Due to possible variation in the population, evaluating cfDNA as a non-invasive biomarker at disease onset alone may not be sensitive enough to accurately diagnose diseases, particularly early stage cancers on a personal level. To understand the factors that define the cfDNA levels on the personal level and for better use as a non-invasive biomarker, we isolated cfDNA from the plasma of healthy individuals with varying degrees of genetic and/or environmental similarities (monozygotic twins, dizygotic twins, sibling pairs, and unrelated individuals) as well as from patients with varying stages of breast and ovarian cancer undergoing treatment. Cell-free DNA levels were quantified by a fluorometer (ng/ml) and/or real-time PCR (copies/ml). The associations between individuals with various degrees of genetic and/or environmental similarities and their plasma cfDNA levels were evaluated. The ACE model (A = additive genetic, C = common environment, and E = specific environmental factors) was used to determine the proportion of each factor on the cfDNA levels. We found a high correlation (r = 0.77; p < 0.0001) in plasma cfDNA levels between monozygotic twins (n = 39). However, the correlation was gradually reduced to moderate (r = 0.47; p = 0.016) between dizygotic twins (n = 13) and low correlation (r = 0.28; p = 0.043) between sibling pairs (n = 26). The ACE model analysis showed that the plasma cfDNA level of a given healthy individual is influenced both by genetic and the environmental components in similar proportions (53% and 47%, respectively; A = 53%, C = 22.5%, E = 24.5%). Moreover, while age had no effect, gender significantly influenced the individual’s plasma cfDNA level. As expected, cfDNA levels were significantly higher in both breast (n = 26) (p<0.0001) and ovarian (n = 64) (p<0.0001) cancer patients compared to the healthy individuals. Our study demonstrated that both genome and environmental factors modulate the individual’s cfDNA level suggesting that its diagnostic sensitivity may be improved only if the person’s cfDNA level is known prior to disease presentation.
Collapse
Affiliation(s)
- Lamyaa Alghofaili
- Translational Cancer Research Section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Alfaisal University Medical School, Riyadh, Saudi Arabia
| | - Hannah Almubarak
- Translational Cancer Research Section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Khawlah Gassem
- Translational Cancer Research Section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Syed S Islam
- Translational Cancer Research Section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Serdar Coskun
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Namik Kaya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Bedri Karakas
- Translational Cancer Research Section, Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
40
|
Wang Y, Li L, Douville C, Cohen JD, Yen TT, Kinde I, Sundfelt K, Kjær SK, Hruban RH, Shih IM, Wang TL, Kurman RJ, Springer S, Ptak J, Popoli M, Schaefer J, Silliman N, Dobbyn L, Tanner EJ, Angarita A, Lycke M, Jochumsen K, Afsari B, Danilova L, Levine DA, Jardon K, Zeng X, Arseneau J, Fu L, Diaz LA, Karchin R, Tomasetti C, Kinzler KW, Vogelstein B, Fader AN, Gilbert L, Papadopoulos N. Evaluation of liquid from the Papanicolaou test and other liquid biopsies for the detection of endometrial and ovarian cancers. Sci Transl Med 2019; 10:10/433/eaap8793. [PMID: 29563323 DOI: 10.1126/scitranslmed.aap8793] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/14/2018] [Indexed: 12/21/2022]
Abstract
We report the detection of endometrial and ovarian cancers based on genetic analyses of DNA recovered from the fluids obtained during a routine Papanicolaou (Pap) test. The new test, called PapSEEK, incorporates assays for mutations in 18 genes as well as an assay for aneuploidy. In Pap brush samples from 382 endometrial cancer patients, 81% [95% confidence interval (CI), 77 to 85%] were positive, including 78% of patients with early-stage disease. The sensitivity in 245 ovarian cancer patients was 33% (95% CI, 27 to 39%), including 34% of patients with early-stage disease. In contrast, only 1.4% of 714 women without cancer had positive Pap brush samples (specificity, ~99%). Next, we showed that intrauterine sampling with a Tao brush increased the detection of malignancy over endocervical sampling with a Pap brush: 93% of 123 (95% CI, 87 to 97%) patients with endometrial cancer and 45% of 51 (95% CI, 31 to 60%) patients with ovarian cancer were positive, whereas none of the samples from 125 women without cancer were positive (specificity, 100%). Finally, in 83 ovarian cancer patients in whom plasma was available, circulating tumor DNA was found in 43% of patients (95% CI, 33 to 55%). When plasma and Pap brush samples were both tested, the sensitivity for ovarian cancer increased to 63% (95% CI, 51 to 73%). These results demonstrate the potential of mutation-based diagnostics to detect gynecologic cancers at a stage when they are more likely to be curable.
Collapse
Affiliation(s)
- Yuxuan Wang
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lu Li
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christopher Douville
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joshua D Cohen
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ting-Tai Yen
- Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | | | - Karin Sundfelt
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Susanne K Kjær
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Rigshospitalet, Copenhagen 2100, Denmark.,Unit of Virus, Lifestyle, and Genes, Danish Cancer Society Research Center, Copenhagen 2100, Denmark
| | - Ralph H Hruban
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ie-Ming Shih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert J Kurman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Simeon Springer
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Janine Ptak
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Maria Popoli
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joy Schaefer
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Natalie Silliman
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lisa Dobbyn
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Edward J Tanner
- Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Ana Angarita
- Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Maria Lycke
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Kirsten Jochumsen
- Department of Obstetrics and Gynecology, Odense University Hospital, Odense 5000, Denmark
| | - Bahman Afsari
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ludmila Danilova
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Douglas A Levine
- Department of Gynecologic Oncology, Laura and Isaac Perlmutter Cancer Centre, New York University Langone Medical Center, New York, NY 10016, USA
| | - Kris Jardon
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, Oncology, and Pathology, McGill University and McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Xing Zeng
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, Oncology, and Pathology, McGill University and McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Jocelyne Arseneau
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, Oncology, and Pathology, McGill University and McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Lili Fu
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, Oncology, and Pathology, McGill University and McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Luis A Diaz
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rachel Karchin
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Cristian Tomasetti
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Kenneth W Kinzler
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Bert Vogelstein
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA. .,Howard Hughes Medical Institute, Baltimore, MD 21287, USA
| | - Amanda N Fader
- Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
| | - Lucy Gilbert
- Division of Gynecologic Oncology, Departments of Obstetrics and Gynecology, Oncology, and Pathology, McGill University and McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics, and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
41
|
Rodriguez BJ, Córdoba GD, Aranda AG, Álvarez M, Vicioso L, Pérez CL, Hernando C, Bermejo B, Parreño AJ, Lluch A, Ryder MB, Jones FS, Fredebohm J, Holtrup F, Queipo-Ortuño MI, Alba E. Detection of TP53 and PIK3CA Mutations in Circulating Tumor DNA Using Next-Generation Sequencing in the Screening Process for Early Breast Cancer Diagnosis. J Clin Med 2019; 8:jcm8081183. [PMID: 31394872 PMCID: PMC6723048 DOI: 10.3390/jcm8081183] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 01/27/2023] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a non-invasive "liquid biopsy" for early breast cancer diagnosis. We evaluated the suitability of ctDNA analysis in the diagnosis of early breast cancer after mammography findings, comparing PIK3CA and TP53 mutations between tumor biopsies and pre-biopsy circulating DNA. Matched plasma and frozen fresh tissue biopsies from patients with Breast Imaging-Reporting and Data System (BIRADS) 4c/5 mammography findings and subsequent diagnosis of primary breast cancer were analyzed using NGS TruSeq Custom Amplicon Low Input Panel (Illumina) and plasma SafeSEQ (Sysmex Inostics). The same plasma and tumor mutations were observed in eight of 29 patients (27.6%) with four in TP53 and five in PIK3CA mutations. Sequencing analysis also revealed four additional ctDNA mutations (three in TP53 and one in PIK3CA) previously not identified in three patients tissue biopsy. One of these patients had mutations in both genes. Age, tumor grade and size, immunohistochemical (IHC) subtype, BIRADS category, and lymph node positivity were significantly associated with the detectability of these blood tumor-derived mutations. In conclusion, ctDNA analysis could be used in early breast cancer diagnosis, providing critical clinical information to improve patient diagnosis.
Collapse
Affiliation(s)
- Begona Jimenez Rodriguez
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain
| | - Gema Diaz Córdoba
- Radiology Department, Hospital Clínico Universitario Virgen de la Victoria de Málaga, 29010 Málaga, Spain
| | - Alicia Garrido Aranda
- Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain
| | - Martina Álvarez
- Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain
| | - Luis Vicioso
- Histopathology Department, Hospital Clínico Universitario Virgen de la Victoria de Málaga, 29010 Málaga, Spain
| | - Casilda Llácer Pérez
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain
| | - Cristina Hernando
- Hospital Clínico de Valencia, Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
| | - Begoña Bermejo
- Hospital Clínico de Valencia, Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
| | - Ana Julve Parreño
- Hospital Clínico de Valencia, Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
| | - Ana Lluch
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
- Hospital Clínico de Valencia, Universidad de Valencia, Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
| | | | | | | | | | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, 29010 Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain.
| | - Emilio Alba
- Unidad de Gestión Clínica Intercentros de Oncología Medica, Hospitales Universitarios Regional y Virgen de la Victoria de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, 29010 Málaga, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
42
|
Hamfjord J, Guren TK, Dajani O, Johansen JS, Glimelius B, Sorbye H, Pfeiffer P, Lingjærde OC, Tveit KM, Kure EH, Pallisgaard N, Spindler KLG. Total circulating cell-free DNA as a prognostic biomarker in metastatic colorectal cancer before first-line oxaliplatin-based chemotherapy. Ann Oncol 2019; 30:1088-1095. [PMID: 31046124 DOI: 10.1093/annonc/mdz139] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Metastatic colorectal cancer (mCRC) is a heterogeneous disease where prognosis is dependent both on tumor biology and host factors. Total circulating cell-free DNA (cfDNA) has shown to harbor prognostic information in mCRC, although less is known about the biological correlates of cfDNA levels in this patient group. The primary objective was to evaluate the prognostic value of pretreatment cfDNA in patients receiving the first-line oxaliplatin-based chemotherapy for mCRC, by using a predefined upper limit of normal (ULN) from a cohort of presumed healthy individuals. The secondary objective was to model cfDNA levels as a function of predefined tumor and host factors. PATIENTS AND METHODS This was a retrospective post hoc study based on a prospective multicenter phase III trial, the NORDIC-VII study. DNA was purified from 547 plasma samples and cfDNA quantified by a droplet digital PCR assay (B2M, PPIA) with controls for lymphocyte contamination. Main clinical end point was overall survival (OS). RESULTS cfDNA was quantified in 493 patients, 54 were excluded mainly due to lymphocyte contamination. Median cfDNA level was 7673 alleles/ml (1050-1 645 000) for B2M and 5959 alleles/ml (555-854 167) for PPIA. High cfDNA levels were associated with impaired outcome; median OS of 16.6 months for levels above ULN and 25.9 months for levels below ULN (hazard ratio = 1.83, 95% confidence interval 1.51-2.21, P < 0.001). The result was confirmed in multivariate OS analysis adjusting for established clinicopathological characteristics. A linear regression model predicted cfDNA levels from sum of longest tumor diameters by RECIST, the presence of liver metastases and systemic inflammatory response as measured by interleukin 6 (F(6, 357) = 62.7, P < 0.001). CONCLUSION cfDNA holds promise as a minimally invasive and clinically relevant prognostic biomarker in mCRC before initiating first-line oxaliplatin-based chemotherapy and may be a complex entity associated with tumor burden, liver metastases and systemic inflammatory response. TRIAL REGISTRATION ClinicalTrials.gov, NCT00145314.
Collapse
Affiliation(s)
- J Hamfjord
- Department of Oncology, Oslo University Hospital, Oslo; Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo
| | - T K Guren
- Department of Oncology, Oslo University Hospital, Oslo; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway.
| | - O Dajani
- Department of Oncology, Oslo University Hospital, Oslo
| | - J S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - B Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - H Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - P Pfeiffer
- Department of Oncology, Odense University Hospital, Odense; Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - O C Lingjærde
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo; Department of Computer Science, University of Oslo, Oslo
| | - K M Tveit
- Department of Oncology, Oslo University Hospital, Oslo; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - E H Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo; Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Bø in Telemark, Norway
| | - N Pallisgaard
- Department of Pathology, Zealand University Hospital, Roskilde
| | - K-L G Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
43
|
Bando H, Kagawa Y, Kato T, Akagi K, Denda T, Nishina T, Komatsu Y, Oki E, Kudo T, Kumamoto H, Yamanaka T, Yoshino T. A multicentre, prospective study of plasma circulating tumour DNA test for detecting RAS mutation in patients with metastatic colorectal cancer. Br J Cancer 2019; 120:982-986. [PMID: 31015557 PMCID: PMC6734650 DOI: 10.1038/s41416-019-0457-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/30/2019] [Accepted: 04/04/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND OncoBEAMTM RAS CRC kit using BEAMing technology is a circulating tumour DNA (ctDNA) test for detecting plasma RAS mutational status in metastatic colorectal cancer (mCRC). We conducted a multicentre, prospective study to investigate the concordance of the RAS mutational status between plasma ctDNA and tumour tissue DNA. METHODS mCRC patients without prior anti-EGFR antibodies or regorafenib treatment were enroled. Plasma- and tissue-based RAS mutational status were determined by BEAMing, respectively. RESULTS A total of 280 patients from eight institutions were eligible. The overall agreement between plasma- and tissue-based analyses was 86.4%, with a positive percent agreement of 82.1% and negative percent agreement of 90.4%. From logistic regression analysis, lung metastasis alone indicated the most significant factor associated with discordance. The agreement between plasma- and tissue-based analyses was 64.5% in patients with lung metastasis alone (n = 31) indicating lower amount of ctDNA. Among the cases with lung metastasis alone, all plasma- and tissue-based analyses were perfectly concordant in cases with ≥20 mm of maximum lesion diameter or ≥10 lesions. CONCLUSION The clinical validity of OncoBEAMTM RAS CRC kit was confirmed. Careful attention should be paid for mCRC patients with lung metastases alone having fewer metastases or smaller diameter lesions.
Collapse
Affiliation(s)
- Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Yoshinori Kagawa
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Hyogo, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Osaka, Japan
| | - Kiwamu Akagi
- Division of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Ina, Saitama, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Chiba, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo, Hokkaido, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Toshihiro Kudo
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Kumamoto
- Scientific Affairs Division, Clinical Affairs, Sysmex Corporation, Kobe, Hyogo, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.
| |
Collapse
|
44
|
A Pipeline for Faecal Host DNA Analysis by Absolute Quantification of LINE-1 and Mitochondrial Genomic Elements Using ddPCR. Sci Rep 2019; 9:5599. [PMID: 30944341 PMCID: PMC6447604 DOI: 10.1038/s41598-019-41753-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/13/2019] [Indexed: 12/25/2022] Open
Abstract
Stool contains DNA shed from cells of the gastrointestinal (GI) tract and has great potential as a bio-specimen for non-invasive, nucleic acid-based detection of GI diseases. Whereas methods for studying faecal microbiome DNA are plentiful, there is a lack of well-characterised procedures for stabilisation, isolation, and quantitative analysis of faecal host DNA. We report an optimised pipeline for faecal host DNA analysis from the point-of-collection to droplet digital PCR (ddPCR) absolute quantification of host-specific gene targets. We evaluated multiple methods for preservation and isolation of host DNA from stool to identify the highest performing methods. To quantify host DNA even if present in partially degraded form, we developed sensitive, human-specific short-amplicon ddPCR assays targeting repetitive nuclear genomic elements (LINE-1) and mitochondrial genes. We validated the ability of these optimised methods to perform absolute quantification of host DNA in 200 stool DNA extracts from samples that were serially collected from three healthy individuals and three hospitalised patients. These specimens allowed assessment of host DNA day-to-day variability in stool specimens with widely varying physical characteristics (i.e., Bristol scores). We further extended this approach to mouse stool analysis, to enable faecal host DNA studies in animal disease models as well.
Collapse
|
45
|
Circulating Cell-Free DNA and RNA Analysis as Liquid Biopsy: Optimal Centrifugation Protocol. Cancers (Basel) 2019; 11:cancers11040458. [PMID: 30935089 PMCID: PMC6521186 DOI: 10.3390/cancers11040458] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 03/27/2019] [Indexed: 01/17/2023] Open
Abstract
The combined analysis of circulating cell-free (tumor) DNA (cfDNA/ctDNA) and circulating cell-free (tumor) RNA (cfRNA/ctRNA) shows great promise in determining the molecular profile of cancer patients. Optimization of the workflow is necessary to achieve consistent and reproducible results. In this study, we compared five centrifugation protocols for the optimal yield of both cfDNA/ctDNA and cfRNA/ctRNA. These protocols varied in centrifugation speed, ambient temperature, time, and number of centrifugation steps. Samples from 33 participants were collected in either BD Vacutainer K₂EDTA (EDTA) tubes or cell-free DNA BCT® (Streck) tubes. cfDNA concentration and fragment size, and cfRNA concentration were quantitated in all samples by digital droplet PCR (ddPCR) and quantitative PCR (qPCR). The KRAS-mutated ctDNA and ctRNA fraction was determined via ddPCR. In EDTA tubes, the protocol generating both plasma and platelets was found to produce high quality cfDNA and cfRNA concentrations. Two-step, high-speed centrifugation protocols were associated with high cfDNA but low cfRNA concentrations. High cfRNA concentrations were generated by a one-step, low-speed protocol. However, this coincided with a high amount of genomic DNA (gDNA) contamination. In Streck tubes, two-step, high-speed centrifugation protocols also generated good quality, high cfDNA concentration. However, these tubes are not compatible with cfRNA analysis.
Collapse
|
46
|
Abstract
To our knowledge, this is the first comprehensive study on the influence of several pre-analytical and demographic parameters that could be a source of variability in the quantification of nuclear and mitochondrial circulating DNA (NcirDNA and McirDNA). We report data from a total of 222 subjects, 104 healthy individuals and 118 metastatic colorectal cancer (mCRC) patients. Approximately 50,000 and 3,000-fold more mitochondrial than nuclear genome copies were found in the plasma of healthy individuals and mCRC patients, respectively. In healthy individuals, NcirDNA concentration was statistically influenced by age (p = 0.009) and gender (p = 0.048). Multivariate analysis with logistic regression specified that age over 47 years-old was predictive to have higher NcirDNA concentration (OR = 2.41; p = 0.033). McirDNA concentration was independent of age and gender in healthy individuals. In mCRC patients, NcirDNA and McirDNA levels were independent of age, gender, delay between food intake and blood collection, and plasma aspect, either with univariate or multivariate analysis. Nonetheless, ad hoc study suggested that menopause and blood collection time might have tendency to influence cirDNA quantification. In addition, high significant statistical differences were found between mCRC patients and healthy individuals for NcirDNA (p < 0.0001), McirDNA (p < 0.0001) and McirDNA/NcirDNA ratio (p < 0.0001). NcirDNA and McirDNA levels do not vary in the same way with regards to cancer vs healthy status, pre-analytical and demographic factors.
Collapse
|
47
|
Woff E, Kehagias P, Vandeputte C, Ameye L, Guiot T, Paesmans M, Hendlisz A, Flamen P. Combining 18F-FDG PET/CT-Based Metabolically Active Tumor Volume and Circulating Cell-Free DNA Significantly Improves Outcome Prediction in Chemorefractory Metastatic Colorectal Cancer. J Nucl Med 2019; 60:1366-1372. [PMID: 30850494 DOI: 10.2967/jnumed.118.222919] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 02/11/2019] [Indexed: 12/26/2022] Open
Abstract
Baseline whole-body metabolically active tumor volume (WB-MATV) measured by 18F-FDG PET/CT and circulating cell-free DNA (cfDNA) have been separately validated as predictors of overall and progression-free survival (OS/PFS) in chemorefractory metastatic colorectal cancer (mCRC) patients. This study assessed the correlation between WB-MATV and cfDNA, evaluating the added prognostic value of these in combination, along with clinical parameters. Methods: Of 141 mCRC patients included in a prospective multicenter trial, 132 were evaluable for OS/PFS. cfDNA was extracted from 3 mL of plasma and quantified using a fluorometer. All target lesions were delineated on 18F-FDG PET/CT, and their metabolic volumes were summed to obtain the WB-MATV. Results: Baseline WB-MATV and cfDNA were strongly correlated (r = 0.70; P < 0.001) but showed discordance in 23 of 132 (17%) patients. A multivariate analysis identified 3 independent negative predictors of PFS (high cfDNA, short time since diagnosis, and body mass index < 30) and 5 of OS (high cfDNA, high WB-MATV, body mass index < 30, poor performance status, and short time since diagnosis). Combining WB-MATV and cfDNA increased the overall prognostic value and allowed identification of a subgroup of patients with low cfDNA and high WB-MATV who were associated with intermediate survival (median OS of 8.1 for low-cfDNA/high-MATV patients vs. 12.7 mo for low-cfDNA/low-MATV patients; hazard ratio, 2.04; P = 0.02). Conclusion: This study confirms the added prognostic value of combined circulating cfDNA and PET-based WB-MATV in chemorefractory mCRC patients. The combination of these two biomarkers should provide a firm basis for risk stratification, both in clinical practice and in research trials.
Collapse
Affiliation(s)
- Erwin Woff
- Nuclear Medicine Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pashalina Kehagias
- Gastro-Oncology Translational Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Caroline Vandeputte
- Gastro-Oncology Translational Laboratory, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Lieveke Ameye
- Data Centre, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium; and
| | - Thomas Guiot
- Nuclear Medicine Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marianne Paesmans
- Data Centre, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium; and
| | - Alain Hendlisz
- Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Patrick Flamen
- Nuclear Medicine Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
48
|
Haselmann V, Ahmad-Nejad P, Geilenkeuser WJ, Duda A, Gabor M, Eichner R, Patton S, Neumaier M. Results of the first external quality assessment scheme (EQA) for isolation and analysis of circulating tumour DNA (ctDNA). Clin Chem Lab Med 2019; 56:220-228. [PMID: 28841569 DOI: 10.1515/cclm-2017-0283] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Circulating tumour DNA (ctDNA) is considered to have a high potential for future management of malignancies. This pilot external quality assessment (EQA) scheme aimed to address issues of analytical quality in this new area of laboratory diagnostics. METHODS The EQA scheme consisted of three 2-mL EDTA-plasma samples spiked with fragmented genomic DNA with a mutant allele frequency ranging from 0% to 10% dedicated to the analysis of nine known sequence variations in KRAS codon 12/13 and of BRAF V600E. Laboratories reported: (1) time elapsed for processing, (2) storage temperatures, (3) methods for extraction and quantification, (4) genotyping methodologies and (5) results. RESULTS Specimens were sent to 42 laboratories from 10 European countries; 72.3% reported to isolate cell-free DNA (cfDNA) manually, 62.5% used the entire plasma volume for cfDNA isolation and 38.5% used >10% of cfDNA extracted for downstream genotyping. Of the methods used for quantification, PicoGreen demonstrated the lowest coefficient of variation (33.7%). For genotyping, 11 different methods were reported with the highest error rate observed for Sanger sequencing and the lowest for highly sensitive approaches like digital PCR. In total, 197 genotypes were determined with an overall error rate of 6.09%. CONCLUSIONS This pilot EQA scheme illustrates the current variability in multiple phases of cfDNA processing and analysis of ctDNA resulting in an overall error rate of 6.09%. The areas with the greatest variance and clinical impact included specimen volume, cfDNA quantification method, and preference of genotyping platform. Regarding quality assurance, there is an urgent need for harmonisation of procedures and workflows.
Collapse
Affiliation(s)
- Verena Haselmann
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| | - Parviz Ahmad-Nejad
- Institute for Medical Laboratory Diagnostics, Centre for Clinical and Translational Research (CCTR), HELIOS Hospital, Witten/Herdecke University, Wuppertal, Germany
| | - Wolf J Geilenkeuser
- Reference-Institute for Bioanalytics, German Society for Clinical Chemistry and Laboratory Medicine (DGKL), Bonn, Germany
| | - Angelika Duda
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| | - Merle Gabor
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| | - Romy Eichner
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| | - Simon Patton
- European Molecular Genetic Quality Network (EMQN), Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester, UK
| | - Michael Neumaier
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, Mannheim, Germany
| |
Collapse
|
49
|
Kuboki Y, Fischer CG, Beleva Guthrie V, Huang W, Yu J, Chianchiano P, Hosoda W, Zhang H, Zheng L, Shao X, Thompson ED, Waters K, Poling J, He J, Weiss MJ, Wolfgang CL, Goggins MG, Hruban RH, Roberts NJ, Karchin R, Wood LD. Single-cell sequencing defines genetic heterogeneity in pancreatic cancer precursor lesions. J Pathol 2019; 247:347-356. [PMID: 30430578 DOI: 10.1002/path.5194] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/28/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022]
Abstract
Intraductal papillary mucinous neoplasms (IPMNs) are precursors to pancreatic cancer; however, little is known about genetic heterogeneity in these lesions. The objective of this study was to characterize genetic heterogeneity in IPMNs at the single-cell level. We isolated single cells from fresh tissue from ten IPMNs, followed by whole genome amplification and targeted next-generation sequencing of pancreatic driver genes. We then determined single-cell genotypes using a novel multi-sample mutation calling algorithm. Our analyses revealed that different mutations in the same driver gene frequently occur in the same IPMN. Two IPMNs had multiple mutations in the initiating driver gene KRAS that occurred in unique tumor clones, suggesting the possibility of polyclonal origin or an unidentified initiating event preceding this critical mutation. Multiple mutations in later-occurring driver genes were also common and were frequently localized to unique tumor clones, raising the possibility of convergent evolution of these genetic events in pancreatic tumorigenesis. Single-cell sequencing of IPMNs demonstrated genetic heterogeneity with respect to early and late occurring driver gene mutations, suggesting a more complex pattern of tumor evolution than previously appreciated in these lesions. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yuko Kuboki
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Catherine G Fischer
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Violeta Beleva Guthrie
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Wenjie Huang
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Yu
- Department of Surgery, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter Chianchiano
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Waki Hosoda
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Lily Zheng
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoshan Shao
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth D Thompson
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin Waters
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Justin Poling
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin He
- Department of Surgery, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew J Weiss
- Department of Surgery, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher L Wolfgang
- Department of Surgery, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas J Roberts
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachel Karchin
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Mair R, Mouliere F, Smith CG, Chandrananda D, Gale D, Marass F, Tsui DWY, Massie CE, Wright AJ, Watts C, Rosenfeld N, Brindle KM. Measurement of Plasma Cell-Free Mitochondrial Tumor DNA Improves Detection of Glioblastoma in Patient-Derived Orthotopic Xenograft Models. Cancer Res 2019; 79:220-230. [PMID: 30389699 PMCID: PMC6753020 DOI: 10.1158/0008-5472.can-18-0074] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/29/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022]
Abstract
The factors responsible for the low detection rate of cell-free tumor DNA (ctDNA) in the plasma of patients with glioblastoma (GBM) are currently unknown. In this study, we measured circulating nucleic acids in patient-derived orthotopically implanted xenograft (PDOX) models of GBM (n = 64) and show that tumor size and cell proliferation, but not the integrity of the blood-brain barrier or cell death, affect the release of ctDNA in treatment-naïve GBM PDOX. Analysis of fragment length profiles by shallow genome-wide sequencing (<0.2× coverage) of host (rat) and tumor (human) circulating DNA identified a peak at 145 bp in the human DNA fragments, indicating a difference in the origin or processing of the ctDNA. The concentration of ctDNA correlated with cell death only after treatment with temozolomide and radiotherapy. Digital PCR detection of plasma tumor mitochondrial DNA (tmtDNA), an alternative to detection of nuclear ctDNA, improved plasma DNA detection rate (82% vs. 24%) and allowed detection in cerebrospinal fluid and urine. Mitochondrial mutations are prevalent across all cancers and can be detected with high sensitivity, at low cost, and without prior knowledge of tumor mutations via capture-panel sequencing. Coupled with the observation that mitochondrial copy number increases in glioma, these data suggest analyzing tmtDNA as a more sensitive method to detect and monitor tumor burden in cancer, specifically in GBM, where current methods have largely failed. SIGNIFICANCE: These findings show that detection of tumor mitochondrial DNA is more sensitive than circulating tumor DNA analysis to detect and monitor tumor burden in patient-derived orthotopic xenografts of glioblastoma.
Collapse
Affiliation(s)
- Richard Mair
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Florent Mouliere
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Christopher G Smith
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Dineika Chandrananda
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Davina Gale
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Francesco Marass
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Dana W Y Tsui
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles E Massie
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Alan J Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Colin Watts
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom.
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom.
- Cancer Research UK Major Centre - Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|