1
|
Bao H, Yang S, Chen X, Dong G, Mao Y, Wu S, Cheng X, Wu X, Tang W, Wu M, Tang S, Liang W, Wang Z, Yang L, Liu J, Wang T, Zhang B, Jiang K, Xu Q, Chen J, Huang H, Peng J, Xia X, Wu Y, Xu S, Tao J, Chong L, Zhu D, Yang R, Chang S, He P, Xu X, Zhang J, Shen Y, Jiang Y, Liu S, Zhang X, Wu X, Wang X, Shao Y. Early detection of multiple cancer types using multidimensional cell-free DNA fragmentomics. Nat Med 2025:10.1038/s41591-025-03735-2. [PMID: 40425843 DOI: 10.1038/s41591-025-03735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 04/24/2025] [Indexed: 05/29/2025]
Abstract
The multicancer early detection (MCED) test has the potential to enhance current cancer-screening methods. We evaluated a new MCED test that analyzes plasma cell-free DNA using genetic- and fragmentomics-based features from whole-genome sequencing. The present study included an internal validation cohort of 3,021 patients with cancer and 3,370 noncancer controls, and an independent cohort of 677 patients with cancer and 687 noncancer individuals. The results demonstrated an overall sensitivity of 87.4%, specificity of 97.8% and tissue-of-origin prediction accuracy of 82.4% in the independent validation cohort. Preliminary results from a prospective study of 3,724 asymptomatic participants showed a sensitivity of 53.5% (predominantly early stage cancers) and specificity of 98.1%. These findings indicate that the MCED test has strong potential to improve early cancer detection and support clinical decision-making.
Collapse
Affiliation(s)
- Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Shanshan Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xiaoxi Chen
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Guangqiang Dong
- Nanjing Jiangbei New Area Center for Public Health Service, Nanjing, China
| | - Yuan Mao
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuyu Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xi Cheng
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xuxiaochen Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Wanxiangfu Tang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Min Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Shiting Tang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zheng Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liu Yang
- Colorectal Center, Jiangsu Cancer Hospital, Nanjing, China
| | - Jiaqi Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center, Cancer Hospital of the Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, Nanjing, China
| | - Bingzhong Zhang
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Xu
- Departments of Gynecology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fujian, China
| | - Jierong Chen
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Hairong Huang
- Department of Thoracic Surgery, Eastern Theater Command Hospital, Nanjing, China
| | - Junjie Peng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaomeng Xia
- Department of Gynaecology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yumei Wu
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing, China
| | - Shun Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ji Tao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Li Chong
- Department of Respiratory Medicine, First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dongqin Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Ruowei Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Shuang Chang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Peng He
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xiuxiu Xu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - JinPeng Zhang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yi Shen
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Ya Jiang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Sisi Liu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xian Zhang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xiaonan Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China.
- School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Imai M, Nakamura Y, Yoshino T. Transforming cancer screening: the potential of multi-cancer early detection (MCED) technologies. Int J Clin Oncol 2025; 30:180-193. [PMID: 39799530 PMCID: PMC11785667 DOI: 10.1007/s10147-025-02694-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
Early cancer detection substantially improves the rate of patient survival; however, conventional screening methods are directed at single anatomical sites and focus primarily on a limited number of cancers, such as gastric, colorectal, lung, breast, and cervical cancer. Additionally, several cancers are inadequately screened, hindering early detection of 45.5% cases. In contrast, Multi-Cancer Early Detection (MCED) assays offer simultaneous screening of multiple cancers from a single liquid biopsy and identify molecular changes before symptom onset. These tests assess DNA mutations, abnormal DNA methylation patterns, fragmented DNA, and other tumor-derived biomarkers, indicating the presence of cancer and predicting its origin. Moreover, MCED assays concurrently detect multiple cancers without recommended screening protocols, potentially revolutionizing cancer screening and management. Large trials have reported promising results, achieving 50-95% sensitivity and 89-99% specificity for multiple cancer types. However, challenges, regarding improving accuracy, addressing ethical issues (e.g., psychosocial impact assessment), and integrating MCED into healthcare systems, must be addressed to achieve widespread adoption. Furthermore, prospective multi-institutional studies are crucial for demonstrating the clinical benefits in diverse populations. This review provides an overview of the principles, development status, and clinical significance of MCED tests, and discusses their potential and challenges.
Collapse
Affiliation(s)
- Mitsuho Imai
- Translational Research Support Section, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Section, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Translational Research Support Section, National Cancer Center Hospital East, Chiba, Japan.
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
3
|
Trosman JR, Weldon CB, Kurian AW, Mrig E, Phillips KA. Implementation of Cancer Genomics in the United States: Views of Payers and Other Stakeholders on Challenges and the Role of Payers in Solutions. JCO Precis Oncol 2025; 9:e2400822. [PMID: 40020207 PMCID: PMC11875457 DOI: 10.1200/po-24-00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
PURPOSE Genomic testing is crucial in cancer risk identification, diagnosis, and treatment. However, health care implementation is challenging, even for tests covered by insurance. US payers are important health care participants and may contribute to addressing implementation challenges. We explored whether and how payers consider their role in genomic test implementation, and the perspectives of nonpayer stakeholders on payers' participation. METHODS We conducted a group interview with private payers (N = 12) to elucidate views on their role in genomic test implementation, implementation challenges, and potential solutions. Subsequently, we conducted individual interviews (N = 10) with nonpayer stakeholders-five cancer advocacy groups and five medical societies working in cancer, examining their reactions to payer input and capturing additional ideas. Qualitative research methods were used to frame the study and analyze results. RESULTS Payers considered cancer genomics implementation important and expressed willingness and ability to be involved. They articulated specific challenges relevant to them, including underutilization of covered tests in clinical practice, inequitable test use, and inconsistencies across guidelines, and suggested specific solutions to collaborate on with other stakeholders. Stakeholders viewed payers' participation in implementation as appropriate and feasible, expressed willingness to work with payers where relevant, and concurred with the key challenges noted by payers. Stakeholders' agreement with payers' solutions varied, but they offered additional ideas for addressing challenges. CONCLUSION Both payers and other stakeholders considered payers' role in addressing genomics implementation challenges appropriate and feasible, and offered specific avenues for payer participation. Our findings inform efforts by payers and other stakeholders to address broad health care implementation. They may also help precision oncology professionals, cancer centers, and health systems to frame their own implementation efforts and influence a broader policy and implementation agenda.
Collapse
Affiliation(s)
- Julia R Trosman
- UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
- Center for Business Models in Healthcare, Glencoe, IL
| | - Christine B Weldon
- UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA
- Center for Business Models in Healthcare, Glencoe, IL
| | - Allison W Kurian
- Departments of Medicine and of Epidemiology and Population Health, Stanford University, Stanford, CA
| | - Emily Mrig
- UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
| | - Kathryn A Phillips
- UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
| |
Collapse
|
4
|
Rubinstein WS, Patriotis C, Dickherber A, Han PKJ, Katki HA, LeeVan E, Pinsky PF, Prorok PC, Skarlupka AL, Temkin SM, Castle PE, Minasian LM. Cancer screening with multicancer detection tests: A translational science review. CA Cancer J Clin 2024; 74:368-382. [PMID: 38517462 PMCID: PMC11226362 DOI: 10.3322/caac.21833] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
Multicancer detection (MCD) tests use a single, easily obtainable biospecimen, such as blood, to screen for more than one cancer concurrently. MCD tests can potentially be used to improve early cancer detection, including cancers that currently lack effective screening methods. However, these tests have unknown and unquantified benefits and harms. MCD tests differ from conventional cancer screening tests in that the organ responsible for a positive test is unknown, and a broad diagnostic workup may be necessary to confirm the location and type of underlying cancer. Among two prospective studies involving greater than 16,000 individuals, MCD tests identified those who had some cancers without currently recommended screening tests, including pancreas, ovary, liver, uterus, small intestine, oropharyngeal, bone, thyroid, and hematologic malignancies, at early stages. Reported MCD test sensitivities range from 27% to 95% but differ by organ and are lower for early stage cancers, for which treatment toxicity would be lowest and the potential for cure might be highest. False reassurance from a negative MCD result may reduce screening adherence, risking a loss in proven public health benefits from standard-of-care screening. Prospective clinical trials are needed to address uncertainties about MCD accuracy to detect different cancers in asymptomatic individuals, whether these tests can detect cancer sufficiently early for effective treatment and mortality reduction, the degree to which these tests may contribute to cancer overdiagnosis and overtreatment, whether MCD tests work equally well across all populations, and the appropriate diagnostic evaluation and follow-up for patients with a positive test.
Collapse
Affiliation(s)
- Wendy S. Rubinstein
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Christos Patriotis
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Anthony Dickherber
- Center for Strategic Scientific Initiatives, US National Cancer Institute, Rockville, Maryland, USA
| | - Paul K. J. Han
- Division of Cancer Control and Population Sciences, US National Cancer Institute, Rockville, Maryland, USA
| | - Hormuzd A. Katki
- Division of Cancer Epidemiology and Genetics, US National Cancer Institute, Rockville, Maryland, USA
| | - Elyse LeeVan
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Paul F. Pinsky
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Philip C. Prorok
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Amanda L. Skarlupka
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| | - Sarah M. Temkin
- National Institutes of Health Office of Research on Women’s Health, Bethesda, Maryland, USA
| | - Philip E. Castle
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
- Division of Cancer Epidemiology and Genetics, US National Cancer Institute, Rockville, Maryland, USA
| | - Lori M. Minasian
- Division of Cancer Prevention, US National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
5
|
Lange JM, Gogebakan KC, Gulati R, Etzioni R. Projecting the Impact of Multi-Cancer Early Detection on Late-Stage Incidence Using Multi-State Disease Modeling. Cancer Epidemiol Biomarkers Prev 2024; 33:830-837. [PMID: 38506751 PMCID: PMC11213491 DOI: 10.1158/1055-9965.epi-23-1470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/29/2024] [Accepted: 03/18/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Downstaging-reduction in late-stage incidence-has been proposed as an endpoint in randomized trials of multi-cancer early detection (MCED) tests. How downstaging depends on test performance and follow-up has been studied for some cancers but is understudied for cancers without existing screening and for MCED tests that include these cancer types. METHODS We develop a model for cancer natural history that can be fit to registry incidence patterns under minimal inputs and can be estimated for solid cancers without existing screening. Fitted models are combined to project downstaging in MCED trials given sensitivity for early- and late-stage cancers. We fit models for 12 cancers using incidence data from the Surveillance, Epidemiology, and End Results program and project downstaging in a simulated trial under variable preclinical latencies and test sensitivities. RESULTS A proof-of-principle lung cancer model approximated downstaging in the National Lung Screening Trial. Given published stage-specific sensitivities for 12 cancers, we projected downstaging ranging from 21% to 43% across plausible preclinical latencies in a hypothetical 3-screen MCED trial. Late-stage incidence reductions manifest soon after screening begins. Downstaging increases with longer early-stage latency or higher early-stage test sensitivity. CONCLUSIONS Even short-term MCED trials could produce substantial downstaging given adequate early-stage test sensitivity. IMPACT Modeling the natural histories of cancers without existing screening facilitates analysis of novel MCED products and trial designs. The framework informs expectations of MCED impact on disease stage at diagnosis and could serve as a building block for designing trials with late-stage incidence as the primary endpoint.
Collapse
Affiliation(s)
- Jane M. Lange
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University
| | | | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Center
- Department of Health Services, University of Washington
| |
Collapse
|
6
|
Greene MP, Vassy JL. Helping patients understand multi-cancer early detection tests: a scoping review. Per Med 2024; 21:131-137. [PMID: 38269535 DOI: 10.2217/pme-2023-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Multi-cancer early detection tests are emerging as a revolutionary technology for the early detection of dozens of cancers from a single blood sample, including cancers without proven screening methods. However, they also come with challenges, including false-positive and false-negative results. To help patients make informed decisions, patient education materials are crucial. A review of available materials reveals that, while some materials provide understandable and actionable information, most lack a balanced presentation of the current benefits and risks of multi-cancer early detection testing. The dynamic nature of this field necessitates continuous updates to educational materials, incorporating current evidence and uncertainties.
Collapse
Affiliation(s)
| | - Jason L Vassy
- Harvard Medical School, Boston, 02115 MA, USA
- Veterans Affairs Boston Healthcare System, Boston, 02130 MA, USA
- Brigham and Women's Hospital, Boston, 02115 MA, USA
- Precision Population Health, Ariadne Labs, Boston, 02215 MA, USA
| |
Collapse
|
7
|
Bamodu OA, Chung CC, Pisanic TR. Harnessing liquid biopsies: Exosomes and ctDNA as minimally invasive biomarkers for precision cancer medicine. THE JOURNAL OF LIQUID BIOPSY 2023; 2:100126. [PMID: 40028482 PMCID: PMC11863985 DOI: 10.1016/j.jlb.2023.100126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 03/05/2025]
Abstract
Liquid biopsies have emerged as groundbreaking tools for minimally invasive monitoring of cancer, encompassing the analysis of Cell-Free DNA (cfDNA), circulating tumor DNA (ctDNA) and exosomes. This paradigm shift offers an emerging approach for understanding tumor dynamics, treatment responses, and disease progression. Leveraging advancements in molecular biology and technology, liquid biopsies enable clinicians to gain intricate insights from peripheral blood, thereby transforming the landscape of cancer care. This review describes the clinical impact, technological innovations, and recent evidence surrounding the integration of ctDNA and exosome analysis in cancer monitoring. Through early detection, real-time treatment response assessment, and the tracking of minimal residual disease, liquid biopsies have redefined the standards of precision oncology. Key advancements in ctDNA analysis, such as high-throughput sequencing and digital PCR, empower the detection of actionable mutations with high sensitivity. Concurrently, the characterization of exosomal cargo, facilitated by next-generation sequencing and mass spectrometry, unveils the molecular nuances of tumors. Recent studies underscore the utility of these approaches, demonstrating their efficacy in predicting relapse, guiding therapeutic decisions, and ultimately improving patient outcomes. As the field continues to evolve, liquid biopsies hold promise not only as diagnostic tools but also as agents of personalized medicine, enabling precise navigation of the intricate landscape of cancer with minimally invasiveness.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Directorate of Postgraduate Studies, School of Medicine, Muhimbili University of Health and Allied Sciences, Ilala District, Dar es Salaam, Tanzania
- Ocean Road Cancer Institute, Ilala District, Dar es Salaam, Tanzania
| | - Chen-Chih Chung
- Department of Neurology, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, 110, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University - Shuang Ho Hospital, New Taipei City, 235, Taiwan
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD, 21218, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Oncology - Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
8
|
Myers RE, Hallman MH, Shimada A, DiCarlo M, Davis K, Leach WT, Jackson H, Indictor A, Chambers CV. Primary Care Patient Interest in Multi-Cancer Early Detection for Cancer Screening. J Pers Med 2023; 13:1613. [PMID: 38003928 PMCID: PMC10671850 DOI: 10.3390/jpm13111613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/27/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Multi-cancer early detection (MCED) tests are being developed, but little is known about patient receptivity to their use for cancer screening. The current study assessed patient interest in such testing. Our team conducted a prospective, observational study among primary care patients in a large, urban health system. They were asked to complete a telephone survey that briefly described a new blood test in development to identify multiple types of cancer, but was not currently recommended or covered by insurance. The survey included items to assess respondent background characteristics, perceptions about MCED testing, and interest in having such an MCED test. We also used multivariable analyses to identify factors associated with patient interest in test use. In 2023, we surveyed 159 (32%) of 500 identified patients. Among respondents, 125 (79%) reported a high level of interest in having an MCED test. Interest was not associated with personal background characteristics, but was positively associated with the following expectations: testing would be recommended for cancer screening, be convenient, and be effective in finding early-stage disease (OR = 11.70, 95% CI: 4.02, 34.04, p < 0.001). Research is needed to assess patient interest and actual uptake when detailed information on testing is presented in routine care.
Collapse
Affiliation(s)
- Ronald E. Myers
- Division of Population Science, Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.); (H.J.); (A.I.)
| | - Mie H. Hallman
- Division of Population Science, Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.); (H.J.); (A.I.)
| | - Ayako Shimada
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Melissa DiCarlo
- Division of Population Science, Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.); (H.J.); (A.I.)
| | - Kaitlyn Davis
- Department of Family and Community Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (K.D.); (W.T.L.); (C.V.C.)
| | - William T. Leach
- Department of Family and Community Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (K.D.); (W.T.L.); (C.V.C.)
| | - Hattie Jackson
- Division of Population Science, Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.); (H.J.); (A.I.)
| | - Amanda Indictor
- Division of Population Science, Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.); (H.J.); (A.I.)
| | - Christopher V. Chambers
- Department of Family and Community Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (K.D.); (W.T.L.); (C.V.C.)
| |
Collapse
|