1
|
LeJeune C, Mora-Soto N, Storgaard L, Pareja R, Amant F. Cervical cancer in pregnancy. Semin Perinatol 2025; 49:152038. [PMID: 40089318 DOI: 10.1016/j.semperi.2025.152038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Cervical cancer is one of the most common and challenging malignancies diagnosed during pregnancy, requiring a complex balance between effective oncological management and pregnancy preservation. This narrative review synthesizes current evidence and clinical experience regarding the management of cervical cancer in pregnant patients. While treatment should generally follow standard protocols for nonpregnant patients, emerging data suggest that pregnancy can often be safely maintained without compromising oncological outcomes. Nevertheless, robust prognostic data from larger patient cohorts are still needed. Individualized treatment approaches that incorporate the patient's preferences and values are crucial. Given the complexities involved, referral to specialized tertiary centers with expertise in the oncological and obstetric care of pregnant cancer patients is strongly recommended to ensure optimal outcomes for both mother and child.
Collapse
Affiliation(s)
- Charlotte LeJeune
- Lab of Gynecological Oncology, Department of Oncology, KU Leuven, Leuven, Belgium; Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium
| | - Nathalia Mora-Soto
- Department of Gynecologic Oncology, Instituto Nacional de Cancerología, Bogotá, Colombia
| | - Lone Storgaard
- Department of Gynecology and Obstetrics, Julian Marie center, Rigshospitalet, Copenhagen, Denmark
| | - René Pareja
- Department of Gynecologic Oncology, Instituto Nacional de Cancerología, Bogotá, Colombia; Gynecologic Oncology, Clínica Astorga, Medellín, Colombia
| | - Frédéric Amant
- Lab of Gynecological Oncology, Department of Oncology, KU Leuven, Leuven, Belgium; Department of Obstetrics and Gynecology, UZ Leuven, Leuven, Belgium; Department of Gynecologic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Goldlust IS, Bianchi DW. Incidental Detection of Maternal Cancer Following Cell-Free DNA Screening for Fetal Aneuploidies. Clin Chem 2025; 71:61-68. [PMID: 39749496 DOI: 10.1093/clinchem/hvae170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/05/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Prenatal cell-free DNA (cfDNA) screening is a success story of clinical genomics that has translated to and transformed obstetric care. It is a highly sensitive and specific method of screening for the most common fetal aneuploidies, including trisomies 13, 18, and 21. While primarily designed to detect fetal chromosomal abnormalities, the test also analyzes maternal cfDNA, which can complicate interpretation of results. Occasionally, abnormalities in cfDNA that do not align with fetal aneuploidy may indicate benign or malignant maternal disease states, presenting unique diagnostic challenges and opportunities. CONTENT This review explores the methods and implications of incidental cancer detection through prenatal cfDNA screening. Early case reports noted instances in which abnormal cfDNA results, initially suggestive of fetal aneuploidies, led to post-pregnancy diagnoses of maternal cancers. Subsequent large retrospective studies have established significant correlations between atypical prenatal cfDNA results and undiagnosed maternal malignancies. Abnormal cfDNA profiles, particularly those indicating multiple aneuploidies, first require fetal diagnostic tests, such as amniocentesis. If the fetal studies are normal, further investigation into a maternal source should proceed. Clinical management of these findings may involve a multidisciplinary approach, incorporating advanced imaging techniques and genetic counseling to ensure timely and accurate maternal diagnoses. SUMMARY The integration of genome-wide analysis and innovative bioinformatics tools into prenatal cfDNA screening has enhanced its ability to identify potential cancer cases. Developing standardized guidelines for reporting and managing incidental findings is crucial to optimizing patient outcomes and mitigating psychological impacts on expectant persons and their partners.
Collapse
Affiliation(s)
- Ian S Goldlust
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Diana W Bianchi
- Prenatal Genomics and Therapy Section, Center for Precision Health Research, National Human Genome Institute, National Institutes of Health, Bethesda, MD, United States
- Office of the Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
3
|
Turriff AE, Annunziata CM, Malayeri AA, Redd B, Pavelova M, Goldlust IS, Rajagopal PS, Lin J, Bianchi DW. Prenatal cfDNA Sequencing and Incidental Detection of Maternal Cancer. N Engl J Med 2024; 391:2123-2132. [PMID: 39774314 PMCID: PMC11711700 DOI: 10.1056/nejmoa2401029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND Cell-free DNA (cfDNA) sequence analysis to screen for fetal aneuploidy can incidentally detect maternal cancer. Additional data are needed to identify DNA-sequencing patterns and other biomarkers that can identify pregnant persons who are most likely to have cancer and to determine the best approach for follow-up. METHODS In this ongoing study we performed cancer screening in pregnant or postpartum persons who did not perceive signs or symptoms of cancer but received unusual clinical cfDNA-sequencing results or results that were nonreportable (i.e., the fetal aneuploidy status could not be assessed) from one of 12 different commercial laboratories in North America. We used a uniform cancer-screening protocol including rapid whole-body magnetic resonance imaging (MRI), laboratory tests, and standardized cfDNA sequencing for research purposes with the use of a genomewide platform. The primary outcome was the presence of cancer in participants after the initial cancer-screening evaluation. Secondary analyses included test performance. RESULTS Cancer was present in 52 of the 107 participants in the initial cohort (48.6%). The sensitivity and specificity of whole-body MRI in detecting occult cancer were 98.0% and 88.5%, respectively. Physical examination and laboratory tests were of limited use in identifying participants with cancer. Research sequencing showed that 49 participants had a combination of copy-number gains and losses across multiple (≥3) chromosomes; cancer was present in 47 of the participants (95.9%) with this sequencing pattern. Sequencing patterns of cfDNA in which there were only chromosomal gains (multiple trisomies) or only chromosomal losses (one or more monosomies) were found in participants with nonmalignant conditions, such as fibroids. CONCLUSIONS In this study, 48.6% of participants who received unusual or nonreportable clinical cfDNA-sequencing results had an occult cancer. Further study of DNA-sequencing patterns that are suggestive of occult cancer during prenatal screening is warranted. (Funded by the NIH Intramural Research Programs; ClinicalTrials.gov number, NCT04049604.).
Collapse
Affiliation(s)
- Amy E. Turriff
- Prenatal Genomics & Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Christina M. Annunziata
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ashkan A. Malayeri
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Bernadette Redd
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD
| | | | - Ian S. Goldlust
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Padma Sheila Rajagopal
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jielu Lin
- Section on Social Network Methods, Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Diana W. Bianchi
- Prenatal Genomics & Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
- Office of the Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
4
|
Heesterbeek CJ, Tjan-Heijnen VC, Heimovaara JH, Lenaerts L, Lok C, Vriens IJ, Van Opstal D, Boon EM, Sie D, de Die-Smulders CE, Amant F, Macville MV. Prenatal cell-free DNA testing of women with pregnancy-associated cancer: a retrospective cross-sectional study. THE LANCET REGIONAL HEALTH. EUROPE 2024; 45:101024. [PMID: 39220433 PMCID: PMC11363838 DOI: 10.1016/j.lanepe.2024.101024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Background Incidentally, the non-invasive prenatal test (NIPT) shows chromosomal aberrations suspicious of a maternal malignancy, especially after genome-wide testing. The aim of this study is to determine how many cases of cancer in pregnancy are diagnosed or missed with NIPT and whether in retrospect subtle changes in NIPT results could have detected cancer. Methods We identified Dutch patients diagnosed in 2017-2021 with pregnancy-associated cancer from the International Network on Cancer, Infertility and Pregnancy (INCIP) Registry, who underwent NIPT in the Dutch NIPT implementation study (TRIDENT-2). We retrospectively assessed how many of these women showed a malignancy suspicious-NIPT, their tumour types and -stages, and the time interval between NIPT and cancer diagnosis. Findings Of 143 women with pregnancy-associated cancer, we included 65 patients that underwent an NIPT. Fifty-four women had a solid tumour and 11 a haematological malignancy. Sixteen (24.6%) NIPTs were malignancy suspicious (15 genome-wide, one targeted). All 10 haematological cancer patients with genome-wide NIPT had a malignancy suspicious-NIPT, irrespective of the disease stage. Only five patients with a solid tumour had a genome-wide malignancy suspicious-NIPT (4/5 advanced cancer stage III or IV). The mean time between date of NIPT and cancer diagnosis was significantly shorter after a malignancy suspicious-NIPT compared to a non-suspicious-NIPT, respectively 49.9 days (± SD 31.8) and 100.7 days (± SD 74.9), p = 0.001. Interpretation All genome-wide NIPT in women with pregnancy-associated haematological malignancies were malignancy suspicious. Women with a solid tumour showed a malignancy suspicious-NIPT in only a minority of cases, mainly the advanced stages. Funding None.
Collapse
Affiliation(s)
- Catharina J. Heesterbeek
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Vivianne C.G. Tjan-Heijnen
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Joosje H. Heimovaara
- Department of Gynaecology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Oncology, Catholic University Leuven, Leuven, Belgium
| | | | - Christianne Lok
- Department of Gynaecologic Oncology, Centre of Gynaecologic Oncology Amsterdam, Amsterdam, the Netherlands
| | - Ingeborg J.H. Vriens
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Diane Van Opstal
- Department of Clinical Genetics, Erasmus MC University Medical Centre, Rotterdam, the Netherlands
| | - Elles M.J. Boon
- Department of Human Genetics, VU University Medical Centre Amsterdam, Amsterdam University Medical Centres, Amsterdam, the Netherlands
| | - Daoud Sie
- Department of Human Genetics, VU University Medical Centre Amsterdam, Amsterdam University Medical Centres, Amsterdam, the Netherlands
| | | | - Frédéric Amant
- Department of Gynaecologic Oncology, Catholic University Leuven, Leuven, Belgium
- Department of Gynaecologic Oncology, Antoni van Leeuwenhoek-The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Merryn V.E. Macville
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
5
|
Turriff AE, Bianchi DW. Oncologists must act to manage cancer detected through prenatal screening. Nat Rev Cancer 2024; 24:649-650. [PMID: 38951713 DOI: 10.1038/s41568-024-00719-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Affiliation(s)
- Amy E Turriff
- Prenatal Genomics & Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Diana W Bianchi
- Prenatal Genomics & Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD, USA.
- Office of the Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA.
| |
Collapse
|
6
|
Baruah S, Rajak J, Mitra AK, Dhara B. Advancements in precision oncology: Investigating the function of circulating DNA in the advancement of liquid biopsy technologies. THE JOURNAL OF LIQUID BIOPSY 2024; 5:100157. [PMID: 40027946 PMCID: PMC11863923 DOI: 10.1016/j.jlb.2024.100157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 03/05/2025]
Abstract
While the traditional method is a biopsy of the potential tumor tissue, more non-invasive methods have been developed such as the liquid biopsy. It is an emerging, powerful cancer detection method that is used for molecular profiling of tumours apart from tissue-specific biopsy. It involves the analysis of free, circulating tumor genetic material and circulating tumor cells (CTCs) that are found in the fluids of the patient. This analysis helps to detect the abnormal genetic anomalies associated with the various forms of cancer as well as identify mutations corresponding to specific tumours. It is an upcoming method of diagnostics applied in the field of precision oncology that helps overcome the limitations of the traditional biopsy. It can interrogate and look through difficult-to-biopsy tumours and also aid in early cancer detection. Sequencing techniques help in building databases with different information on mutations in the genetic as well as epigenetic levels. In this review, we understand the role of liquid biopsy as not only a diagnostic tool but its role in therapeutics and its potential in the future.
Collapse
Affiliation(s)
- Stootee Baruah
- Department of Microbiology, St. Xavier's College (Autonomous), Kolkata, 700016, India
| | - Jenifer Rajak
- Department of Microbiology, St. Xavier's College (Autonomous), Kolkata, 700016, India
| | - Arup Kumar Mitra
- Department of Microbiology, St. Xavier's College (Autonomous), Kolkata, 700016, India
| | - Bikram Dhara
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- Department of Health Sciences, Novel Global Community and Educational Foundation, Hebersham, NSW, Australia
| |
Collapse
|
7
|
Merrill MH, Cahill SR, Pepprock HW, Redd R, Rana HQ, Economy KE, Garber JE, LaCasce AS. Detection of Maternal Malignancy After Abnormal Noninvasive Prenatal Testing: A Single-Center Case Series. JCO Precis Oncol 2024; 8:e2400058. [PMID: 39298712 DOI: 10.1200/po.24.00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/13/2024] [Accepted: 08/15/2024] [Indexed: 09/22/2024] Open
|
8
|
Heesterbeek CJ, Lenaerts L, Tjan-Heijnen VCG, Amant F, van Rij MC, Theunis M, de Die-Smulders CEM, Vermeesch JR, Macville MVE. Comprehensive Recommendations for the Clinical Management of Pregnant Women With Noninvasive Prenatal Test Results Suspicious of a Maternal Malignancy. JCO Oncol Pract 2024; 20:1027-1034. [PMID: 38608208 DOI: 10.1200/op.23.00594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/12/2024] [Accepted: 02/29/2024] [Indexed: 04/14/2024] Open
Abstract
In this article, we defined comprehensive recommendations for the clinical follow-up of pregnant women with a malignancy-suspicious NIPT result, on the basis of the vast experience with population-based NIPT screening programs in two European countries complemented with published large data sets. These recommendations provide a tool for classifying NIPT results as malignancy-suspicious, and guide health care professionals in structured clinical decision making for the diagnostic process of pregnant women who receive such a malignancy-suspicious NIPT result.
Collapse
Affiliation(s)
- Catharina J Heesterbeek
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - Vivianne C G Tjan-Heijnen
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Frédéric Amant
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Gynecologic Oncology, Catholic University Leuven, Leuven, Belgium
- Department of Gynecologic Oncology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maartje C van Rij
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Miel Theunis
- Centre of Human Genetics, Catholic University Leuven, Leuven, Belgium
| | - Christine E M de Die-Smulders
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joris R Vermeesch
- Centre of Human Genetics, Catholic University Leuven, Leuven, Belgium
| | - Merryn V E Macville
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
9
|
Rosenblum J, Blaumeiser B, Janssens K. The impact of confined placental mosaicism on prenatal cell-free DNA screening: Insights from a monocentric study of 99 cases. Placenta 2024; 152:17-22. [PMID: 38744036 DOI: 10.1016/j.placenta.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/01/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Confined placental mosaicism (CPM) is thought to be one of the main sources of false-positive prenatal cell-free DNA (cfDNA) screening results, but extensive and systematic studies to prove this statement are limited. We evaluate the contribution of CPM to false-positive prenatal cfDNA screening results in the largest cohort published to date. METHOD We systematically offered postnatal analysis on placenta and umbilical cord to women who had a negative amniocentesis following a positive prenatal cfDNA screening result. A standardized protocol was used in which (when available) biopsies were taken at five locations in the placenta and umbilical cord. RESULTS We analyzed a series of 99 placentas. CPM could be confirmed in 32.3 % of cases (32/99). CPM was detected across all subtypes of chromosomal aberrations (common and rare autosomal trisomies, sex chromosome abnormalities, copy number variations and autosomal monosomies). A lower detection rate was present in umbilical cord biopsies in comparison with placental biopsies. When comparing different sections of the placenta, no clear difference could be observed with regard to the probability of CPM being present nor to the grade of mosaicism. DISCUSSION We confirm an important role for CPM in explaining false-positive prenatal cfDNA screening results. Placental regional differences are common. Given its limited clinical relevance, we do however not advocate placental studies in a diagnostic setting.
Collapse
Affiliation(s)
- Jessica Rosenblum
- Department of Medical Genetics, Antwerp University and University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium.
| | - Bettina Blaumeiser
- Department of Medical Genetics and Department of Obstetrics and Gynecology, Antwerp University and University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium.
| | - Katrien Janssens
- Department of Medical Genetics, Antwerp University and University Hospital, Drie Eikenstraat 655, 2650, Edegem, Belgium.
| |
Collapse
|
10
|
Abstract
This review delves into the rapidly evolving landscape of liquid biopsy technologies based on cell-free DNA (cfDNA) and cell-free RNA (cfRNA) and their increasingly prominent role in precision medicine. With the advent of high-throughput DNA sequencing, the use of cfDNA and cfRNA has revolutionized noninvasive clinical testing. Here, we explore the physical characteristics of cfDNA and cfRNA, present an overview of the essential engineering tools used by the field, and highlight clinical applications, including noninvasive prenatal testing, cancer testing, organ transplantation surveillance, and infectious disease testing. Finally, we discuss emerging technologies and the broadening scope of liquid biopsies to new areas of diagnostic medicine.
Collapse
Affiliation(s)
- Conor Loy
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Lauren Ahmann
- Department of Pathology, Stanford University, Stanford, California, USA;
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Wei Gu
- Department of Pathology, Stanford University, Stanford, California, USA;
| |
Collapse
|
11
|
Boukovala M, Westphalen CB, Probst V. Liquid biopsy into the clinics: Current evidence and future perspectives. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100146. [PMID: 40027149 PMCID: PMC11863819 DOI: 10.1016/j.jlb.2024.100146] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2025]
Abstract
As precision oncology has become a major part of the treatment landscape in oncology, liquid biopsies have developed as a particularly powerful tool as it surmounts several limitations of traditional tissue biopsies. These biopsies involve most commonly the isolation of circulating extracellular nucleic acids, including cell-free DNA (cfDNA) and circulating tumor DNA (ctDNA), as well as circulating tumor cells (CTCs), typically from blood. The clinical applications of liquid biopsies are diverse, encompassing the initial diagnosis and cancer detection, the application as a tool for prognostication in early and advanced tumor settings, the identification of potentially actionable alterations, the monitoring of response and resistance under systemic therapy and the detection of resistance mechanisms, the differentiation of distinct immune checkpoint blockade response patterns through serial samples, the prediction of immune checkpoint blockade responses based on initial liquid biopsy characteristics and the assessment of tumor heterogeneity. Moreover, molecular relapse monitoring in early-stage cancers and the personalization of adjuvant or additive therapy via MRD have become a major field of research in recent years. Compared to tissue biopsies, liquid biopsies are less invasive and can be collected serially, offering real-time molecular insights. Furthermore, liquid biopsies may allow for a more holistic evaluation of a patient's disease, as they assess material from all tumor sites and can theoretically reflect tumor heterogeneity. Furthermore, quicker turnaround-time also constitutes an advantage of liquid biopsies. Disadvantages or hurdles include the challenge of detecting low amounts of tumor deposits in peripheral blood or other fluids and the potential of different amounts tumor-shedding from different metastatic sites, as well as potentially false-positive from clonal hematopoietic mutations of indeterminate potential (CHIP) mutations. The clinical utility of liquid biopsies still must be validated in most settings and further research has to be done. Clinal trials including alternate bodily fluids and leveraging AI-technology are expected to revolutionize the field of liquid biopsies.
Collapse
|
12
|
Chen CP. False positive non-invasive prenatal testing (NIPT) for partial deletion of chromosome 7q in a pregnancy associated with maternal uterine fibroids and a normal maternal blood cytogenetic result. Taiwan J Obstet Gynecol 2024; 63:279-280. [PMID: 38485334 DOI: 10.1016/j.tjog.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 03/19/2024] Open
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Institute of Clinical and Community Health Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
13
|
Chang Y, Li S, Li Z, Wang X, Chang F, Geng S, Zhu D, Zhong G, Wu W, Chang Y, Tu S, Mao M. Non-invasive detection of lymphoma with circulating tumor DNA features and protein tumor markers. Front Oncol 2024; 14:1341997. [PMID: 38313801 PMCID: PMC10834776 DOI: 10.3389/fonc.2024.1341997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
Background According to GLOBOCAN 2020, lymphoma ranked as the 9th most common cancer and the 12th leading cause of cancer-related deaths worldwide. Traditional diagnostic methods rely on the invasive excisional lymph node biopsy, which is an invasive approach with some limitations. Most lymphoma patients are diagnosed at an advanced stage since they are asymptomatic at the beginning, which has significantly impacted treatment efficacy and prognosis of the disease. Method This study assessed the performance and utility of a newly developed blood-based assay (SeekInCare) for lymphoma early detection. SeekInCare utilized protein tumor markers and a comprehensive set of cancer-associated genomic features, including copy number aberration (CNA), fragment size (FS), end motif, and lymphoma-related virus, which were profiled by shallow WGS of cfDNA. Results Protein marker CA125 could be used for lymphoma detection independent of gender, and the sensitivity was 27.8% at specificity of 98.0%. After integrating these multi-dimensional features, 77.8% sensitivity was achieved at specificity of 98.0%, while its NPV and PPV were both more than 92% for lymphoma detection. The sensitivity of early-stage (I-II) lymphoma was up to 51.3% (47.4% and 55.0% for stage I and II respectively). After 2 cycles of treatment, the molecular response of SeekInCare was correlated with the clinical outcome. Conclusion In summary, a blood-based assay can be an alternative to detect lymphoma with adequate performance. This approach becomes particularly valuable in cases where obtaining tissue biopsy is difficult to obtain or inconclusive.
Collapse
Affiliation(s)
- Yu Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shiyong Li
- Research and Development, SeekIn Inc, Shenzhen, China
| | - Zhiming Li
- Department of Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | - Dandan Zhu
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Guolin Zhong
- Research and Development, SeekIn Inc, Shenzhen, China
| | - Wei Wu
- Research and Development, SeekIn Inc, Shenzhen, China
| | - Yinyin Chang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Shichun Tu
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Mao Mao
- Research and Development, SeekIn Inc, Shenzhen, China
- Yonsei Song-Dang Institute for Cancer Research, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
14
|
Lenaerts L, Theunis M, Amant F, Vermeesch JR. Non-invasive prenatal testing: when results suggests maternal cancer. MED GENET-BERLIN 2023; 35:285-295. [PMID: 38835737 PMCID: PMC11006267 DOI: 10.1515/medgen-2023-2055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
It is now well-established that non-invasive prenatal testing (NIPT), originally designed to screen cell-free DNA (cfDNA) in maternal blood for the presence of common fetal trisomies, can lead to incidental detection of occult maternal malignancies. Retrospective evaluations have demonstrated that the detection of multiple copy number alterations in cfDNA is particularly suggestive of an incipient tumor and that cancer detection rates not only depend on tumor biology but also on applied NIPT technologies and downstream diagnostic investigations. Since the identification of a maternal cancer in pregnancy has implications for both woman and the unborn child, prospective studies are needed to provide evidence on best clinical practices and on clinical utility in terms of patient outcomes.
Collapse
Affiliation(s)
- Liesbeth Lenaerts
- Catholic University Leuven Department of Oncology, Laboratory of Gynecological Oncology Herestraat 49 - box 818 3000 Leuven Belgium
| | - Miel Theunis
- Catholic University Leuven Centre of Human Genetics Herestraat 49 - box 818 3000 Leuven Belgium
| | - Frédéric Amant
- Catholic University Leuven Department of Oncology, Laboratory of Gynecological Oncology Herestraat 49 - box 818 3000 Leuven Belgium
- Catholic University Leuven Department of Oncology, Laboratory for Gynecological Oncology Leuven Belgium
- University Hospitals Leuven Centre of Human Genetics Leuven Belgium
| | - Joris R Vermeesch
- Catholic University Leuven Centre of Human Genetics Herestraat 49 - box 818 3000 Leuven Belgium
| |
Collapse
|
15
|
Creswell L, Doddy F, Manning C, Nazir SF, Lindow SW, Lynch C, O'Gorman N. Cell free DNA screening for fetal aneuploidy in Ireland: An observational study of outcomes following insufficient fetal fraction. Eur J Obstet Gynecol Reprod Biol 2023; 290:143-149. [PMID: 37797414 DOI: 10.1016/j.ejogrb.2023.09.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVES To determine maternal factors associated with low fetal fraction (FF). To determine the proportion of women who receive a result from repeat non-invasive prenatal testing (NIPT) testing. To identify any significant associations between pregnancy interventions or outcomes and low FF. STUDY DESIGN Retrospective observational study of 4465 women undergoing antenatal screening by targeted cell free DNA (cfDNA) testing at an Irish tertiary maternity hospital between January 2017 and December 2022. Patients who failed to obtain a result after the first NIPT were analyzed in two cohorts; those who received a result on a repeat sample and those who failed to ever achieve a result despite a second, third or fourth cfDNA test. RESULTS Risk of insufficient FF significantly increased with elevated maternal BMI (OR 1.07; 95% CI 1.01-1.13, p = 0.03) and in-vitro fertilization (IVF) (OR 3.4; 95% CI 1.19-9.4, p = 0.02). Women with no result were more likely to have diagnostic invasive testing (p < 0.01), but had no increased risk of aneuploidy. Repeated failed NIPT attempts due to low FF were significantly associated with the subsequent development of hypertensive diseases of pregnancy (p = 0.03). Greater than 70% of patients who were unsuccessful in a first or second attempt at NIPT due to low FF yielded a result following a second or third sample. CONCLUSIONS High BMI and IVF conceptions are greater contributors to low FF than fetal aneuploidy. Repeating NIPT yields a result in greater than 70% of cases. WHAT'S ALREADY KNOWN ABOUT THIS TOPIC?: Fetal fraction (FF) in prenatal cfDNA testing is influenced by maternal and pregnancy factors including body mass index (BMI) and IVF. Low FF has been associated with adverse pregnancy outcomes including fetal aneuploidy and hypertensive diseases of pregnancy. WHAT DOES THIS STUDY ADD?: In a large Irish population, increasing maternal BMI and in-vitro fertilization are the most significant contributors to repeated test failures due to low FF. Greater than 70% of patients with test failure due to low FF will receive a result on 2nd and 3rd NIPT attempts. Patients with no result from NIPT were more likely to undergo diagnostic invasive testing but the risk of aneuploidy was not significantly increased.
Collapse
Affiliation(s)
| | - F Doddy
- The Coombe Hospital, Dublin, Ireland
| | - C Manning
- The Coombe Hospital, Dublin, Ireland
| | - S F Nazir
- The Coombe Hospital, Dublin, Ireland
| | | | - C Lynch
- The Coombe Hospital, Dublin, Ireland
| | | |
Collapse
|
16
|
Rink BD, Stevens BK. Incidental Detection of Malignancies With Cell-Free DNA Screening. Clin Obstet Gynecol 2023; 66:607-615. [PMID: 37443422 DOI: 10.1097/grf.0000000000000800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023]
Abstract
Cell-free circulating DNA is an evolving technology with important clinical applications in both obstetric care and oncology. In the challenging patient with pregnancy and co-existing malignancy, the utility of cell-free DNA both for aneuploidy screening and cancer identification is an area of active research. Understanding the physiology associated with circulating cell-free DNA and subsequent laboratory evaluation is critical for clinicians caring for the obstetric patient with cell-free fetal DNA screening results suggestive of malignancy. Ongoing research is necessary to determine best practices for the evaluation and management of these patients with promising applications in the advancement of precision medicine.
Collapse
Affiliation(s)
- Britton D Rink
- Mount Carmel Health Systems, Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Blair K Stevens
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Texas McGovern Medical School, Houston, Texas
| |
Collapse
|
17
|
Benn P, Cuckle H. Overview of Noninvasive Prenatal Testing (NIPT) for the Detection of Fetal Chromosome Abnormalities; Differences in Laboratory Methods and Scope of Testing. Clin Obstet Gynecol 2023; 66:536-556. [PMID: 37650667 DOI: 10.1097/grf.0000000000000803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Although nearly all noninvasive prenatal testing is currently based on analyzing circulating maternal cell-free DNA, the technical methods usedvary considerably. We review the different methods. Based on validation trials and clinical experience, there are mostly relatively small differences in screening performance for trisomies 21, 18, and 13 in singleton pregnancies. Recent reports show low no-call rates for all methods, diminishing its importance when choosing a laboratory. However, method can be an important consideration for twin pregnancies, screening for sex chromosome abnormalities, microdeletion syndromes, triploidy, molar pregnancies, rare autosomal trisomies, and segmental imbalances, and detecting maternal chromosome abnormalities.
Collapse
Affiliation(s)
- Peter Benn
- Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, Connecticut
| | - Howard Cuckle
- Department of Obstetrics and Gynecology, Faculty of Medicine, Tel Aviv University, Israel
| |
Collapse
|
18
|
Moufarrej MN, Bianchi DW, Shaw GM, Stevenson DK, Quake SR. Noninvasive Prenatal Testing Using Circulating DNA and RNA: Advances, Challenges, and Possibilities. Annu Rev Biomed Data Sci 2023; 6:397-418. [PMID: 37196360 PMCID: PMC10528197 DOI: 10.1146/annurev-biodatasci-020722-094144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Prenatal screening using sequencing of circulating cell-free DNA has transformed obstetric care over the past decade and significantly reduced the number of invasive diagnostic procedures like amniocentesis for genetic disorders. Nonetheless, emergency care remains the only option for complications like preeclampsia and preterm birth, two of the most prevalent obstetrical syndromes. Advances in noninvasive prenatal testing expand the scope of precision medicine in obstetric care. In this review, we discuss advances, challenges, and possibilities toward the goal of providing proactive, personalized prenatal care. The highlighted advances focus mainly on cell-free nucleic acids; however, we also review research that uses signals from metabolomics, proteomics, intact cells, and the microbiome. We discuss ethical challenges in providing care. Finally, we look to future possibilities, including redefining disease taxonomy and moving from biomarker correlation to biological causation.
Collapse
Affiliation(s)
| | - Diana W Bianchi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development and Section on Prenatal Genomics and Fetal Therapy, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gary M Shaw
- Department of Pediatrics and March of Dimes Prematurity Research Center at Stanford University, Stanford University School of Medicine, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics and March of Dimes Prematurity Research Center at Stanford University, Stanford University School of Medicine, Stanford, California, USA
| | - Stephen R Quake
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, California, USA
- Chan Zuckerberg Initiative, Redwood City, California, USA
| |
Collapse
|
19
|
Prensky L, Persson F. Apparent versus effective test performance: The importance of understanding and reviewing performance metrics. Prenat Diagn 2023; 43:1255-1258. [PMID: 37458530 DOI: 10.1002/pd.6408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
|
20
|
Klein EA, Madhavan S, Beer TM, Bettegowda C, Liu MC, Hartman AR, Hackshaw A. Dying To Find Out: The Cost of Time at the Dawn of the Multicancer Early Detection Era. Cancer Epidemiol Biomarkers Prev 2023; 32:1003-1010. [PMID: 37255363 PMCID: PMC10390858 DOI: 10.1158/1055-9965.epi-22-1275] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/21/2023] [Accepted: 05/09/2023] [Indexed: 05/17/2023] Open
Abstract
Cancer is a significant burden worldwide that adversely impacts life expectancy, quality of life, health care costs, and workforce productivity. Although currently recommended screening tests for individual cancers reduce mortality, they detect only a minority of all cancers and sacrifice specificity for high sensitivity, resulting in a high cumulative rate of false positives. Blood-based multicancer early detection tests (MCED) based on next-generation sequencing (NGS) and other technologies hold promise for broadening the number of cancer types detected in screened populations and hope for reducing cancer mortality. The promise of this new technology to improve cancer detection rates and make screening more efficient at the population level demands the development of novel trial designs that accelerate clinical adoption. Carefully designed clinical trials are needed to address these issues.
Collapse
Affiliation(s)
- Eric A. Klein
- GRAIL, Inc, and Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Tomasz M. Beer
- Exact Sciences Corporation and OHSU Knight Cancer Institute, Portland, Oregon
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | - Allan Hackshaw
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, United Kingdom
| |
Collapse
|
21
|
Jha P, Lenaerts L, Vermeesch J, Norton M, Amant F, Glanc P, Poder L. Noninvasive prenatal screening and maternal malignancy: role of imaging. Abdom Radiol (NY) 2023; 48:1590-1598. [PMID: 37095202 DOI: 10.1007/s00261-023-03913-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 04/26/2023]
Abstract
Noninvasive prenatal screening (NIPS) tests for fetal chromosomal anomalies through maternal blood sampling. It is becoming widely available and standard of care for pregnant women in many countries. It is performed in the first trimester of pregnancy, usually between 9 and 12 weeks. Fragments of fetal cell-free deoxyribonucleic acid (DNA) floating in maternal plasma are detected and analyzed by this test to assess for chromosomal aberrations. Similarly, maternal tumor-derived cell-free DNA (ctDNA) released from the tumor cells also circulates in the plasma. Hence, the presence of genomic anomalies originating from maternal tumor-derived DNA may be detected on the NIPS-based fetal risk assessment in pregnant patients. Presence of multiple aneuploidies or autosomal monosomies are the most commonly reported NIPS abnormalities detected with occult maternal malignancies. When such results are received, the search for an occult maternal malignancy begins, in which imaging plays a crucial role. The most commonly detected malignancies via NIPS are leukemia, lymphoma, breast and colon cancers. Ultrasound is a reasonable radiation-free modality for imaging during pregnancy, specially when there are localizing symptoms or findings, such as palpable lumps. While there are no consensus guidelines on the imaging evaluation for these patients, when there are no localizing symptoms or clinically palpable findings, whole body MRI is recommended as the radiation-free modality of choice to search for an occult malignancy. Based on clinical symptoms, practice patterns, and available resources, breast ultrasound, chest radiographs, and targeted ultrasound evaluations can also be performed initially or as a follow-up for MRI findings. CT is reserved for exceptional circumstances due to its higher radiation dose. This article intends to increase awareness of this rare but stressful clinical scenario and guide imaging evaluation for occult malignancy detected via NIPS during pregnancy.
Collapse
Affiliation(s)
- Priyanka Jha
- Department of Radiology, Division of Body Imaging, Stanford University School of Medicine, Palo Alto, CA, USA.
| | | | - Joris Vermeesch
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Centre of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Mary Norton
- Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA
| | - 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, Amsterdam, The Netherlands
- The Netherlands Cancer Institute, Antoni Van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Phyllis Glanc
- Department of Radiology, Obstetrics & Gynecology, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Liina Poder
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
22
|
Desarrollo genital prenatal. Clasificación, diagnóstico y manejo de las anomalías de la diferenciación sexual. CLINICA E INVESTIGACION EN GINECOLOGIA Y OBSTETRICIA 2023. [DOI: 10.1016/j.gine.2022.100828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
23
|
A View on Uterine Leiomyoma Genesis through the Prism of Genetic, Epigenetic and Cellular Heterogeneity. Int J Mol Sci 2023; 24:ijms24065752. [PMID: 36982825 PMCID: PMC10056617 DOI: 10.3390/ijms24065752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Uterine leiomyomas (ULs), frequent benign tumours of the female reproductive tract, are associated with a range of symptoms and significant morbidity. Despite extensive research, there is no consensus on essential points of UL initiation and development. The main reason for this is a pronounced inter- and intratumoral heterogeneity resulting from diverse and complicated mechanisms underlying UL pathobiology. In this review, we comprehensively analyse risk and protective factors for UL development, UL cellular composition, hormonal and paracrine signalling, epigenetic regulation and genetic abnormalities. We conclude the need to carefully update the concept of UL genesis in light of the current data. Staying within the framework of the existing hypotheses, we introduce a possible timeline for UL development and the associated key events—from potential prerequisites to the beginning of UL formation and the onset of driver and passenger changes.
Collapse
|
24
|
Goldring G, Trotter C, Meltzer JT, Souter V, Pais L, DiNonno W, Xu W, Weitzel JN, Vora NL. Maternal Malignancy After Atypical Findings on Single-Nucleotide Polymorphism-Based Prenatal Cell-Free DNA Screening. Obstet Gynecol 2023; 141:791-800. [PMID: 36897127 PMCID: PMC10026947 DOI: 10.1097/aog.0000000000005107] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/15/2022] [Indexed: 03/11/2023]
Abstract
OBJECTIVE To evaluate the incidence and clinical outcomes of cell-free DNA results suspicious for maternal malignancy on prenatal cell-free DNA screening with single-nucleotide polymorphism (SNP)-based technology. METHODS This retrospective cohort study included data from SNP-based, noninvasive prenatal screening samples from a commercial laboratory from January 2015 to October 2021. Maternal plasma was screened for trisomy 21, 18, and 13; monosomy X; and triploidy. Cases were considered suspicious for maternal malignancy if retrospective bioinformatics and visual inspection of the SNP plot were suggestive of multiple maternal copy number variants across at least two of the tested chromosomes. Clinical follow-up on patients was obtained by contacting individual referring clinician offices by telephone, facsimile, or email. RESULTS A total of 2,004,428 noninvasive prenatal screening samples during the study period met criteria for inclusion in the analysis. Of these, 38 samples (0.002% or 1 in 52,748, 95% CI 1:74,539-1:38,430) had SNP-plot results that were suspicious for maternal malignancy. Maternal health outcomes were obtained in 30 of these patients (78.9%); eight were lost to follow-up. Maternal malignancy or suspected malignancy was identified in 66.7% (20/30) of the 30 patients with clinical follow-up provided by the clinic. The most common maternal malignancies were lymphoma (n=10), breast cancer (n=5), and colon cancer (n=3). CONCLUSION Results suspicious for maternal malignancy are rare with SNP-based noninvasive prenatal screening (1:53,000), but two thirds of patients who had a noninvasive prenatal screening result concerning for malignancy in this study had a cancer diagnosis. Investigation for malignancy should be recommended for all pregnant patients with this type of result. FUNDING SOURCE This study was funded by Natera, Inc.
Collapse
Affiliation(s)
- Georgina Goldring
- Natera, Inc., Austin, Texas; and the Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Small Cell Carcinoma of the Vagina: First Systematic Review of Case Reports and Proposal of a Management Algorithm. J Low Genit Tract Dis 2023; 27:56-67. [PMID: 36282979 PMCID: PMC9770126 DOI: 10.1097/lgt.0000000000000712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Small cell carcinoma of the vagina (SmCCV) is an extremely rare disease. Evidence-based data and specific guidelines are lacking. We conducted the first systematic review of case reports to provide the most overall picture of SmCCV. MATERIALS AND METHODS Literature search in PubMed and Scopus was performed using the terms "small cell carcinoma" and "vagina." English-language case reports of primary SmCCV up to January 2022 were included. RESULTS Twenty-nine articles describing 44 cases met our inclusion criteria. We report a new case of our hospital. The global median overall survival (mOS) was 12.00 months (95% CI = 9.31-14.69). The mOS was not reached for stage I, and it was 12.00, 12.00, 9.00, and 8.00 months for stages II, III, IVA, and IVB, respectively (statistically significant differences between stage I and stages II, III, or IVA [log rank p = .003-.017]). Thirty-five cases received local treatments (77.8%). The mOS of patients treated with surgery ± complementary chemotherapy, radiotherapy ± complementary chemotherapy, chemoradiation ± complementary chemotherapy, and surgery + radiotherapy ± complementary chemotherapy were 11.00, 12.00, 17.00, and 29.00 months, respectively. The use of adjuvant or neoadjuvant chemotherapy (64.5%, mostly platinum + etoposide) showed longer mOS (77.00 vs 15.00 months). Four of 5 tested cases presented human papillomavirus infection, 3 of them presenting type 18. CONCLUSIONS Small cell carcinoma of the vagina shows dismal prognosis. Multimodal local management plus complementary chemotherapy seems to achieve better outcomes. Human papillomavirus could be related to the development of SmCCV. A diagnostic-therapeutic algorithm is proposed.
Collapse
|
26
|
Moellgaard MH, Lund ICB, Becher N, Skytte A, Andreasen L, Srebniak MI, Vogel I. Incidental finding of maternal malignancy in an unusual non-invasive prenatal test and a review of similar cases. Clin Case Rep 2022; 10:e6280. [PMID: 36245448 PMCID: PMC9552546 DOI: 10.1002/ccr3.6280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/07/2022] [Indexed: 11/11/2022] Open
Abstract
We present a clinical case where a complex abnormal non-invasive prenatal test (NIPT) result in a research project revealed carcinoma of the breast in the pregnant woman. Furthermore, the NIPT result did not demonstrate the same fetal chromosomal aberration as the chorion villus sample. A literature search for similar cases was performed identifying 43 unique cases, where abnormal NIPT results were related to maternal malignancy. Malignancy is a rare but important cause of complex abnormal non-invasive prenatal test (NIPT) results and should be considered when fetal karyotype and abnormal NIPT results are discordant. Furthermore, a follow-up invasive sample is essential for correct fetal diagnosis when abnormal NIPT results are found.
Collapse
Affiliation(s)
| | - Ida Charlotte Bay Lund
- Center for Fetal diagnosticsAarhus University/Aarhus University HospitalAarhusDenmark
- Department of Clinical GeneticsAarhus University/Aarhus University HospitalAarhusDenmark
| | - Naja Becher
- Department of Clinical GeneticsAarhus University/Aarhus University HospitalAarhusDenmark
| | - Anne‐Bine Skytte
- Department of Clinical GeneticsAarhus University/Aarhus University HospitalAarhusDenmark
- Cryos InternationalAarhusDenmark
| | - Lotte Andreasen
- Department of Clinical GeneticsAarhus University/Aarhus University HospitalAarhusDenmark
| | | | - Ida Vogel
- Center for Fetal diagnosticsAarhus University/Aarhus University HospitalAarhusDenmark
- Department of Clinical GeneticsAarhus University/Aarhus University HospitalAarhusDenmark
| |
Collapse
|
27
|
Nikanjam M, Kato S, Kurzrock R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol 2022; 15:131. [PMID: 36096847 PMCID: PMC9465933 DOI: 10.1186/s13045-022-01351-y] [Citation(s) in RCA: 376] [Impact Index Per Article: 125.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022] Open
Abstract
Liquid biopsies are increasingly used for cancer molecular profiling that enables a precision oncology approach. Circulating extracellular nucleic acids (cell-free DNA; cfDNA), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs) can be isolated from the blood and other body fluids. This review will focus on current technologies and clinical applications for liquid biopsies. ctDNA/cfDNA has been isolated and analyzed using many techniques, e.g., droplet digital polymerase chain reaction, beads, emulsion, amplification, and magnetics (BEAMing), tagged-amplicon deep sequencing (TAm-Seq), cancer personalized profiling by deep sequencing (CAPP-Seq), whole genome bisulfite sequencing (WGBS-Seq), whole exome sequencing (WES), and whole genome sequencing (WGS). CTCs have been isolated using biomarker-based cell capture, and positive or negative enrichment based on biophysical and other properties. ctDNA/cfDNA and CTCs are being exploited in a variety of clinical applications: differentiating unique immune checkpoint blockade response patterns using serial samples; predicting immune checkpoint blockade response based on baseline liquid biopsy characteristics; predicting response and resistance to targeted therapy and chemotherapy as well as immunotherapy, including CAR-T cells, based on serial sampling; assessing shed DNA from multiple metastatic sites; assessing potentially actionable alterations; analyzing prognosis and tumor burden, including after surgery; interrogating difficult-to biopsy tumors; and detecting cancer at early stages. The latter can be limited by the small amounts of tumor-derived components shed into the circulation; furthermore, cfDNA assessment in all cancers can be confounded by clonal hematopoeisis of indeterminate potential, especially in the elderly. CTCs can be technically more difficult to isolate that cfDNA, but permit functional assays, as well as evaluation of CTC-derived DNA, RNA and proteins, including single-cell analysis. Blood biopsies are less invasive than tissue biopsies and hence amenable to serial collection, which can provide critical molecular information in real time. In conclusion, liquid biopsy is a powerful tool, and remarkable advances in this technology have impacted multiple aspects of precision oncology, from early diagnosis to management of refractory metastatic disease. Future research may focus on fluids beyond blood, such as ascites, effusions, urine, and cerebrospinal fluid, as well as methylation patterns and elements such as exosomes.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, 1200 Garden View Road, Encinitas, CA, 92024, USA.
| | - Shumei Kato
- Division of Hematology-Oncology, University of California San Diego, La Jolla, 1200 Garden View Road, Encinitas, CA, 92024, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.,WIN Consortium, Paris, France
| |
Collapse
|
28
|
Rink BD, Stevens BK, Norton ME. Incidental Detection of Maternal Malignancy by Fetal Cell-Free DNA Screening. Obstet Gynecol 2022. [DOI: 10.109710.1097/aog.0000000000004833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
29
|
Incidental Detection of Maternal Malignancy by Fetal Cell-Free DNA Screening. Obstet Gynecol 2022; 140:121-131. [DOI: 10.1097/aog.0000000000004833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
|
30
|
Behera TR, Song JM, Ko J, Eicher D, Arbesman J, Gastman B, Farkas DH, Funchain P. Circulating Tumor DNA Testing Supports Rapid Therapeutic Decision-Making in Metastatic Melanoma: A Case Report. Front Oncol 2022; 12:846187. [PMID: 35756682 PMCID: PMC9231430 DOI: 10.3389/fonc.2022.846187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Treatment of metastatic melanoma includes the option of targeted therapy in patients with driver BRAF mutations. BRAF-MEK inhibitor drugs improve survival in the approximately 50% of patients with melanoma that harbor BRAF mutations. As BRAF mutation detection in tissue often takes days to weeks, it is not always possible or timely to obtain BRAF status in tissue using immunohistochemistry or next generation sequencing. Plasma-derived circulating tumor DNA (ctDNA) is a potential alternative analyte in such treatment settings. We present a case of metastatic melanoma that was treated in an emergent setting using therapy supported by rapid PCR-based detection of ctDNA positive for a BRAF V600 mutation. In this rapidly deteriorating 53-year-old male with diffuse melanoma metastases and unknown BRAF mutation status requiring hospital admission, a plasma-based BRAF mutation detection supported treatment with targeted therapy, dabrafenib and trametinib. Same-day initiation of therapy resulted in swift amelioration allowing discharge within a week, followed by substantial clinical improvement over the following weeks. In cases requiring urgent clinical decision making, a plasma-based, near point-of-care detection system is useful in supporting targeted therapy decisions without the need for invasive and time-consuming biopsy.
Collapse
Affiliation(s)
| | - Jung Min Song
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jennifer Ko
- Anatomic Pathology, Cleveland Clinic, Cleveland, OH, United States
| | - Donald Eicher
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Joshua Arbesman
- Dermatology & Plastic Surgery Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brian Gastman
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Daniel H Farkas
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Pauline Funchain
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
31
|
Turriff AE, Annunziata CM, Bianchi DW. Prenatal DNA Sequencing for Fetal Aneuploidy Also Detects Maternal Cancer: Importance of Timely Workup and Management in Pregnant Women. J Clin Oncol 2022; 40:2398-2401. [PMID: 35704839 PMCID: PMC9467676 DOI: 10.1200/jco.22.00733] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Amy E Turriff
- Prenatal Genomics & Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD
| | | | - Diana W Bianchi
- Prenatal Genomics & Therapy Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health (NIH), Bethesda, MD.,Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| |
Collapse
|
32
|
Di Giosaffatte N, Bottillo I, Laino L, Iaquinta G, Ferraris A, Garzia M, Bargiacchi S, Mulargia C, Angelitti MR, Palumbo F, Grammatico B, Bartolelli C, Salerno MG, Rigacci L, Grammatico P. Discordant cfDNA-NIPT result unraveling a trisomy 12 chronic lymphocytic leukemia in a 37 years old pregnant woman. Prenat Diagn 2022; 42:1000-1003. [PMID: 35506546 PMCID: PMC9546086 DOI: 10.1002/pd.6158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/07/2022]
Abstract
What's already known about this topic?
Discordant NIPT results can rarely unravel maternal malignancies, especially when multiple chromosomal imbalances are reported. Both solid and hematological neoplasms have been described.
What does this study add?
This is the first case of a discordant NIPT result due to Chronic Lymphocytic Leukemia associated with trisomy of the chromosome 12. Putative maternal malignancy should be considered and investigated through sensitive techniques even in presence of a single chromosomal anomaly. This must be considered especially when the imbalance is known to recur in hematological neoplasms.
Collapse
Affiliation(s)
- Niccolò Di Giosaffatte
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Irene Bottillo
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Luigi Laino
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Giovanni Iaquinta
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Alessandro Ferraris
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Mariagrazia Garzia
- Haematology and Stem Cell Transplantation Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Simone Bargiacchi
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Claudia Mulargia
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Maria Rosaria Angelitti
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Fabiana Palumbo
- Haematology and Stem Cell Transplantation Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Barbara Grammatico
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Cinzia Bartolelli
- Obstetrics and Gynecology Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | | | - Luigi Rigacci
- Haematology and Stem Cell Transplantation Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Paola Grammatico
- Department of Molecular Medicine, Laboratory of Medical Genetics, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| |
Collapse
|
33
|
Clinical validation of a next-generation sequencing-based multi-cancer early detection "liquid biopsy" blood test in over 1,000 dogs using an independent testing set: The CANcer Detection in Dogs (CANDiD) study. PLoS One 2022; 17:e0266623. [PMID: 35471999 PMCID: PMC9041869 DOI: 10.1371/journal.pone.0266623] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Abstract
Cancer is the leading cause of death in dogs, yet there are no established screening paradigms for early detection. Liquid biopsy methods that interrogate cancer-derived genomic alterations in cell-free DNA in blood are being adopted for multi-cancer early detection in human medicine and are now available for veterinary use. The CANcer Detection in Dogs (CANDiD) study is an international, multi-center clinical study designed to validate the performance of a novel multi-cancer early detection “liquid biopsy” test developed for noninvasive detection and characterization of cancer in dogs using next-generation sequencing (NGS) of blood-derived DNA; study results are reported here. In total, 1,358 cancer-diagnosed and presumably cancer-free dogs were enrolled in the study, representing the range of breeds, weights, ages, and cancer types seen in routine clinical practice; 1,100 subjects met inclusion criteria for analysis and were used in the validation of the test. Overall, the liquid biopsy test demonstrated a 54.7% (95% CI: 49.3–60.0%) sensitivity and a 98.5% (95% CI: 97.0–99.3%) specificity. For three of the most aggressive canine cancers (lymphoma, hemangiosarcoma, osteosarcoma), the detection rate was 85.4% (95% CI: 78.4–90.9%); and for eight of the most common canine cancers (lymphoma, hemangiosarcoma, osteosarcoma, soft tissue sarcoma, mast cell tumor, mammary gland carcinoma, anal sac adenocarcinoma, malignant melanoma), the detection rate was 61.9% (95% CI: 55.3–68.1%). The test detected cancer signal in patients representing 30 distinct cancer types and provided a Cancer Signal Origin prediction for a subset of patients with hematological malignancies. Furthermore, the test accurately detected cancer signal in four presumably cancer-free subjects before the onset of clinical signs, further supporting the utility of liquid biopsy as an early detection test. Taken together, these findings demonstrate that NGS-based liquid biopsy can offer a novel option for noninvasive multi-cancer detection in dogs.
Collapse
|
34
|
Heesterbeek CJ, Aukema SM, Galjaard RJH, Boon EMJ, Srebniak MI, Bouman K, Faas BHW, Govaerts LCP, Hoffer MJV, den Hollander NS, Lichtenbelt KD, van Maarle MC, van Prooyen Schuurman L, van Rij MC, Schuring-Blom GH, Stevens SJC, Tan-Sindhunata G, Zamani Esteki M, de Die-Smulders CEM, Tjan-Heijnen VCG, Henneman L, Sistermans EA, Macville MVE. Noninvasive Prenatal Test Results Indicative of Maternal Malignancies: A Nationwide Genetic and Clinical Follow-Up Study. J Clin Oncol 2022; 40:2426-2435. [PMID: 35394817 DOI: 10.1200/jco.21.02260] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Noninvasive prenatal testing (NIPT) for fetal aneuploidy screening using cell-free DNA derived from maternal plasma can incidentally raise suspicion for cancer. Diagnostic routing after malignancy suspicious-NIPT faces many challenges. Here, we detail malignancy suspicious-NIPT cases, and describe the clinical characteristics, chromosomal aberrations, and diagnostic routing of the patients with a confirmed malignancy. Clinical lessons can be learned from our experience. METHODS Patients with NIPT results indicative of a malignancy referred for tumor screening between April 2017 and April 2020 were retrospectively included from a Dutch nationwide NIPT implementation study, TRIDENT-2. NIPT profiles from patients with confirmed malignancies were reviewed, and the pattern of chromosomal aberrations related to tumor type was analyzed. We evaluated the diagnostic contribution of clinical and genetic examinations. RESULTS Malignancy suspicious-NIPT results were reported in 0.03% after genome-wide NIPT, and malignancies confirmed in 16 patients (16/48, 33.3%). Multiple chromosomal aberrations were seen in 23 of 48 patients with genome-wide NIPT, and a malignancy was confirmed in 16 patients (16/23, 69.6%). After targeted NIPT, 0.005% malignancy suspicious-NIPT results were reported, in 2/3 patients a malignancy was confirmed. Different tumor types and stages were diagnosed, predominantly hematologic malignancies (12/18). NIPT data showed recurrent gains and losses in primary mediastinal B-cell lymphomas and classic Hodgkin lymphomas. Magnetic resonance imaging and computed tomography were most informative in diagnosing the malignancy. CONCLUSION In 231,896 pregnant women, a low percentage (0.02%) of NIPT results were assessed as indicative of a maternal malignancy. However, when multiple chromosomal aberrations were found, the risk of a confirmed malignancy was considerably high. Referral for extensive oncologic examination is recommended, and may be guided by tumor-specific hallmarks in the NIPT profile.
Collapse
Affiliation(s)
- Catharina J Heesterbeek
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Sietse M Aukema
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - Elles M J Boon
- Department of Human Genetics, and Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Katelijne Bouman
- Department of Human Genetics, and Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Brigitte H W Faas
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Mariëtte J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Klaske D Lichtenbelt
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Merel C van Maarle
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lisanne van Prooyen Schuurman
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Human Genetics, and Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Maartje C van Rij
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Servi J C Stevens
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Gita Tan-Sindhunata
- Department of Human Genetics, and Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Masoud Zamani Esteki
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Public Health, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Christine E M de Die-Smulders
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Vivianne C G Tjan-Heijnen
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lidewij Henneman
- Department of Human Genetics, and Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Erik A Sistermans
- Department of Human Genetics, and Amsterdam Reproduction & Development Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Merryn V E Macville
- Department of Clinical Genetics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, the Netherlands
| | | |
Collapse
|
35
|
Li J, Ju J, Zhao Q, Liu W, Yuan Y, Liu Q, Zhou L, Han Y, Yuan W, Huang Y, Xie Y, Li Z, Chen J, Huang S, Chen R, Li W, Tan M, Wang D, Zhou S, Zhang J, Zeng F, Yu N, Su F, Chen M, Ge Y, Huang Y, Jin X. Effective Identification of Maternal Malignancies in Pregnancies Undergoing Noninvasive Prenatal Testing. Front Genet 2022; 13:802865. [PMID: 35265103 PMCID: PMC8900746 DOI: 10.3389/fgene.2022.802865] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/10/2022] [Indexed: 11/22/2022] Open
Abstract
Background: The existence of maternal malignancy may cause false-positive results or failed tests of NIPT. Though recent studies have shown multiple chromosomal aneuploidies (MCA) are associated with malignancy, there is still no effective solution to identify maternal cancer patients from pregnant women with MCA results using NIPT. We aimed to develop a new method to effectively detect maternal cancer in pregnant women with MCA results using NIPT and a random forest classifier to identify the tissue origin of common maternal cancer types. Methods: For examination, 496 participants with MCA results via NIPT were enrolled from January 2016 to June 2019 at BGI. Cancer and non-cancer participants were confirmed through the clinical follow-up. The cohort comprising 42 maternal cancer cases and 294 non-cancer cases enrolled from January 2016 to December 2017 was utilized to develop a method named mean of the top five chromosome z scores (MTOP5Zscores). The remaining 160 participants enrolled from January 2018 to June 2019 were used to validate the performance of MTOP5Zscores. We established a random forest model to classify three common cancer types using normalized Pearson correlation coefficient (NPCC) values, z scores of 22 chromosomes, and seven plasma tumor markers (PTMs) as predictor variables. Results: 62 maternal cancer cases were confirmed with breast cancer, liver cancer, and lymphoma, the most common cancer types. MTOP5Zscores showed a sensitivity of 85% (95% confidence interval (CI), 62.11–96.79%) and specificity of 80% (95% CI, 72.41–88.28%) in the detection of maternal cancer among pregnant women with MCA results. The sensitivity of the classifier was 93.33, 66.67, and 50%, while specificity was 66.67, 90, and 97.06%, and positive predictive value (PPV) was 60.87, 72.73, and 80% for the prediction of breast cancer, liver cancer, and lymphoma, respectively. Conclusion: This study presents a solution to identify maternal cancer patients from pregnant women with MCA results using NIPT, indicating it as a value-added application of NIPT in the detection of maternal malignancies in addition to screening for fetal aneuploidies with no extra cost.
Collapse
Affiliation(s)
- Jia Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co., Ltd., Shijiazhuang, China
| | - Jia Ju
- BGI-Shenzhen, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Qiang Zhao
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Jiangmen, China
- Reproductive Medicine Center, Jiangmen Central Hospital, Jiangmen, China
| | - Weiqiang Liu
- Central Lab, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | | | - Qiang Liu
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Lijun Zhou
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Yuan Han
- BGI-Wuhan, BGI-Shenzhen, Wuhan, China
| | - Wen Yuan
- BGI-Wuhan, BGI-Shenzhen, Wuhan, China
| | - Yonghua Huang
- Department of Obstetrics and Gynecology, Jiangmen Central Hospital, Jiangmen, China
| | - Yingjun Xie
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhihua Li
- Department of Prenatal Diagnosis and Fetal Medical, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jingsi Chen
- Department of Prenatal Diagnosis and Fetal Medical, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuyu Huang
- The Department of Obstetrics, Foshan First People’s Hospital, Foshan, China
| | - Rufang Chen
- The Department of Obstetrics, Foshan First People’s Hospital, Foshan, China
| | - Wei Li
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Meihua Tan
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Danchen Wang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Si Zhou
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Shijiazhuang BGI Genomics Co., Ltd., Shijiazhuang, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Zhang
- Department of Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, Fujian province, China
| | | | - Nan Yu
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | | | - Min Chen
- Department of Prenatal Diagnosis and Fetal Medical, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xin Jin, ; Min Chen, ; Yunsheng Ge, ; Yanming Huang,
| | - Yunsheng Ge
- Department of Prenatal Diagnosis Center, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, Fujian province, China
- *Correspondence: Xin Jin, ; Min Chen, ; Yunsheng Ge, ; Yanming Huang,
| | - Yanming Huang
- Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, Guangdong province, China
- *Correspondence: Xin Jin, ; Min Chen, ; Yunsheng Ge, ; Yanming Huang,
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, China
- School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Xin Jin, ; Min Chen, ; Yunsheng Ge, ; Yanming Huang,
| |
Collapse
|
36
|
Harasim T, Neuhann T, Behnecke A, Stampfer M, Holinski-Feder E, Abicht A. Initial Clinical Experience with NIPT for Rare Autosomal Aneuploidies and Large Copy Number Variations. J Clin Med 2022; 11:jcm11020372. [PMID: 35054066 PMCID: PMC8777675 DOI: 10.3390/jcm11020372] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE Amniocentesis, chorionic villi sampling and first trimester combined testing are able to screen for common trisomies 13, 18, and 21 and other atypical chromosomal anomalies (ACA). The most frequent atypical aberrations reported are rare autosomal aneuploidies (RAA) and copy number variations (CNV), which are deletions or duplications of various sizes. We evaluated the clinical outcome of non-invasive prenatal testing (NIPT) results positive for RAA and large CNVs to determine the clinical significance of these abnormal results. METHODS Genome-wide NIPT was performed on 3664 eligible patient samples at a single genetics center. For patients with positive NIPT reports, the prescribing physician was asked retrospectively to provide clinical follow-up information using a standardized questionnaire. RESULTS RAAs and CNVs (>7 Mb) were detected in 0.5%, and 0.2% of tested cases, respectively. Follow up on pregnancies with an NIPT-positive result for RAA revealed signs of placental insufficiency or intra-uterine death in 50% of the cases and normal outcome at the time of birth in the other 50% of cases. We showed that CNV testing by NIPT allows for the detection of unbalanced translocations and relevant maternal health conditions. CONCLUSION NIPT for aneuploidies of all autosomes and large CNVs of at least 7 Mb has a low "non-reportable"-rate (<0.2%) and allows the detection of additional conditions of clinical significance.
Collapse
|
37
|
Dow E, Freimund A, Smith K, Hicks RJ, Jurcevic P, Shackleton M, James PA, Fellowes A, Delatycki MB, Fawcett S, Flowers N, Pertile MD, McGillivray G, Mileshkin L. Cancer Diagnoses Following Abnormal Noninvasive Prenatal Testing: A Case Series, Literature Review, and Proposed Management Model. JCO Precis Oncol 2022; 5:1001-1012. [PMID: 34994626 DOI: 10.1200/po.20.00429] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Noninvasive prenatal testing (NIPT) is a screening test for fetal chromosomal aneuploidy using cell-free DNA derived from maternal blood. It has been rapidly accepted into obstetric practice because of its application from 10-weeks' gestation, and its high sensitivity and specificity. NIPT results can be influenced by several factors including placental or maternal mosaicism and co-twin demise; cell-free DNA from a maternal origin can also complicate interpretation, with evidence that NIPT can detect previously unsuspected malignancies. This study aimed to develop management guidelines for women with NIPT results suspicious of maternal malignancy. The Peter MacCallum Cancer Center's experience of seven cases where abnormal NIPT results led to investigation for maternal malignancy between 2016 and 2019 were reviewed, along with the published literature. Six of the seven women (86%) referred for investigation were diagnosed with advanced malignancies, including colorectal cancer, breast cancer, melanoma, and Hodgkin lymphoma. Based on our single-center experience, as well as the available literature, guidelines for the investigation of women with NIPT results suspicious of malignancy are proposed, including utilization of fluorodeoxyglucose positron emission tomography-computed tomography, which had a high concordance with other investigations and diagnoses. These guidelines include maternal and fetal investigations, as well as consideration of the complex medical, psychologic, social, and ethical needs of these patients and their families.
Collapse
Affiliation(s)
- Eryn Dow
- Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Alison Freimund
- Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Kortnye Smith
- Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Rodney J Hicks
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Peter Jurcevic
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Australia
| | - Mark Shackleton
- Department of Oncology, Alfred Health, Melbourne, Australia.,Department of Medicine, Monash University, Clayton, Australia
| | - Paul A James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Andrew Fellowes
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Martin B Delatycki
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia.,Murdoch Children's Research Institute, Parkville, Australia
| | - Susan Fawcett
- Clinical Genetics Service, Royal Women's Hospital, Parkville, Australia
| | - Nicola Flowers
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Parkville, Australia
| | - Mark D Pertile
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Parkville, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - George McGillivray
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Parkville, Australia.,Clinical Genetics Service, Royal Women's Hospital, Parkville, Australia
| | - Linda Mileshkin
- Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
38
|
Yang YD, Cao Q, Li DZ. Influence of fibroids on cell-free DNA screening accuracy: what we need to know. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 59:127-128. [PMID: 34985817 DOI: 10.1002/uog.24818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/24/2021] [Indexed: 06/14/2023]
Affiliation(s)
- Y-D Yang
- Department of Ultrasound, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Q Cao
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| | - D-Z Li
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Scott F, Menezes M, Smet ME, Carey K, Hardy T, Fullston T, Rolnik DL, McLennan A. Influence of fibroids on cell-free DNA screening accuracy. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 59:114-119. [PMID: 34396623 DOI: 10.1002/uog.23763] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Cell-free DNA (cfDNA) screening assesses both maternal and placental cfDNA. Fibroids are common and release cfDNA into maternal serum. Genetic abnormality is seen in 50% of fibroids. We aimed to assess the impact of fibroids on the accuracy of genome-wide cfDNA screening. METHODS This was a prospective cohort study of singleton pregnancies examined at one of two centers in Melbourne and Sydney, Australia, between 1 November 2019 and 31 December 2020. All cases underwent pretest ultrasound examination to confirm an ongoing pregnancy of at least 10 weeks' gestation, and, at this stage, the number and volume of any uterine fibroid were documented. Genome-wide cfDNA screening was performed to detect all copy-number variants (CNV) > 7 megabases. The incidence of a false-positive result was compared between cases with and those without fibroids. RESULTS Over the 14-month study period, 13 184 patients underwent cfDNA screening, of whom 1017 (7.7%) had fibroids. Fibroids were not identified in any of the 17 participants who had a false-positive result for chromosomes 13, 18, 21, X or Y. Ninety-five (0.7%) cases were screen-positive for subchromosomal aberration (SA), rare autosomal trisomy (RAT) or multiple abnormalities (MA), with 10 of these cases having a fetal genetic abnormality. The incidence of a false-positive RAT, MA or SA result was significantly higher in participants with fibroids (20/1017 (2.0%)) than in those without fibroids (64/12 167 (0.5%)). Women with fibroids were approximately six times as likely to have a false-positive result for SA, and this was associated positively with both fibroid number and volume. CONCLUSIONS Most women with fibroids do not have an abnormal result on genome-wide cfDNA screening. However, CNVs due to fibroids are associated with false-positive SA findings, although fibroids do not appear to influence cfDNA screening accuracy for the common autosomal trisomies or sex-chromosomal abnormalities. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- F Scott
- Sydney Ultrasound for Women, Sydney, Australia
- University of New South Wales, Sydney, Australia
| | - M Menezes
- Monash Ultrasound for Women, Melbourne, Australia
| | - M E Smet
- Sydney Ultrasound for Women, Sydney, Australia
- Department of Obstetrics and Gynaecology, Westmead Hospital, Sydney, Australia
| | - K Carey
- Sydney Ultrasound for Women, Sydney, Australia
| | - T Hardy
- Repromed, Adelaide, Australia
- South Australia Pathology, Adelaide, Australia
| | | | - D L Rolnik
- Monash Ultrasound for Women, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - A McLennan
- Sydney Ultrasound for Women, Sydney, Australia
- University of Sydney, Sydney, Australia
| |
Collapse
|
40
|
OUP accepted manuscript. Clin Chem 2022; 68:627-632. [DOI: 10.1093/clinchem/hvac040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 11/12/2022]
|
41
|
Chen S, Yang M, Zhong N, Yu D, Jian J, Jiang D, Xiao Y, Wei W, Wang T, Lou Y, Zhou Z, Xu W, Wan W, Wu Z, Wei H, Liu T, Zhao J, Yang X, Xiao J. Quantified CIN Score From Cell-free DNA as a Novel Noninvasive Predictor of Survival in Patients With Spinal Metastasis. Front Cell Dev Biol 2021; 9:767340. [PMID: 34957099 PMCID: PMC8696126 DOI: 10.3389/fcell.2021.767340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: Most currently available scores for survival prediction of patients with bone metastasis lack accuracy. In this study, we present a novel quantified CIN (Chromosome Instability) score modeled from cfDNA copy number variation (CNV) for survival prediction. Experimental Design: Plasma samples collected from 67 patients with bone metastases from 11 different cancer types between November 2015 and May 2016 were sent through low-coverage whole genome sequencing followed by CIN computation to make a correlation analysis between the CIN score and survival prognosis. The results were validated in an independent cohort of 213 patients. Results: During the median follow-up period of 598 (95% CI 364-832) days until December 25, 2018, 124 (44.3%) of the total 280 patients died. Analysis of the discovery dataset showed that CIN score = 12 was the optimal CIN cutoff. Validation dataset showed that CIN was elevated (score ≥12) in 87 (40.8%) patients, including 5 (5.75%) with head and neck cancer, 11 (12.6%) with liver and gallbladder cancer, 11 (12.6%) with cancer from unidentified sites, 21 (24.1%) with lung cancer, 7 (8.05%) with breast cancer, 4 (4.60%) with thyroid cancer, 6 (6.90%) with colorectal cancer, 4 (4.60%) with kidney cancer, 2 (2.30%) with prostate cancer, and 16 (18.4%) with other types of cancer. Further analysis showed that patients with elevated CIN were associated with worse survival (p < 0.001). For patients with low Tokuhashi score (≤8) who had predictive survival of less than 6 months, the CIN score was able to distinguish patients with a median overall survival (OS) of 443 days (95% CI 301-585) from those with a median OS of 258 days (95% CI 184-332). Conclusion: CNV examination in bone metastatic cancer from cfDNA is superior to the traditional predictive model in that it provides a noninvasive and objective method of monitoring the survival of patients with spine metastasis.
Collapse
Affiliation(s)
- Su Chen
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Minglei Yang
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Nanzhe Zhong
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Dong Yu
- Center of Translational Medicine, Naval Medical University, Shanghai, China
| | - Jiao Jian
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Dongjie Jiang
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yasong Xiao
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wei Wei
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | | | - Yan Lou
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhenhua Zhou
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wei Xu
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wan Wan
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhipeng Wu
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Haifeng Wei
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Tielong Liu
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhao
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xinghai Yang
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianru Xiao
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
42
|
Gu W, Rauschecker AM, Hsu E, Zorn KC, Sucu Y, Federman S, Gopez A, Arevalo S, Sample HA, Talevich E, Nguyen ED, Gottschall M, Nourbakhsh B, Gold CA, Cree BAC, Douglas VC, Richie MB, Shah MP, Josephson SA, Gelfand JM, Miller S, Wang L, Tihan T, DeRisi JL, Chiu CY, Wilson MR. Detection of Neoplasms by Metagenomic Next-Generation Sequencing of Cerebrospinal Fluid. JAMA Neurol 2021; 78:1355-1366. [PMID: 34515766 DOI: 10.1001/jamaneurol.2021.3088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Importance Cerebrospinal fluid (CSF) cytologic testing and flow cytometry are insensitive for diagnosing neoplasms of the central nervous system (CNS). Such clinical phenotypes can mimic infectious and autoimmune causes of meningoencephalitis. Objective To ascertain whether CSF metagenomic next-generation sequencing (mNGS) can identify aneuploidy, a hallmark of malignant neoplasms, in difficult-to-diagnose cases of CNS malignant neoplasm. Design, Setting, and Participants Two case-control studies were performed at the University of California, San Francisco (UCSF). The first study used CSF specimens collected at the UCSF Clinical Laboratories between July 1, 2017, and December 31, 2019, and evaluated test performance in specimens from patients with a CNS malignant neoplasm (positive controls) or without (negative controls). The results were compared with those from CSF cytologic testing and/or flow cytometry. The second study evaluated patients who were enrolled in an ongoing prospective study between April 1, 2014, and July 31, 2019, with presentations that were suggestive of neuroinflammatory disease but who were ultimately diagnosed with a CNS malignant neoplasm. Cases of individuals whose tumors could have been detected earlier without additional invasive testing are discussed. Main Outcomes and Measures The primary outcome measures were the sensitivity and specificity of aneuploidy detection by CSF mNGS. Secondary subset analyses included a comparison of CSF and tumor tissue chromosomal abnormalities and the identification of neuroimaging characteristics that were associated with test performance. Results Across both studies, 130 participants were included (median [interquartile range] age, 57.5 [43.3-68.0] years; 72 men [55.4%]). The test performance study used 125 residual laboratory CSF specimens from 47 patients with a CNS malignant neoplasm and 56 patients with other neurological diseases. The neuroinflammatory disease study enrolled 12 patients and 17 matched control participants. The sensitivity of the CSF mNGS assay was 75% (95% CI, 63%-85%), and the specificity was 100% (95% CI, 96%-100%). Aneuploidy was detected in 64% (95% CI, 41%-83%) of the patients in the test performance study with nondiagnostic cytologic testing and/or flow cytometry, and in 55% (95% CI, 23%-83%) of patients in the neuroinflammatory disease study who were ultimately diagnosed with a CNS malignant neoplasm. Of the patients in whom aneuploidy was detected, 38 (90.5%) had multiple copy number variations with tumor fractions ranging from 31% to 49%. Conclusions and Relevance This case-control study showed that CSF mNGS, which has low specimen volume requirements, does not require the preservation of cell integrity, and was orginally developed to diagnose neurologic infections, can also detect genetic evidence of a CNS malignant neoplasm in patients in whom CSF cytologic testing and/or flow cytometry yielded negative results with a low risk of false-positive results.
Collapse
Affiliation(s)
- Wei Gu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco.,Department of Pathology, Stanford University, Stanford, California
| | - Andreas M Rauschecker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco
| | - Elaine Hsu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Kelsey C Zorn
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco
| | - Yasemin Sucu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Scot Federman
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Allan Gopez
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Shaun Arevalo
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Hannah A Sample
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco
| | | | - Eric D Nguyen
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Marc Gottschall
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland
| | - Carl A Gold
- Department of Neurology, Stanford University, Stanford, California
| | - Bruce A C Cree
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Vanja C Douglas
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Megan B Richie
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Maulik P Shah
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - S Andrew Josephson
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco.,Editor in Chief, JAMA Neurology
| | - Jeffrey M Gelfand
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| | - Steve Miller
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Linlin Wang
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco
| | - Tarik Tihan
- Department of Pathology, University of California, San Francisco, San Francisco
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco.,Chan Zuckerberg Biohub, San Francisco, California
| | - Charles Y Chiu
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, California.,Department of Medicine, Division of Infectious Diseases, University of California, San Francisco, San Francisco
| | - Michael R Wilson
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco
| |
Collapse
|
43
|
Jensen TJ, Goodman AM, Ellison CK, Holden KA, Kato S, Kim L, Daniels GA, Fitzgerald K, McCarthy E, Nakashe P, Mazloom AR, Almasri E, McLennan G, Grosu DS, Eisenberg M, Kurzrock R. Genome-wide Sequencing of Cell-free DNA Enables Detection of Copy-number Alterations in Patients with Cancer Where Tissue Biopsy is Not Feasible. Mol Cancer Ther 2021; 20:2274-2279. [PMID: 34465593 PMCID: PMC9398131 DOI: 10.1158/1535-7163.mct-20-1066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/03/2021] [Accepted: 08/20/2021] [Indexed: 01/07/2023]
Abstract
When tissue biopsy is not medically prudent or tissue is insufficient for molecular testing, alternative methods are needed. Because cell-free DNA (cfDNA) has been shown to provide a representative surrogate for tumor tissue, we sought to evaluate its utility in this clinical scenario. cfDNA was isolated from the plasma of patients and assayed with low-coverage (∼0.3×), genome-wide sequencing. Copy-number alterations (CNA) were identified and characterized using analytic methods originally developed for noninvasive prenatal testing (NIPT) and quantified using the genomic instability number (GIN), a metric that reflects the quantity and magnitude of CNAs across the genome. The technical variability of the GIN was first evaluated in an independent cohort comprising genome-wide sequencing results from 27,754 women who consented to have their samples used for research and whose NIPT results yielded no detected CNAs to establish a detection threshold. Subsequently, cfDNA sequencing data from 96 patients with known cancers but for whom a tissue biopsy could not be obtained are presented. An elevated GIN was detected in 35% of patients and detection rates varied by tumor origin. Collectively, CNAs covered 96.6% of all autosomes. Survival was significantly reduced in patients with an elevated GIN relative to those without. Overall, these data provide a proof of concept for the use of low-coverage, genome-wide sequencing of cfDNA from patients with cancer to obtain relevant molecular information in instances where tissue is difficult to access. These data may ultimately serve as an informative complement to other molecular tests.
Collapse
Affiliation(s)
- Taylor J. Jensen
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California.,Laboratory Corporation of America, Durham, North Carolina.,Corresponding Author: Taylor J. Jensen, Research and Development, Laboratory Corporation of America, 1912 TW Alexander, Durham, NC 27703. Phone: 858-242-6842; E-mail:
| | - Aaron M. Goodman
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego.,Department of Medicine, Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California, San Diego
| | - Christopher K. Ellison
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Kimberly A. Holden
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Shumei Kato
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego.,Department of Medicine, Division of Precision Medicine, Moores Cancer Center, University of California, San Diego
| | - Lisa Kim
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego
| | - Gregory A. Daniels
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego
| | - Kerry Fitzgerald
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Erin McCarthy
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Prachi Nakashe
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Amin R. Mazloom
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Eyad Almasri
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Graham McLennan
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Daniel S. Grosu
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | | | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego.,Department of Medicine, Division of Precision Medicine, Moores Cancer Center, University of California, San Diego
| |
Collapse
|
44
|
Schobers G, Koeck R, Pellaers D, Stevens SJC, Macville MVE, Paulussen ADC, Coonen E, van den Wijngaard A, de Die-Smulders C, de Wert G, Brunner HG, Zamani Esteki M. Liquid biopsy: state of reproductive medicine and beyond. Hum Reprod 2021; 36:2824-2839. [PMID: 34562078 PMCID: PMC8523207 DOI: 10.1093/humrep/deab206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 08/06/2021] [Indexed: 01/23/2023] Open
Abstract
Liquid biopsy is the process of sampling and analyzing body fluids, which enables non-invasive monitoring of complex biological systems in vivo. Liquid biopsy has myriad applications in health and disease as a wide variety of components, ranging from circulating cells to cell-free nucleic acid molecules, can be analyzed. Here, we review different components of liquid biopsy, survey state-of-the-art, non-invasive methods for detecting those components, demonstrate their clinical applications and discuss ethical considerations. Furthermore, we emphasize the importance of artificial intelligence in analyzing liquid biopsy data with the aim of developing ethically-responsible non-invasive technologies that can enhance individualized healthcare. While previous reviews have mainly focused on cancer, this review primarily highlights applications of liquid biopsy in reproductive medicine.
Collapse
Affiliation(s)
- Gaby Schobers
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rebekka Koeck
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Dominique Pellaers
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Servi J C Stevens
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Merryn V E Macville
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Aimée D C Paulussen
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Edith Coonen
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Center for Reproductive Medicine, Maastricht University Medical Centre+, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Christine de Die-Smulders
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Guido de Wert
- Faculty of Health, Medicine and Life Sciences, Department of Health, Ethics and Society, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Han G Brunner
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Masoud Zamani Esteki
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
45
|
Goodman AM, Holden KA, Jeong AR, Kim L, Fitzgerald KD, Almasri E, McLennan G, Eisenberg M, Jahromi AH, Hoh C, Hurley M, Mulroney C, Tzachanis D, Ball ED, Jensen TJ, Kurzrock R. Assessing CAR T-Cell Therapy Response Using Genome-Wide Sequencing of Cell-Free DNA in Patients With B-Cell Lymphomas. Transplant Cell Ther 2021; 28:30.e1-30.e7. [PMID: 34655803 DOI: 10.1016/j.jtct.2021.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
Methods that enable monitoring of therapeutic efficacy of autologous chimeric antigen receptor (CAR) T-cell therapy will be clinically useful. The aim of this study is to demonstrate the feasibility of blood-derived cell-free DNA (cfDNA) to predict CAR T-cell therapy response in patients with refractory B-cell lymphomas. Whole blood was collected before and throughout CAR T-cell therapy until day 154. Low-coverage (∼0.4×), genome-wide cfDNA sequencing, similar to that established for noninvasive prenatal testing, was performed. The genomic instability number (GIN) was used to quantify plasma copy number alteration level. Twelve patients were enrolled. Seven (58%) patients achieved a complete response (CR); 2 (25%), a partial response. Median progression-free survival was 99 days; median overall survival was not reached (median follow-up, 247 days). Altogether, 127 blood samples were analyzed (median, 10 samples/patient [range 8-13]). All 5 patients who remained in CR at the time of last measurement had GIN <170 (threshold). Two patients who attained CR, but later relapsed, and all but one patient who had best response other than CR had last GIN measurement of >170. In 5 of 6 patients with relapsed or progressive disease, increasing GIN was observed before the diagnosis by imaging. The abundance of CAR T-cell construct (absolute number of construct copies relative to the number of human genome equivalents) also showed a trend to correlate with outcome (day 10, P = .052). These data describe a proof-of-concept for the use of multiple liquid biopsy technologies to monitor therapeutic response in B-cell lymphoma patients receiving CAR T-cell therapy.
Collapse
Affiliation(s)
- Aaron M Goodman
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California.
| | | | - Ah-Reum Jeong
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, California
| | - Lisa Kim
- Laboratory Corporation of America, San Diego, California
| | | | - Eyad Almasri
- Laboratory Corporation of America, San Diego, California
| | | | | | - Amin H Jahromi
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Carl Hoh
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Michael Hurley
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California
| | - Carolyn Mulroney
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California
| | - Dimitrios Tzachanis
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California
| | - Edward D Ball
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, California
| | - Taylor J Jensen
- Laboratory Corporation of America, San Diego, California; Laboratory Corporation of America, Durham, North Carolina
| | | |
Collapse
|
46
|
Lannoo L, Lenaerts L, Van Den Bogaert K, Che H, Brison N, Devriendt K, Amant F, Vermeesch JR, Van Calsteren K. Non-invasive prenatal testing suggesting a maternal malignancy: What do we tell the prospective parents in Belgium? Prenat Diagn 2021; 41:1264-1272. [PMID: 34405430 DOI: 10.1002/pd.6031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 12/20/2022]
Abstract
Cancer is diagnosed in one in 1000 to 1500 pregnancies. Most frequently encountered malignancies during pregnancy are breast cancer, hematological cancer, cervical cancer and malignant melanoma. Maternal cancer is associated with an increased risk of IUGR and preterm labor, especially in patients with systemic disease or those receiving chemotherapy during pregnancy, requiring a high-risk obstetrical follow-up. Fetal aneuploidy screening by non-invasive prenatal testing (NIPT) can lead to the incidental identification of copy number alterations derived from non-fetal cell-free DNA (cfDNA), as seen in certain cases of maternal malignancy. The identification of tumor-derived cfDNA requires further clinical, biochemical, radiographic and histological investigations to confirm the diagnosis. In such cases, reliable risk estimation for fetal trisomy 21, 18 and 13 is impossible. Therefore, invasive testing should be offered when ultrasonographic screening reveals an increased risk for chromosomal anomalies, or when a more accurate test is desired. When the fetal karyotype is normal, long term implications for the fetus refer to the consequences of the maternal disease and treatment during pregnancy. This manuscript addresses parental questions when NIPT suggests a maternal malignancy. Based on current evidence and our own experience, a clinical management scheme in a multidisciplinary setting is proposed.
Collapse
Affiliation(s)
- Lore Lannoo
- Department of Development and Regeneration, Division Woman and Child, Clinical Department Obstetrics and Gynaecology, University Hospital Leuven, KULeuven, Leuven, Belgium
| | | | | | - Huiwen Che
- Department of Human Genetics, KULeuven, Leuven, Belgium
| | | | | | - Frédéric Amant
- Department of Gynaecological Oncology, KULeuven, Leuven, Belgium.,Center for Gynecological Oncology Amsterdam, Academic Medical Centre Amsterdam, University of Amsterdam, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | | | - Kristel Van Calsteren
- Department of Development and Regeneration, Division Woman and Child, Clinical Department Obstetrics and Gynaecology, University Hospital Leuven, KULeuven, Leuven, Belgium
| |
Collapse
|
47
|
Calculation of Fetal Fraction for Non-Invasive Prenatal Testing. BIOTECH 2021; 10:biotech10030017. [PMID: 35822771 PMCID: PMC9245487 DOI: 10.3390/biotech10030017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/05/2022] Open
Abstract
Estimating the fetal fraction of DNA in a pregnant mother’s blood is a risk-free, non-invasive way of predicting fetal aneuploidy. It is a rapidly developing field of study, offering researchers a plethora of different complementary methods. Such methods include examining the differences in methylation profiles between the fetus and the mother. Others include calculating the average allele frequency based on the difference in genotype of a number of single-nucleotide polymorphisms. Differences in the length distribution of DNA fragments between the mother and the fetus as well as measuring the proportion of DNA reads mapping to the Y chromosome also constitute fetal fraction estimation methods. The advantages and disadvantages of each of these main method types are discussed. Moreover, several well-known fetal fraction estimation methods, such as SeqFF, are described and compared with other methods. These methods are amenable to not only the estimation of fetal fraction but also paternity, cancer, and transplantation monitoring studies. NIPT is safe, and should aneuploidy be detected, this information can help parents prepare mentally and emotionally for the birth of a special needs child.
Collapse
|
48
|
Scott F, Smet ME, Hardy T, Sundercombe S, Friedlander M, Carey L, Kirk E, Li B, McLennan A. Concurrent maternal malignancy and fetal trisomy detected using genome-wide noninvasive prenatal screening. Prenat Diagn 2021; 41:1273-1276. [PMID: 34318961 DOI: 10.1002/pd.6020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Fergus Scott
- Sydney Ultrasound for Women, Sydney, Australia.,University of New South Wales, Sydney, Australia
| | - Maria-Elisabeth Smet
- Sydney Ultrasound for Women, Sydney, Australia.,Fetal Medicine Unit, Westmead Hospital, Sydney, Australia
| | | | | | - Michael Friedlander
- University of New South Wales, Sydney, Australia.,Department of Medical Oncology, Prince of Wales Clinical School, The Prince of Wales Hospital, Sydney, Australia
| | - Louise Carey
- NSW Health Pathology Randwick Genomics, Randwick, Australia.,School of Women's and Children's Health, UNSW, Sydney Children's Hospital, Randwick, Australia
| | - Edwin Kirk
- NSW Health Pathology Randwick Genomics, Randwick, Australia.,School of Women's and Children's Health, UNSW, Sydney Children's Hospital, Randwick, Australia
| | - Biao Li
- Department of Bioinformatics, Illumina Inc., San Diego, CA, USA
| | - Andrew McLennan
- Sydney Ultrasound for Women, Sydney, Australia.,Discipline of Obstetrics, Gynaecology and Neonatology, The University of Sydney, Sydney, Australia
| |
Collapse
|
49
|
Gu W, Talevich E, Hsu E, Qi Z, Urisman A, Federman S, Gopez A, Arevalo S, Gottschall M, Liao L, Tung J, Chen L, Lim H, Ho C, Kasowski M, Oak J, Holmes BJ, Yeh I, Yu J, Wang L, Miller S, DeRisi JL, Prakash S, Simko J, Chiu CY. Detection of cryptogenic malignancies from metagenomic whole genome sequencing of body fluids. Genome Med 2021; 13:98. [PMID: 34074327 PMCID: PMC8167833 DOI: 10.1186/s13073-021-00912-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 05/20/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Metagenomic next-generation sequencing (mNGS) of body fluids is an emerging approach to identify occult pathogens in undiagnosed patients. We hypothesized that metagenomic testing can be simultaneously used to detect malignant neoplasms in addition to infectious pathogens. METHODS From two independent studies (n = 205), we used human data generated from a metagenomic sequencing pipeline to simultaneously screen for malignancies by copy number variation (CNV) detection. In the first case-control study, we analyzed body fluid samples (n = 124) from patients with a clinical diagnosis of either malignancy (positive cases, n = 65) or infection (negative controls, n = 59). In a second verification cohort, we analyzed a series of consecutive cases (n = 81) sent to cytology for malignancy workup that included malignant positives (n = 32), negatives (n = 18), or cases with an unclear gold standard (n = 31). RESULTS The overall CNV test sensitivity across all studies was 87% (55 of 63) in patients with malignancies confirmed by conventional cytology and/or flow cytometry testing and 68% (23 of 34) in patients who were ultimately diagnosed with cancer but negative by conventional testing. Specificity was 100% (95% CI 95-100%) with no false positives detected in 77 negative controls. In one example, a patient hospitalized with an unknown pulmonary illness had non-diagnostic lung biopsies, while CNVs implicating a malignancy were detectable from bronchoalveolar fluid. CONCLUSIONS Metagenomic sequencing of body fluids can be used to identify undetected malignant neoplasms through copy number variation detection. This study illustrates the potential clinical utility of a single metagenomic test to uncover the cause of undiagnosed acute illnesses due to cancer or infection using the same specimen.
Collapse
Affiliation(s)
- Wei Gu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA.
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA.
| | | | - Elaine Hsu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Zhongxia Qi
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Anatoly Urisman
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Scot Federman
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Allan Gopez
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Shaun Arevalo
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Marc Gottschall
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Linda Liao
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA
| | - Jack Tung
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Lei Chen
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA
| | - Harumi Lim
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA
| | - Chandler Ho
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA
| | - Maya Kasowski
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Jean Oak
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA
| | - Brittany J Holmes
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
- Stanford Health Care, Stanford University, Stanford, CA, 94305, USA
| | - Iwei Yeh
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Jingwei Yu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Linlin Wang
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Steve Miller
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 94107, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94107, USA
| | - Sonam Prakash
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Jeff Simko
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94107, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94107, USA.
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, 91407, USA.
- Department of Medicine, Division of Infectious Diseases, University of California San Francisco, San Francisco, CA, 94107, USA.
| |
Collapse
|
50
|
Lenaerts L, Che H, Brison N, Neofytou M, Jatsenko T, Lefrère H, Maggen C, Villela D, Verheecke M, Dehaspe L, Croitor A, Hatse S, Wildiers H, Neven P, Vandecaveye V, Floris G, Vermeesch JR, Amant F. Breast Cancer Detection and Treatment Monitoring Using a Noninvasive Prenatal Testing Platform: Utility in Pregnant and Nonpregnant Populations. Clin Chem 2021; 66:1414-1423. [PMID: 33141904 DOI: 10.1093/clinchem/hvaa196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/24/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Numerous publications have reported the incidental detection of occult malignancies upon routine noninvasive prenatal testing (NIPT). However, these studies were not designed to evaluate the NIPT performance for cancer detection. METHODS We investigated the sensitivity of a genome-wide NIPT pipeline, called GIPSeq, for detecting cancer-specific copy number alterations (CNAs) in plasma tumor DNA (ctDNA) of patients with breast cancer. To assess whether a pregnancy itself, with fetal cell-free DNA (cfDNA) in the maternal circulation, might influence the detection of ctDNA, results were compared in pregnant (n = 25) and nonpregnant (n = 25) cancer patients. Furthermore, the ability of GIPSeq to monitor treatment response was assessed. RESULTS Overall GIPSeq sensitivity for detecting cancer-specific CNAs in plasma cfDNA was 26%. Fifteen percent of detected cases were asymptomatic at the time of blood sampling. GIPSeq sensitivity mainly depended on the tumor stage. Also, triple negative breast cancers (TNBC) were more frequently identified compared to hormone-positive or HER2-enriched tumors. This might be due to the presence of high-level gains and losses of cfDNA or high ctDNA loads in plasma of TNBC. Although higher GIPSeq sensitivity was noted in pregnant (36%) than in nonpregnant women (16%), the limited sample size prohibits a definite conclusion. Finally, GIPSeq profiling of cfDNA during therapy allowed monitoring of early treatment response. CONCLUSIONS The results underscore the potential of NIPT-based tests, analyzing CNAs in plasma cfDNA in a genome-wide and unbiased fashion for breast cancer detection, cancer subtyping and treatment monitoring in a pregnant and nonpregnant target population.
Collapse
Affiliation(s)
- Liesbeth Lenaerts
- Department of Oncology, Laboratory of Gynecological Oncology, KU Leuven, Leuven, Belgium
| | - Huiwen Che
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium
| | - Nathalie Brison
- Centre of Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Maria Neofytou
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium.,Cancer Research UK Cambridge Institute, Molecular and Computational Diagnostics, University of Cambridge, Cambridge, UK
| | - Tatjana Jatsenko
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium
| | - Hanne Lefrère
- Department of Oncology, Laboratory of Gynecological Oncology, KU Leuven, Leuven, Belgium
| | - Charlotte Maggen
- Department of Oncology, Laboratory of Gynecological Oncology, KU Leuven, Leuven, Belgium.,Department of Gynecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium
| | - Darine Villela
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium.,Departamento de Genética e Biologia Evolutiva, University of São Paulo, São Paulo, Brazil
| | - Magali Verheecke
- Gynaecology and Obstetrics Department, General Hospital Turnhout, Turnhout, Belgium
| | - Luc Dehaspe
- Genomics Core Facility, University Hospitals Leuven, Leuven, Belgium
| | - Anca Croitor
- Department of Imaging and Pathology, Unit of Biomedical MRI, KU Leuven, Leuven, Belgium
| | - Sigrid Hatse
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Hans Wildiers
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium.,Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Oncology, Laboratory of Gynecological Oncology, KU Leuven, Leuven, Belgium.,Department of Gynecology and Obstetrics, University Hospitals Leuven, Leuven, Belgium
| | - Vincent Vandecaveye
- Department of Imaging and Pathology, Unit of Translational MRI, KU Leuven, Leuven, Belgium.,Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Imaging and Pathology, Unit of Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Joris Robert Vermeesch
- Department of Human Genetics, Laboratory for Cytogenetics and Genome Research, KU Leuven, Leuven, Belgium.,Centre of Human Genetics, University Hospitals Leuven, Leuven, Belgium.,Genomics Core Facility, University Hospitals Leuven, Leuven, Belgium
| | - Frédéric Amant
- Department of Oncology, Laboratory of Gynecological Oncology, KU Leuven, Leuven, Belgium.,Department of 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
| |
Collapse
|