1
|
Banadaki MD, Rummel NG, Backus S, Butterfield DA, St Clair DK, Campbell JM, Zhong W, Mayer K, Berry SM, Chaiswing L. Extraction of redox extracellular vesicles using exclusion-based sample preparation. Anal Bioanal Chem 2024; 416:6317-6331. [PMID: 39243301 DOI: 10.1007/s00216-024-05518-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Studying specific subpopulations of cancer-derived extracellular vesicles (EVs) could help reveal their role in cancer progression. In cancer, an increase in reactive oxygen species (ROS) happens which results in lipid peroxidation with a major product of 4-hydroxynonenal (HNE). Adduction by HNE causes alteration to the structure of proteins, leading to loss of function. Blebbing of EVs carrying these HNE-adducted proteins as a cargo or carrying HNE-adducted on EV membrane are methods for clearing these molecules by the cells. We have referred to these EVs as Redox EVs. Here, we utilize a surface tension-mediated extraction process, termed exclusion-based sample preparation (ESP), for the rapid and efficient isolation of intact Redox EVs, from a mixed population of EVs derived from human glioblastoma cell line LN18. After optimizing different parameters, two populations of EVs were analyzed, those isolated from the sample (Redox EVs) and those remaining in the original sample (Remaining EVs). Electron microscopic imaging was used to confirm the presence of HNE adducts on the outer leaflet of Redox EVs. Moreover, the population of HNE-adducted Redox EVs shows significantly different characteristics to those of Remaining EVs including smaller size EVs and a more negative zeta potential EVs. We further treated glioblastoma cells (LN18), radiation-resistant glioblastoma cells (RR-LN18), and normal human astrocytes (NHA) with both Remaining and Redox EV populations. Our results indicate that Redox EVs promote the growth of glioblastoma cells, likely through the production of H2O2, and cause injury to normal astrocytes. In contrast, Remaining EVs have minimal impact on the viability of both glioblastoma cells and NHA cells. Thus, isolating a subpopulation of EVs employing ESP-based immunoaffinity could pave the way for a deeper mechanistic understanding of how subtypes of EVs, such as those containing HNE-adducted proteins, induce biological changes in the cells that take up these EVs.
Collapse
Affiliation(s)
| | - Nicole G Rummel
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Spencer Backus
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, 40506, USA
| | - David Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Daret K St Clair
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - James M Campbell
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kristy Mayer
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Scott M Berry
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, 40506, USA.
| | - Luksana Chaiswing
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
2
|
Le MCN, Smith KA, Dopico PJ, Greer B, Alipanah M, Zhang Y, Siemann DW, Lagmay JP, Fan ZH. Investigating surface proteins and antibody combinations for detecting circulating tumor cells of various sarcomas. Sci Rep 2024; 14:12374. [PMID: 38811642 PMCID: PMC11137101 DOI: 10.1038/s41598-024-61651-w] [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: 01/21/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
Circulating tumor cells (CTCs) have gathered attention as a biomarker for carcinomas. However, CTCs in sarcomas have received little attention. In this work, we investigated cell surface proteins and antibody combinations for immunofluorescence detection of sarcoma CTCs. A microfluidic device that combines filtration and immunoaffinity using gangliosides 2 and cell surface vimentin (CSV) antibodies was employed to capture CTCs. For CTC detection, antibodies against cytokeratins 7 and 8 (CK), pan-cytokeratin (panCK), or a combination of panCK and CSV were used. Thirty-nine blood samples were collected from 21 patients of various sarcoma subtypes. In the independent samples study, samples were subjected to one of three antibody combination choices. Significant difference in CTC enumeration was found between CK and panCK + CSV, and between panCK and panCK + CSV. Upon stratification of CK+ samples, those of metastatic disease had a higher CTC number than those of localized disease. In the paired samples study involving cytokeratin-positive sarcoma subtypes, using panCK antibody detected more CTCs than CK. Similarly, for osteosarcoma, using panCK + CSV combination resulted in a higher CTC count than panCK. This study emphasized deliberate selection of cell surface proteins for sarcoma CTC detection and subtype stratification for studying cancers as heterogeneous as sarcomas.
Collapse
Affiliation(s)
- Minh-Chau N Le
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Kierstin A Smith
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Pablo J Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Beate Greer
- Department of Pediatrics, Division of Hematology-Oncology, University of Florida, Gainesville, FL, 32610, USA
| | - Morteza Alipanah
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Yang Zhang
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, 32610, USA
| | - Joanne P Lagmay
- Department of Pediatrics, Division of Hematology-Oncology, University of Florida, Gainesville, FL, 32610, USA.
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, PO Box 116250, Gainesville, FL, 32611, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
3
|
Guo X, Hu F, Yong Z, Zhao S, Wan Y, Wang B, Peng N. Magnetic Nanoparticle-Based Microfluidic Platform for Automated Enrichment of High-Purity Extracellular Vesicles. Anal Chem 2024; 96:7212-7219. [PMID: 38660946 DOI: 10.1021/acs.analchem.4c00795] [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: 04/26/2024]
Abstract
Extracellular vesicles (EVs) are available in various biological fluids and have highly heterogeneous sizes, origins, contents, and functions. Rapid enrichment of high-purity EVs remains crucial for enhancing research on EVs in tumors. In this work, we present a magnetic nanoparticle-based microfluidic platform (ExoCPR) for on-chip isolation, purification, and mild recovery of EVs from cell culture supernatant and plasma within 29 min. The ExoCPR chip integrates bubble-driven micromixers and immiscible filtration assisted by surface tension (IFAST) technology. The bubble-driven micromixer enhances the mixing between immunomagnetic beads and EVs, eliminating the need for manual pipetting or off-chip oscillatory incubation. The high-purity EVs were obtained after passing through the immiscible phase interface where hydrophilic or hydrophobic impurities nonspecifically bound to SIMI were removed. The ExoCPR chip had a capture efficiency of 75.8% and a release efficiency of 62.7% for model EVs. We also demonstrated the powerful performance of the ExoCPR in isolating EVs from biological samples (>90% purity). This chip was further employed in clinical plasma samples and showed that the number of GPC3-positive EVs isolated from hepatocellular carcinoma patients was significantly higher than that of healthy individuals. This ExoCPR chip may provide a promising tool for EV-based liquid biopsy and other fundamental research.
Collapse
Affiliation(s)
- Xiaoniu Guo
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Zhang Yong
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Shuhao Zhao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Yong Wan
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Bingqing Wang
- Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| | - Niancai Peng
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| |
Collapse
|
4
|
Crocetto F, Falcone A, Mirto BF, Sicignano E, Pagano G, Dinacci F, Varriale D, Machiella F, Giampaglia G, Calogero A, Varlese F, Balsamo R, Trama F, Sciarra A, Del Giudice F, Busetto GM, Ferro M, Lucarelli G, Lasorsa F, Imbimbo C, Barone B. Unlocking Precision Medicine: Liquid Biopsy Advancements in Renal Cancer Detection and Monitoring. Int J Mol Sci 2024; 25:3867. [PMID: 38612677 PMCID: PMC11011885 DOI: 10.3390/ijms25073867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Renal cell carcinoma (RCC) remains a formidable diagnostic challenge, especially in the context of small renal masses. The quest for non-invasive screening tools and biomarkers has steered research towards liquid biopsy, focusing on microRNAs (miRNAs), exosomes, and circulating tumor cells (CTCs). MiRNAs, small non-coding RNAs, exhibit notable dysregulation in RCC, offering promising avenues for diagnosis and prognosis. Studies underscore their potential across various biofluids, including plasma, serum, and urine, for RCC detection and subtype characterization. Encouraging miRNA signatures show correlations with overall survival, indicative of their future relevance in RCC management. Exosomes, with their diverse molecular cargo, including miRNAs, emerge as enticing biomarkers, while CTCs, emanating from primary tumors into the bloodstream, provide valuable insights into cancer progression. Despite these advancements, clinical translation necessitates further validation and standardization, encompassing larger-scale studies and robust evidence generation. Currently lacking approved diagnostic assays for renal cancer, the potential future applications of liquid biopsy in follow-up care, treatment selection, and outcome prediction in RCC patients are profound. This review aims to discuss and highlight recent advancements in liquid biopsy for RCC, exploring their strengths and weaknesses in the comprehensive management of this disease.
Collapse
Affiliation(s)
- Felice Crocetto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Alfonso Falcone
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Benito Fabio Mirto
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Enrico Sicignano
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Giovanni Pagano
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Fabrizio Dinacci
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Domenico Varriale
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Fabio Machiella
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Gaetano Giampaglia
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Armando Calogero
- Department of Advanced Biomedical Science, University of Naples “Federico II”, 80131 Naples, Italy; (A.C.); (F.V.)
| | - Filippo Varlese
- Department of Advanced Biomedical Science, University of Naples “Federico II”, 80131 Naples, Italy; (A.C.); (F.V.)
| | | | - Francesco Trama
- ASL Napoli 2 Nord, P.O. Santa Maria delle Grazie, 80078 Pozzuoli, Italy;
| | - Antonella Sciarra
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Francesco Del Giudice
- Department of Maternal Infant and Urological Sciences, Umberto I Polyclinic Hospital, Sapienza University, 00161 Rome, Italy;
| | - Gian Maria Busetto
- Department of Urology and Renal Transplantation, University of Foggia, 71122 Foggia, Italy;
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology (IEO)-IRCCS, 20141 Milan, Italy;
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy; (G.L.); (F.L.)
| | - Francesco Lasorsa
- Urology, Andrology and Kidney Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, 70124 Bari, Italy; (G.L.); (F.L.)
| | - Ciro Imbimbo
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples “Federico II”, 80131 Naples, Italy; (F.C.); (A.F.); (B.F.M.); (E.S.); (G.P.); (F.D.); (D.V.); (F.M.); (G.G.); (C.I.)
| | - Biagio Barone
- Urology Unit, Department of Surgical Sciences, AORN Sant’Anna e San Sebastiano, 81100 Caserta, Italy
| |
Collapse
|
5
|
Yu HJ, Jang E, Woo A, Han IW, Jeon HG, Linh VTN, Park SG, Jung HS, Lee MY. Cancer screening through surface-enhanced Raman spectroscopy fingerprinting analysis of urinary metabolites using surface-carbonized silver nanowires on a filter membrane. Anal Chim Acta 2024; 1292:342233. [PMID: 38309850 DOI: 10.1016/j.aca.2024.342233] [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: 08/04/2023] [Revised: 11/05/2023] [Accepted: 01/09/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Label-free surface-enhanced Raman spectroscopy (SERS)-based metabolic profiling has great potential for early cancer diagnosis, but further advancements in analytical methods and clinical evidence studies are required for clinical applications. To improve the cancer diagnostic accuracy of label-free SERS spectral analysis of complex biological fluids, it is necessary to obtain specifically enhanced SERS signals of cancer-related metabolites present at low concentrations. RESULTS This study presents a novel 3D SERS sensor, comprising a surface-carbonized silver nanowire (AgNW)-stacked filter membrane, alongside an optimized urine/methanol/chloroform extraction technique, which specifically changes the molecular adsorption and orientation of aromatic metabolites onto SERS substrates. By analyzing the pretreated urine samples on the surface-carbonized AgNW 3D SERS sensor, distinct and highly enhanced SERS peaks derived from semi-polar aromatic metabolites were observed for pancreatic cancer and prostate cancer samples compared with normal controls. Urine metabolite analysis using SERS fingerprinting successfully differentiated pancreatic cancer and prostate cancer groups from normal control group: normal control (n = 56), pancreatic cancer (n = 40), and prostate cancer (n = 39). SIGNIFICANCE AND NOVELTY We confirmed the clinical feasibility of performing fingerprint analysis of urinary metabolites based on the surface-carbonized AgNW 3D SERS sensor and methanol/chloroform extraction for noninvasive cancer screening. This technology holds potential for large-scale screening owing to its high accuracy, and cost effective, simple and rapid detection method.
Collapse
Affiliation(s)
- Ho-Jae Yu
- Medical Device Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - Eunji Jang
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Ayoung Woo
- Medical Device Research Center, Samsung Medical Center, Seoul, 06351, Republic of Korea
| | - In Woong Han
- Division of Hepato Biliary Pancreatic Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Vo Thi Nhat Linh
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Sung-Gyu Park
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
| | - Ho Sang Jung
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea.
| | - Min-Young Lee
- Nano-Bio Convergence Department, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea.
| |
Collapse
|
6
|
Zieren RC, Zondervan PJ, Pienta KJ, Bex A, de Reijke TM, Bins AD. Diagnostic liquid biopsy biomarkers in renal cell cancer. Nat Rev Urol 2024; 21:133-157. [PMID: 37758847 DOI: 10.1038/s41585-023-00818-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
The clinical presentation of renal cell cancer (RCC) is shifting towards incidental and early detection, creating new challenges in RCC diagnosis. Overtreatment might be reduced with the development of new diagnostic biomarkers to distinguish benign from malignant small renal masses (SRMs). Differently from tissue biopsies, liquid biopsies are obtained from a patient's blood or urine and, therefore, are minimally invasive and suitable for longitudinal monitoring. The most promising types of liquid biopsy biomarkers for RCC diagnosis are circulating tumour cells, extracellular vesicles (EVs) and cell-free DNA. Circulating tumour cell assays have the highest specificity, with low processing time and costs. However, the biological characteristics and low sensitivity limit the use of these markers in SRM diagnostics. Cell-free DNA might complement the diagnosis of high-volume RCC, but the potential for clinical application in SRMs is limited. EVs have the highest biological abundance and the highest sensitivity in identifying low-volume disease; moreover, the molecular characteristics of these markers make EVs suitable for multiple analytical applications. Thus, currently, EV assays have the greatest potential for diagnostic application in RCC (including identification of SRMs). All these liquid biomarkers have potential in clinical practice, pending validation studies. Biomarker implementation will be needed to also improve characterization of RCC subtypes. Last, diagnostic biomarkers might be extended to prognostic or predictive applications.
Collapse
Affiliation(s)
- Richard C Zieren
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Patricia J Zondervan
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, United Kingdom
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Theo M de Reijke
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan D Bins
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Bae SY, Kamalanathan KJ, Galeano-Garces C, Konety BR, Antonarakis ES, Parthasarathy J, Hong J, Drake JM. Dissemination of Circulating Tumor Cells in Breast and Prostate Cancer: Implications for Early Detection. Endocrinology 2024; 165:bqae022. [PMID: 38366552 PMCID: PMC10904107 DOI: 10.1210/endocr/bqae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Burgeoning evidence suggests that circulating tumor cells (CTCs) may disseminate into blood vessels at an early stage, seeding metastases in various cancers such as breast and prostate cancer. Simultaneously, the early-stage CTCs that settle in metastatic sites [termed disseminated tumor cells (DTCs)] can enter dormancy, marking a potential source of late recurrence and therapy resistance. Thus, the presence of these early CTCs poses risks to patients but also holds potential benefits for early detection and treatment and opportunities for possibly curative interventions. This review delves into the role of early DTCs in driving latent metastasis within breast and prostate cancer, emphasizing the importance of early CTC detection in these diseases. We further explore the correlation between early CTC detection and poor prognoses, which contribute significantly to increased cancer mortality. Consequently, the detection of CTCs at an early stage emerges as a critical imperative for enhancing clinical diagnostics and allowing for early interventions.
Collapse
Affiliation(s)
| | | | | | - Badrinath R Konety
- Astrin Biosciences, St. Paul, MN 55114, USA
- Allina Health Cancer Institute, Minneapolis, MN 55407, USA
- Department of Urology, University of Minnesota, Minneapolis, MN 55454, USA
| | - Emmanuel S Antonarakis
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Jiarong Hong
- Astrin Biosciences, St. Paul, MN 55114, USA
- Department of Mechanical Engineering and St. Anthony Falls Laboratory, University of Minnesota, Minneapolis, MN 55414, USA
| | - Justin M Drake
- Astrin Biosciences, St. Paul, MN 55114, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Alqualo NO, Campos-Fernandez E, Picolo BU, Ferreira EL, Henriques LM, Lorenti S, Moreira DC, Simião MPS, Oliveira LBT, Alonso-Goulart V. Molecular biomarkers in prostate cancer tumorigenesis and clinical relevance. Crit Rev Oncol Hematol 2024; 194:104232. [PMID: 38101717 DOI: 10.1016/j.critrevonc.2023.104232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Prostate cancer (PCa) is the second most frequent type of cancer in men and assessing circulating tumor cells (CTCs) by liquid biopsy is a promising tool to help in cancer early detection, staging, risk of recurrence evaluation, treatment prediction and monitoring. Blood-based liquid biopsy approaches enable the enrichment, detection and characterization of CTCs by biomarker analysis. Hence, comprehending the molecular markers, their role on each stage of cancer development and progression is essential to provide information that can help in future implementation of these biomarkers in clinical assistance. In this review, we studied the molecular markers most associated with PCa CTCs to better understand their function on tumorigenesis and metastatic cascade, the methodologies utilized to analyze these biomarkers and their clinical significance, in order to summarize the available information to guide researchers in their investigations, new hypothesis formulation and target choice for the development of new diagnostic and treatment tools.
Collapse
Affiliation(s)
- Nathalia Oliveira Alqualo
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Esther Campos-Fernandez
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Bianca Uliana Picolo
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Emanuelle Lorrayne Ferreira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Laila Machado Henriques
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Sabrina Lorenti
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Danilo Caixeta Moreira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Maria Paula Silva Simião
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Luciana Beatriz Tiago Oliveira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Vivian Alonso-Goulart
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil.
| |
Collapse
|
9
|
Rodriguez-Mateos P, Ngamsom B, Ameyo D, Wakaba P, Shiluli C, Iles A, Gitaka J, Pamme N. Integrated microscale immiscible phase extraction and isothermal amplification for colorimetric detection of Neisseria gonorrhoeae. Anal Bioanal Chem 2023; 415:5129-5137. [PMID: 37198361 PMCID: PMC10191819 DOI: 10.1007/s00216-023-04734-3] [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] [Received: 01/31/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
Gonorrhea is the second most common sexually transmitted infection (STI) with around 87 million cases worldwide estimated in 2016 by the World Health Organization. With over half of the cases being asymptomatic, potential life-threatening complications and increasing numbers of drug-resistant strains, routine monitoring of prevalence and incidence of infections are key preventive measures. Whilst gold standard qPCR tests have excellent accuracy, they are neither affordable nor accessible in low-resource settings. In this study, we developed a lab-on-a-chip platform based on microscale immiscible filtration to extract, concentrate and purify Neisseria gonorrhoeae DNA with an integrated detection assay based on colorimetric isothermal amplification. The platform was capable of detecting as low as 500 copies/mL from spiked synthetic urine and showed no cross-reactivity when challenged with DNAs from other common STIs. The credit card-size device allows DNA extraction and purification without power or centrifuges, and the detection reaction only needs a low-tech block heater, providing a straightforward and visual positive/negative result within 1 h. These advantages offer great potential for accurate, affordable and accessible monitoring of gonorrhea infection in resource-poor settings.
Collapse
Affiliation(s)
- Pablo Rodriguez-Mateos
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, Sweden
| | - Bongkot Ngamsom
- Centre for Biomedicine, Hull York Medical School, University of Hull, Hull, UK
| | - Daglus Ameyo
- Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Patrick Wakaba
- Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Clement Shiluli
- Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Alexander Iles
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, Sweden
| | - Jesse Gitaka
- Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya.
| | - Nicole Pamme
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, Sweden.
- School of Natural Sciences, University of Hull, Hull, UK.
| |
Collapse
|
10
|
Sperger JM, Helzer KT, Stahlfeld CN, Jiang D, Singh A, Kaufmann KR, Niles DJ, Heninger E, Rydzewski NR, Wang L, Wang L, Yang R, Ren Y, Engle JW, Huang P, Kyriakopoulos CE, Slovin SF, Soule HR, Zhao SG, Kohli M, Tagawa ST, Cai W, Dehm SM, Lang JM. Expression and Therapeutic Targeting of TROP-2 in Treatment-Resistant Prostate Cancer. Clin Cancer Res 2023; 29:2324-2335. [PMID: 36939530 PMCID: PMC10261916 DOI: 10.1158/1078-0432.ccr-22-1305] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/29/2022] [Accepted: 03/14/2023] [Indexed: 03/21/2023]
Abstract
PURPOSE Men with metastatic castration-resistant prostate cancer (mCRPC) frequently develop resistance to androgen receptor signaling inhibitor (ARSI) treatment; therefore, new therapies are needed. Trophoblastic cell-surface antigen (TROP-2) is a transmembrane protein identified in prostate cancer and overexpressed in multiple malignancies. TROP-2 is a therapeutic target for antibody-drug conjugates (ADC). EXPERIMENTAL DESIGN TROP-2 gene (TACSTD2) expression and markers of treatment resistance from prostate biopsies were analyzed using data from four previously curated cohorts of mCRPC (n = 634) and the PROMOTE study (dbGaP accession phs001141.v1.p1, n = 88). EPCAM or TROP-2-positive circulating tumor cells (CTC) were captured from peripheral blood for comparison of protein (n = 15) and gene expression signatures of treatment resistance (n = 40). We assessed the efficacy of TROP-2-targeting agents in a mouse xenograft model generated from prostate cancer cell lines. RESULTS We demonstrated that TACSTD2 is expressed in mCRPC from luminal and basal tumors but at lower levels in patients with neuroendocrine prostate cancer. Patients previously treated with ARSI showed no significant difference in TACSTD2 expression, whereas patients with detectable AR-V7 expression showed increased expression. We observed that TROP-2 can serve as a cell surface target for isolating CTCs, which may serve as a predictive biomarker for ADCs. We also demonstrated that prostate cancer cell line xenografts can be targeted specifically by labeled anti-TROP-2 agents in vivo. CONCLUSIONS These results support further studies on TROP-2 as a therapeutic and diagnostic target for mCRPC.
Collapse
Affiliation(s)
- Jamie M. Sperger
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | - Kyle T. Helzer
- Department of Human Oncology, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Dawei Jiang
- Department of Radiology, University of Wisconsin, Madison, Wisconsin
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, China
| | - Anupama Singh
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - David J. Niles
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin
| | - Erika Heninger
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | | | | | | | - Rendong Yang
- Masonic Cancer Center and Departments of Laboratory Medicine and Pathology and Urology, University of Minnesota, Minneapolis, Minnesota
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Yanan Ren
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin
| | - Peng Huang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Laboratory of Evolutionary Theranostics, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| | - Christos E. Kyriakopoulos
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Howard R. Soule
- Department of Science, Prostate Cancer Foundation, Santa Monica, California
| | - Shuang G. Zhao
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
- Department of Human Oncology, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Scott T. Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York
| | - Weibo Cai
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
- Department of Radiology, University of Wisconsin, Madison, Wisconsin
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin
| | - Scott M. Dehm
- Masonic Cancer Center and Departments of Laboratory Medicine and Pathology and Urology, University of Minnesota, Minneapolis, Minnesota
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| |
Collapse
|
11
|
Li M, Li L, Zheng J, Li Z, Li S, Wang K, Chen X. Liquid biopsy at the frontier in renal cell carcinoma: recent analysis of techniques and clinical application. Mol Cancer 2023; 22:37. [PMID: 36810071 PMCID: PMC9942319 DOI: 10.1186/s12943-023-01745-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/11/2023] [Indexed: 02/23/2023] Open
Abstract
Renal cell carcinoma (RCC) is a major pathological type of kidney cancer and is one of the most common malignancies worldwide. The unremarkable symptoms of early stages, proneness to postoperative metastasis or recurrence, and low sensitivity to radiotherapy and chemotherapy pose a challenge for the diagnosis and treatment of RCC. Liquid biopsy is an emerging test that measures patient biomarkers, including circulating tumor cells, cell-free DNA/cell-free tumor DNA, cell-free RNA, exosomes, and tumor-derived metabolites and proteins. Owing to its non-invasiveness, liquid biopsy enables continuous and real-time collection of patient information for diagnosis, prognostic assessment, treatment monitoring, and response evaluation. Therefore, the selection of appropriate biomarkers for liquid biopsy is crucial for identifying high-risk patients, developing personalized therapeutic plans, and practicing precision medicine. In recent years, owing to the rapid development and iteration of extraction and analysis technologies, liquid biopsy has emerged as a low cost, high efficiency, and high accuracy clinical detection method. Here, we comprehensively review liquid biopsy components and their clinical applications over the past 5 years. Additionally, we discuss its limitations and predict its future prospects.
Collapse
Affiliation(s)
- Mingyang Li
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning Shenyang, 110004 People’s Republic of China
| | - Lei Li
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning Shenyang, 110004 People’s Republic of China
| | - Jianyi Zheng
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning Shenyang, 110004 People’s Republic of China
| | - Zeyu Li
- grid.412467.20000 0004 1806 3501Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning Shenyang, 110004 People’s Republic of China
| | - Shijie Li
- Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.
| | - Xiaonan Chen
- Department of Urology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Liaoning, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
12
|
Bootsma M, McKay RR, Emamekhoo H, Bade RM, Schehr JL, Mannino MC, Singh A, Wolfe SK, Schultz ZD, Sperger J, Xie W, Signoretti S, Kyriakopoulos CE, Kosoff D, Abel EJ, Helzer KT, Rydzewski N, Bakhtiar H, Shi Y, Blitzer G, Bassetti M, Floberg J, Yu M, Sethakorn N, Sharifi M, Harari PM, Choueiri TK, Lang JM, Zhao SG. Longitudinal Molecular Profiling of Circulating Tumor Cells in Metastatic Renal Cell Carcinoma. J Clin Oncol 2022; 40:3633-3641. [PMID: 35617646 PMCID: PMC9622626 DOI: 10.1200/jco.22.00219] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/29/2022] [Accepted: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Liquid biopsies in metastatic renal cell carcinoma (mRCC) provide a unique approach to understand the molecular basis of treatment response and resistance. This is particularly important in the context of immunotherapies, which target key immune-tumor interactions. Unlike metastatic tissue biopsies, serial real-time profiling of mRCC is feasible with our noninvasive circulating tumor cell (CTC) approach. METHODS We collected 457 longitudinal liquid biopsies from 104 patients with mRCC enrolled in one of two studies, either a prospective cohort or a phase II multicenter adaptive immunotherapy trial. Using a novel CTC capture and automated microscopy platform, we profiled CTC enumeration and expression of HLA I and programmed cell death-ligand 1 (PD-L1). Given their diametric immunological roles, we focused on the HLA I to PD-L1 ratio (HP ratio). RESULTS Patients with radiographic responses showed significantly lower CTC abundances throughout treatment. Furthermore, patients whose CTC enumeration trajectory was in the highest quartile (> 0.12 CTCs/mL annually) had shorter overall survival (median 17.0 months v 21.1 months, P < .001). Throughout treatment, the HP ratio decreased in patients receiving immunotherapy but not in patients receiving tyrosine kinase inhibitors. Patients with an HP ratio trajectory in the highest quartile (≥ 0.0012 annually) displayed significantly shorter overall survival (median 18.4 months v 21.2 months, P = .003). CONCLUSION In the first large longitudinal CTC study in mRCC to date to our knowledge, we identified the prognostic importance of CTC enumeration and the change over time of both CTC enumeration and the HP ratio. These insights into changes in both tumor burden and the molecular profile of tumor cells in response to different treatments provide potential biomarkers to predict and monitor response to immunotherapy in mRCC.
Collapse
Affiliation(s)
- Matthew Bootsma
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Rana R. McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Hamid Emamekhoo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Rory M. Bade
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Jennifer L. Schehr
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Matthew C. Mannino
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Anupama Singh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Serena K. Wolfe
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Zachery D. Schultz
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Jamie Sperger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | | | - Sabina Signoretti
- Dana-Farber Cancer Institute, Boston, MA
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Christos E. Kyriakopoulos
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - David Kosoff
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Edwin J. Abel
- Urology, University of Wisconsin-Madison, Madison, WI
| | - Kyle T. Helzer
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Nicholas Rydzewski
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Hamza Bakhtiar
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Yue Shi
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Grace Blitzer
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Michael Bassetti
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - John Floberg
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | - Menggang Yu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI
| | - Nan Sethakorn
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Marina Sharifi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
| | | | - Joshua M. Lang
- Department of Medicine, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Shuang G. Zhao
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
- William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
13
|
Zhao SG, Sperger JM, Schehr JL, McKay RR, Emamekhoo H, Singh A, Schultz ZD, Bade RM, Stahlfeld CN, Gilsdorf CS, Hernandez CI, Wolfe SK, Mayberry RD, Krause HM, Bootsma M, Helzer KT, Rydzewski N, Bakhtiar H, Shi Y, Blitzer G, Kyriakopoulos CE, Kosoff D, Wei XX, Floberg J, Sethakorn N, Sharifi M, Harari PM, Huang W, Beltran H, Choueiri TK, Scher HI, Rathkopf DE, Halabi S, Armstrong AJ, Beebe DJ, Yu M, Sundling KE, Taplin ME, Lang JM. A clinical-grade liquid biomarker detects neuroendocrine differentiation in prostate cancer. J Clin Invest 2022; 132:e161858. [PMID: 36317634 PMCID: PMC9621140 DOI: 10.1172/jci161858] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/23/2022] [Indexed: 11/07/2022] Open
Abstract
BackgroundNeuroendocrine prostate cancer (NEPC) is an aggressive subtype, the presence of which changes the prognosis and management of metastatic prostate cancer.MethodsWe performed analytical validation of a Circulating Tumor Cell (CTC) multiplex RNA qPCR assay to identify the limit of quantification (LOQ) in cell lines, synthetic cDNA, and patient samples. We next profiled 116 longitudinal samples from a prospectively collected institutional cohort of 17 patients with metastatic prostate cancer (7 NEPC, 10 adenocarcinoma) as well as 265 samples from 139 patients enrolled in 3 adenocarcinoma phase II trials of androgen receptor signaling inhibitors (ARSIs). We assessed a NEPC liquid biomarker via the presence of neuroendocrine markers and the absence of androgen receptor (AR) target genes.ResultsUsing the analytical validation LOQ, liquid biomarker NEPC detection in the longitudinal cohort had a per-sample sensitivity of 51.35% and a specificity of 91.14%. However, when we incorporated the serial information from multiple liquid biopsies per patient, a unique aspect of this study, the per-patient predictions were 100% accurate, with a receiver-operating-curve (ROC) AUC of 1. In the adenocarcinoma ARSI trials, the presence of neuroendocrine markers, even while AR target gene expression was retained, was a strong negative prognostic factor.ConclusionOur analytically validated CTC biomarker can detect NEPC with high diagnostic accuracy when leveraging serial samples that are only feasible using liquid biopsies. Patients with expression of NE genes while retaining AR-target gene expression may indicate the transition to neuroendocrine differentiation, with clinical characteristics consistent with this phenotype.FundingNIH (DP2 OD030734, 1UH2CA260389, R01CA247479, and P30 CA014520), Department of Defense (PC190039 and PC200334), and Prostate Cancer Foundation (Movember Foundation - PCF Challenge Award).
Collapse
Affiliation(s)
- Shuang G. Zhao
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Jamie M. Sperger
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Jennifer L. Schehr
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Rana R. McKay
- Moores Cancer Center, University of California, San Diego, La Jolla, California, USA
| | - Hamid Emamekhoo
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Anupama Singh
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Zachery D. Schultz
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Rory M. Bade
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Charlotte N. Stahlfeld
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Cole S. Gilsdorf
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Camila I. Hernandez
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Serena K. Wolfe
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Hannah M. Krause
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Matt Bootsma
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Kyle T. Helzer
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Nicholas Rydzewski
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Hamza Bakhtiar
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Yue Shi
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Grace Blitzer
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Christos E. Kyriakopoulos
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - David Kosoff
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Xiao X. Wei
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - John Floberg
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Nan Sethakorn
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Marina Sharifi
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Paul M. Harari
- Department of Human Oncology and
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Wei Huang
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Himisha Beltran
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Toni K. Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Howard I. Scher
- Genitourinary Oncology Service, Department of Medicine and
- Biomarker Development Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Susan Halabi
- Department of Biostatistics and Bioinformatics and
| | - Andrew J. Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - David J. Beebe
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Biomedical Engineering and
| | - Menggang Yu
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Kaitlin E. Sundling
- Wisconsin State Lab of Hygiene, Madison, Wisconsin, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Mary-Ellen Taplin
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Joshua M. Lang
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Rodriguez-Mateos P, Ngamsom B, Iles A, Pamme N. Microscale immiscible phase magnetic processing for bioanalytical applications. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
15
|
Rodems TS, Heninger E, Stahlfeld CN, Gilsdorf CS, Carlson KN, Kircher MR, Singh A, Krueger TEG, Beebe DJ, Jarrard DF, McNeel DG, Haffner MC, Lang JM. Reversible epigenetic alterations regulate class I HLA loss in prostate cancer. Commun Biol 2022; 5:897. [PMID: 36050516 PMCID: PMC9437063 DOI: 10.1038/s42003-022-03843-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 08/15/2022] [Indexed: 11/09/2022] Open
Abstract
Downregulation of HLA class I (HLA-I) impairs immune recognition and surveillance in prostate cancer and may underlie the ineffectiveness of checkpoint blockade. However, the molecular mechanisms regulating HLA-I loss in prostate cancer have not been fully explored. Here, we conducted a comprehensive analysis of HLA-I genomic, epigenomic and gene expression alterations in primary and metastatic human prostate cancer. Loss of HLA-I gene expression was associated with repressive chromatin states including DNA methylation, histone H3 tri-methylation at lysine 27, and reduced chromatin accessibility. Pharmacological DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibition decreased DNA methylation and increased H3 lysine 27 acetylation and resulted in re-expression of HLA-I on the surface of tumor cells. Re-expression of HLA-I on LNCaP cells by DNMT and HDAC inhibition increased activation of co-cultured prostate specific membrane antigen (PSMA)27-38-specific CD8+ T-cells. HLA-I expression is epigenetically regulated by functionally reversible DNA methylation and chromatin modifications in human prostate cancer. Methylated HLA-I was detected in HLA-Ilow circulating tumor cells (CTCs), which may serve as a minimally invasive biomarker for identifying patients who would benefit from epigenetic targeted therapies.
Collapse
Affiliation(s)
- Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Erika Heninger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Charlotte N Stahlfeld
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole S Gilsdorf
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Kristin N Carlson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Madison R Kircher
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Anupama Singh
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Timothy E G Krueger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David J Beebe
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Pathology, University of Wisconsin, Madison, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - David F Jarrard
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Urology, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Michael C Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, N., Seattle, WA, 98109, USA.,Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA.,Department of Pathology, Johns Hopkins School of Medicine, 600N Wolfe St., Baltimore, MD, 21287, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
16
|
Schehr JL, Sethakorn N, Schultz ZD, Hernandez CI, Bade RM, Eyzaguirre D, Singh A, Niles DJ, Henderson L, Warrick JW, Berry SM, Sundling KE, Beebe DJ, Leal TA, Lang JM. Analytical validation and initial clinical testing of quantitative microscopic evaluation for PD-L1 and HLA I expression on circulating tumor cells from patients with non-small cell lung cancer. Biomark Res 2022; 10:26. [PMID: 35468853 PMCID: PMC9040226 DOI: 10.1186/s40364-022-00370-8] [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: 11/24/2021] [Accepted: 03/25/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION PD-L1 expression in non-small cell lung cancer (NSCLC) predicts response to immune checkpoint blockade, however is an imperfect biomarker given tumor heterogeneity, and the antigen presentation pathway requiring other components including HLA I expression. HLA I downregulation may contribute to resistance, warranting its evaluation in attempts to guide patient selection. In addition, earlier detection of acquired resistance could prompt earlier change in treatment and prolong patient survival. Analysis of circulating tumor cells (CTCs) captures heterogeneity across multiple sites of metastases, enables detection of changes in tumor burden that precede radiographic response, and can be obtained in serial fashion. METHODS To quantify the expression of both PD-L1 and HLA I on CTCs, we developed exclusion-based sample preparation technology, achieving high-yield with gentle magnetic movement of antibody-labeled cells through virtual barriers of surface tension. To achieve clinical-grade quantification of rare cells, we employ high quality fluorescence microscopy image acquisition and automated image analysis together termed quantitative microscopy. RESULTS In preparation for clinical laboratory implementation, we demonstrate high precision and accuracy of these methodologies using a diverse set of control materials. Preliminary testing of CTCs isolated from patients with NSCLC demonstrate heterogeneity in PD-L1 and HLA I expression and promising clinical value in predicting PFS in response to PD-L1 targeted therapies. CONCLUSIONS By confirming high performance, we ensure compatibility for clinical laboratory implementation and future application to better predict and detect resistance to PD-L1 targeted therapy in patients with NSCLC.
Collapse
Affiliation(s)
| | - Nan Sethakorn
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | | | | | - Rory M Bade
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Diego Eyzaguirre
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Anupama Singh
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - David J Niles
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53705, USA
| | | | - Jay W Warrick
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53705, USA
| | - Scott M Berry
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53705, USA
| | - Kaitlin E Sundling
- Wisconsin State Lab of Hygiene, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53705, USA
| | - Ticiana A Leal
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- Department of Medicine, University of Wisconsin, Madison, WI, USA.
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, 1111 Highland Avenue, WIMR 7151, Madison, WI, 53705, USA.
| |
Collapse
|
17
|
Rodems TS, Juang DS, Stahlfeld CN, Gilsdorf CS, Krueger TEG, Heninger E, Zhao SG, Sperger JM, Beebe DJ, Haffner MC, Lang JM. SEEMLIS: a flexible semi-automated method for enrichment of methylated DNA from low-input samples. Clin Epigenetics 2022; 14:37. [PMID: 35272673 PMCID: PMC8908705 DOI: 10.1186/s13148-022-01252-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/18/2022] [Indexed: 01/02/2023] Open
Abstract
Background DNA methylation alterations have emerged as hallmarks of cancer and have been proposed as screening, prognostic, and predictive biomarkers. Traditional approaches for methylation analysis have relied on bisulfite conversion of DNA, which can damage DNA and is not suitable for targeted gene analysis in low-input samples. Here, we have adapted methyl-CpG-binding domain protein 2 (MBD2)-based DNA enrichment for use on a semi-automated exclusion-based sample preparation (ESP) platform for robust and scalable enrichment of methylated DNA from low-input samples, called SEEMLIS. Results We show that combining methylation-sensitive enzyme digestion with ESP-based MBD2 enrichment allows for single gene analysis with high sensitivity for GSTP1 in highly impure, heterogenous samples. We also show that ESP-based MBD2 enrichment coupled with targeted pre-amplification allows for analysis of multiple genes with sensitivities approaching the single cell level in pure samples for GSTP1 and RASSF1 and sensitivity down to 14 cells for these genes in highly impure samples. Finally, we demonstrate the potential clinical utility of SEEMLIS by successful detection of methylated gene signatures in circulating tumor cells (CTCs) from patients with prostate cancer with varying CTC number and sample purity. Conclusions SEEMLIS is a robust assay for targeted DNA methylation analysis in low-input samples, with flexibility at multiple steps. We demonstrate the feasibility of this assay to analyze DNA methylation in prostate cancer cells using CTCs from patients with prostate cancer as a real-world example of a low-input analyte of clinical importance. In summary, this novel assay provides a platform for determining methylation signatures in rare cell populations with broad implications for research as well as clinical applications. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01252-4.
Collapse
Affiliation(s)
- Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Duane S Juang
- Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - Charlotte N Stahlfeld
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole S Gilsdorf
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Tim E G Krueger
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Erika Heninger
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Jamie M Sperger
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David J Beebe
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Pathology, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Michael C Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, N., Seattle, WA, 98109, USA.,Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA.,Department of Pathology, Johns Hopkins School of Medicine, 600 N Wolfe St., Baltimore, MD, 21287, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,7151 WI Institutes for Medical Research, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
18
|
Morrison G, Buckley J, Ostrow D, Varghese B, Cen SY, Werbin J, Ericson N, Cunha A, Lu YT, George T, Smith J, Quinn D, Duddalwar V, Triche T, Goldkorn A. Non-Invasive Profiling of Advanced Prostate Cancer via Multi-Parametric Liquid Biopsy and Radiomic Analysis. Int J Mol Sci 2022; 23:2571. [PMID: 35269713 PMCID: PMC8910093 DOI: 10.3390/ijms23052571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/16/2022] Open
Abstract
Integrating liquid biopsies of circulating tumor cells (CTCs) and cell-free DNA (cfDNA) with other minimally invasive measures may yield more comprehensive disease profiles. We evaluated the feasibility of concurrent cellular and molecular analysis of CTCs and cfDNA combined with radiomic analysis of CT scans from patients with metastatic castration-resistant PC (mCRPC). CTCs from 22 patients were enumerated, stained for PC-relevant markers, and clustered based on morphometric and immunofluorescent features using machine learning. DNA from single CTCs, matched cfDNA, and buffy coats was sequenced using a targeted amplicon cancer hotspot panel. Radiomic analysis was performed on bone metastases identified on CT scans from the same patients. CTCs were detected in 77% of patients and clustered reproducibly. cfDNA sequencing had high sensitivity (98.8%) for germline variants compared to WBC. Shared and unique somatic variants in PC-related genes were detected in cfDNA in 45% of patients (MAF > 0.1%) and in CTCs in 92% of patients (MAF > 10%). Radiomic analysis identified a signature that strongly correlated with CTC count and plasma cfDNA level. Integration of cellular, molecular, and radiomic data in a multi-parametric approach is feasible, yielding complementary profiles that may enable more comprehensive non-invasive disease modeling and prediction.
Collapse
Affiliation(s)
- Gareth Morrison
- Division of Medical Oncology, Department of Medicine and Department of Biochemistry & Molecular Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; (G.M.); (A.C.); (Y.-T.L.); (D.Q.)
| | - Jonathan Buckley
- Department of Population and Public Health Sciences, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA;
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (D.O.); (T.T.)
| | - Dejerianne Ostrow
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (D.O.); (T.T.)
| | - Bino Varghese
- Department of Radiology, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA;
| | - Steven Y. Cen
- Departments of Radiology and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA;
| | - Jeffrey Werbin
- RareCyte, Inc., Seattle, WA 98121, USA; (J.W.); (N.E.); (T.G.)
| | - Nolan Ericson
- RareCyte, Inc., Seattle, WA 98121, USA; (J.W.); (N.E.); (T.G.)
| | - Alexander Cunha
- Division of Medical Oncology, Department of Medicine and Department of Biochemistry & Molecular Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; (G.M.); (A.C.); (Y.-T.L.); (D.Q.)
| | - Yi-Tsung Lu
- Division of Medical Oncology, Department of Medicine and Department of Biochemistry & Molecular Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; (G.M.); (A.C.); (Y.-T.L.); (D.Q.)
| | - Thaddeus George
- RareCyte, Inc., Seattle, WA 98121, USA; (J.W.); (N.E.); (T.G.)
| | - Jeffrey Smith
- Clinical Sequencing Division, Thermo Fisher Scientific, San Francisco, CA 94080, USA;
| | - David Quinn
- Division of Medical Oncology, Department of Medicine and Department of Biochemistry & Molecular Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; (G.M.); (A.C.); (Y.-T.L.); (D.Q.)
| | - Vinay Duddalwar
- Departments of Radiology and Urology, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA;
| | - Timothy Triche
- Center for Personalized Medicine, Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA; (D.O.); (T.T.)
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine and Department of Biochemistry & Molecular Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; (G.M.); (A.C.); (Y.-T.L.); (D.Q.)
| |
Collapse
|
19
|
Fakhraldeen SA, Berry SM, Beebe DJ, Roopra A, Bisbach CM, Spiegelman VS, Niemi NM, Alexander CM. Enhanced immunoprecipitation techniques for the identification of RNA-binding protein partners: IGF2BP1 interactions in mammary epithelial cells. J Biol Chem 2022; 298:101649. [PMID: 35104504 PMCID: PMC8891971 DOI: 10.1016/j.jbc.2022.101649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 11/24/2022] Open
Abstract
RNA-binding proteins (RBPs) regulate the expression of large cohorts of RNA species to produce programmatic changes in cellular phenotypes. To describe the function of RBPs within a cell, it is key to identify their mRNA-binding partners. This is often done by crosslinking nucleic acids to RBPs, followed by chemical release of the nucleic acid fragments for analysis. However, this methodology is lengthy, which involves complex processing with attendant sample losses, thus large amounts of starting materials and prone to artifacts. To evaluate potential alternative technologies, we tested “exclusion-based” purification of immunoprecipitates (IFAST or SLIDE) and report here that these methods can efficiently, rapidly, and specifically isolate RBP–RNA complexes. The analysis requires less than 1% of the starting material required for techniques that include crosslinking. Depending on the antibody used, 50% to 100% starting protein can be retrieved, facilitating the assay of endogenous levels of RBPs; the isolated ribonucleoproteins are subsequently analyzed using standard techniques, to provide a comprehensive portrait of RBP complexes. Using exclusion-based techniques, we show that the mRNA-binding partners for RBP IGF2BP1 in cultured mammary epithelial cells are enriched in mRNAs important for detoxifying superoxides (specifically glutathione peroxidase [GPX]-1 and GPX-2) and mRNAs encoding mitochondrial proteins. We show that these interactions are functionally significant, as loss of function of IGF2BP1 leads to destabilization of GPX mRNAs and reduces mitochondrial membrane potential and oxygen consumption. We speculate that this underlies a consistent requirement for IGF2BP1 for the expression of clonogenic activity in vitro.
Collapse
Affiliation(s)
- Saja A Fakhraldeen
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Scott M Berry
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Celia M Bisbach
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vladimir S Spiegelman
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University in St Louis
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
20
|
Bos MK, Kraan J, Sleijfer S, Martens JWM, Beije N. Prognostic Value of Circulating Tumor Cell Characteristics May Be Biased by Their Quantity. J Clin Oncol 2021; 40:519-520. [PMID: 34878805 DOI: 10.1200/jco.21.01959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Manouk K Bos
- Manouk K. Bos, MD, Jaco Kraan, PhD, Stefan Sleijfer, MD, PhD, John W. M. Martens, PhD, and Nick Beije, MD, PhD, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jaco Kraan
- Manouk K. Bos, MD, Jaco Kraan, PhD, Stefan Sleijfer, MD, PhD, John W. M. Martens, PhD, and Nick Beije, MD, PhD, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Manouk K. Bos, MD, Jaco Kraan, PhD, Stefan Sleijfer, MD, PhD, John W. M. Martens, PhD, and Nick Beije, MD, PhD, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John W M Martens
- Manouk K. Bos, MD, Jaco Kraan, PhD, Stefan Sleijfer, MD, PhD, John W. M. Martens, PhD, and Nick Beije, MD, PhD, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Nick Beije
- Manouk K. Bos, MD, Jaco Kraan, PhD, Stefan Sleijfer, MD, PhD, John W. M. Martens, PhD, and Nick Beije, MD, PhD, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
21
|
Sperger JM, Feng FY, Armstrong AJ, Zhao SG, Lang JM. Reply to M. K. Bos et al. J Clin Oncol 2021; 40:520-522. [PMID: 34878806 DOI: 10.1200/jco.21.02238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jamie M Sperger
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Felix Y Feng
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Andrew J Armstrong
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Shuang G Zhao
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Joshua M Lang
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| |
Collapse
|
22
|
Kim EJ, Liu P, Zhang S, Donahue K, Wang Y, Schehr J, Wolfe S, Dickerson A, Lu L, Rui L, Zhong X, Wisinski K, Yu M, Suzuki A, Lang J, Ong I, Xu W. BAF155 methylation drives metastasis by hijacking super-enhancers and subverting anti-tumor immunity. Nucleic Acids Res 2021; 49:12211-12233. [PMID: 34865122 PMCID: PMC8643633 DOI: 10.1093/nar/gkab1122] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022] Open
Abstract
Subunits of the chromatin remodeler SWI/SNF are the most frequently disrupted genes in cancer. However, how post-translational modifications (PTM) of SWI/SNF subunits elicit epigenetic dysfunction remains unknown. Arginine-methylation of BAF155 by coactivator-associated arginine methyltransferase 1 (CARM1) promotes triple-negative breast cancer (TNBC) metastasis. Herein, we discovered the dual roles of methylated-BAF155 (me-BAF155) in promoting tumor metastasis: activation of super-enhancer-addicted oncogenes by recruiting BRD4, and repression of interferon α/γ pathway genes to suppress host immune response. Pharmacological inhibition of CARM1 and BAF155 methylation not only abrogated the expression of an array of oncogenes, but also boosted host immune responses by enhancing the activity and tumor infiltration of cytotoxic T cells. Moreover, strong me-BAF155 staining was detected in circulating tumor cells from metastatic cancer patients. Despite low cytotoxicity, CARM1 inhibitors strongly inhibited TNBC cell migration in vitro, and growth and metastasis in vivo. These findings illustrate a unique mechanism of arginine methylation of a SWI/SNF subunit that drives epigenetic dysregulation, and establishes me-BAF155 as a therapeutic target to enhance immunotherapy efficacy.
Collapse
Affiliation(s)
- Eui-Jun Kim
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Peng Liu
- Department of Biostatistics and Medical Informatics. School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Shengjie Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kristine Donahue
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Yidan Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer L Schehr
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Serena K Wolfe
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Amber Dickerson
- Department of Stem Cell Biology and Regenerative Medicine, and USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Li Lu
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison WI, USA
| | - Lixin Rui
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Xuehua Zhong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison WI, USA
| | - Kari B Wisinski
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, and USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Irene M Ong
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Department of Biostatistics and Medical Informatics. School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
23
|
Bade RM, Schehr JL, Emamekhoo H, Gibbs BK, Rodems TS, Mannino MC, Desotelle JA, Heninger E, Stahlfeld CN, Sperger JM, Singh A, Wolfe SK, Niles DJ, Arafat W, Steinharter JA, Jason Abel E, Beebe DJ, Wei XX, McKay RR, Choueri TK, Lang JM. Development and initial clinical testing of a multiplexed circulating tumor cell assay in patients with clear cell renal cell carcinoma. Mol Oncol 2021; 15:2330-2344. [PMID: 33604999 PMCID: PMC8410529 DOI: 10.1002/1878-0261.12931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/31/2020] [Accepted: 02/07/2021] [Indexed: 12/21/2022] Open
Abstract
Although therapeutic options for patients with advanced renal cell carcinoma (RCC) have increased in the past decade, no biomarkers are yet available for patient stratification or evaluation of therapy resistance. Given the dynamic and heterogeneous nature of clear cell RCC (ccRCC), tumor biopsies provide limited clinical utility, but liquid biopsies could overcome these limitations. Prior liquid biopsy approaches have lacked clinically relevant detection rates for patients with ccRCC. This study employed ccRCC-specific markers, CAIX and CAXII, to identify circulating tumor cells (CTC) from patients with metastatic ccRCC. Distinct subtypes of ccRCC CTCs were evaluated for PD-L1 and HLA-I expression and correlated with patient response to therapy. CTC enumeration and expression of PD-L1 and HLA-I correlated with disease progression and treatment response, respectively. Longitudinal evaluation of a subset of patients demonstrated potential for CTC enumeration to serve as a pharmacodynamic biomarker. Further evaluation of phenotypic heterogeneity among CTCs is needed to better understand the clinical utility of this new biomarker.
Collapse
Affiliation(s)
- Rory M. Bade
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
| | | | | | | | | | | | | | - Erika Heninger
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
| | | | - Jamie M. Sperger
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
- Department of MedicineUniversity of Wisconsin‐MadisonWIUSA
| | - Anupama Singh
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
| | | | - David J. Niles
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonWIUSA
| | - Waddah Arafat
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
- Department of MedicineUniversity of Wisconsin‐MadisonWIUSA
| | - John A. Steinharter
- Lank Center for Genitourinary OncologyDana‐Farber Cancer InstituteHarvard UniversityBostonMAUSA
| | - E. Jason Abel
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
- Department of MedicineUniversity of Wisconsin‐MadisonWIUSA
| | - David J. Beebe
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonWIUSA
| | - Xiao X. Wei
- Lank Center for Genitourinary OncologyDana‐Farber Cancer InstituteHarvard UniversityBostonMAUSA
| | - Rana R. McKay
- Lank Center for Genitourinary OncologyDana‐Farber Cancer InstituteHarvard UniversityBostonMAUSA
- Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Toni K. Choueri
- Lank Center for Genitourinary OncologyDana‐Farber Cancer InstituteHarvard UniversityBostonMAUSA
| | - Joshua M. Lang
- Carbone Cancer CenterUniversity of Wisconsin‐MadisonWIUSA
- Department of MedicineUniversity of Wisconsin‐MadisonWIUSA
| |
Collapse
|
24
|
Daniel M, Knutson TP, Sperger JM, Li Y, Singh A, Stahlfeld CN, Passow C, Auch B, Lang JM, Dehm SM. AR gene rearrangement analysis in liquid biopsies reveals heterogeneity in lethal prostate cancer. Endocr Relat Cancer 2021; 28:645-655. [PMID: 34280123 PMCID: PMC8363559 DOI: 10.1530/erc-21-0157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 01/01/2023]
Abstract
Castration-resistant prostate cancer (CRPC) is driven by AR gene aberrations that arise during androgen receptor (AR)-targeted therapy. AR amplification and mutations have been profiled in circulating tumor cells (CTCs), but whether AR gene rearrangements can be assessed in CTCs is unknown. In this study, we leveraged CRPC cell lines with defined AR gene rearrangements to develop and validate a CTC DNA analysis approach that utilized whole genome amplification and targeted DNA-sequencing of AR and other genes important in CRPC. We tested the utility of this approach by analyzing matched CTC DNA and plasma cell-free DNA (cfDNA) from a case series of ten CRPC patients. One of ten CTC samples and two of ten cfDNA samples were positive for AR gene rearrangements. All AR gene rearrangements were discordant between matched liquid biopsy samples. One patient harbored separate AR gene rearrangements in CTC DNA and cfDNA, but concordant AR amplification and AR T878A mutation. This patient also displayed concordant loss of TP53 and PTEN, but the loss of RB1 in cfDNA only. The overall frequency of discordant alterations in these genes between matched CTC DNA and cfDNA was high. This study establishes the technical feasibility of analyzing structural rearrangements, mutations, and copy number variants in AR and other CRPC genes using two different sources of DNA from a single blood sample. Paired CTC DNA and cfDNA analysis may have utility for capturing the heterogeneity of genetic alterations in CRPC patients.
Collapse
Affiliation(s)
- Mark Daniel
- Masonic Cancer Center, University of Minnesota; Minneapolis, MN, USA
- Graduate Program in Microbiology, Immunology, and Cancer Biology, University of Minnesota; Minneapolis, MN, USA
| | - Todd P. Knutson
- University of Minnesota Supercomputing Institute, University of Minnesota; Minneapolis, MN, USA
| | - Jamie M. Sperger
- Department of Medicine, University of Wisconsin-Madison; Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison; Madison, WI, USA
| | - Yingming Li
- Masonic Cancer Center, University of Minnesota; Minneapolis, MN, USA
| | - Anupama Singh
- Carbone Cancer Center, University of Wisconsin-Madison; Madison, WI, USA
| | - Charlotte N. Stahlfeld
- Department of Medicine, University of Wisconsin-Madison; Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison; Madison, WI, USA
| | - Courtney Passow
- University of Minnesota Genomics Center, University of Minnesota; Minneapolis, MN, USA
| | - Benjamin Auch
- University of Minnesota Genomics Center, University of Minnesota; Minneapolis, MN, USA
| | - Joshua M. Lang
- Department of Medicine, University of Wisconsin-Madison; Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison; Madison, WI, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota; Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota; Minneapolis, MN, USA
- Department of Urology, University of Minnesota; Minneapolis, MN, USA
- Corresponding Author: Scott M. Dehm, MCRB 560D, Mayo Mail Code 806, 420 Delaware St. SE, Minneapolis, MN 55455. Tel: (612) 625-1504. Fax: (612) 626-4915.
| |
Collapse
|
25
|
Sperger JM, Emamekhoo H, McKay RR, Stahlfeld CN, Singh A, Chen XE, Kwak L, Gilsdorf CS, Wolfe SK, Wei XX, Silver R, Zhang Z, Morris MJ, Bubley G, Feng FY, Scher HI, Rathkopf D, Dehm SM, Choueiri TK, Halabi S, Armstrong AJ, Wyatt AW, Taplin ME, Zhao SG, Lang JM. Prospective Evaluation of Clinical Outcomes Using a Multiplex Liquid Biopsy Targeting Diverse Resistance Mechanisms in Metastatic Prostate Cancer. J Clin Oncol 2021; 39:2926-2937. [PMID: 34197212 PMCID: PMC8425833 DOI: 10.1200/jco.21.00169] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nearly all men with prostate cancer treated with androgen receptor (AR) signaling inhibitors (ARSIs) develop resistance via diverse mechanisms including constitutive activation of the AR pathway, driven by AR genomic structural alterations, expression of AR splice variants (AR-Vs), or loss of AR dependence and lineage plasticity termed neuroendocrine prostate cancer. Understanding these de novo acquired ARSI resistance mechanisms is critical for optimizing therapy.
Collapse
Affiliation(s)
- Jamie M Sperger
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Hamid Emamekhoo
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Rana R McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | | | - Anupama Singh
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Xinyi E Chen
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Lucia Kwak
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Cole S Gilsdorf
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Serena K Wolfe
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Xiao X Wei
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Rebecca Silver
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Zhenwei Zhang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Michael J Morris
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Glenn Bubley
- Beth Israel Deaconess Medical Center, Boston, MA
| | - Felix Y Feng
- Division of Hematology and Oncology, Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA.,Department of Radiation Oncology, University of California San Francisco, San Francisco, CA.,Department of Urology, University of California San Francisco, San Francisco, CA
| | - Howard I Scher
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Dana Rathkopf
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Scott M Dehm
- Departments of Laboratory Medicine and Pathology and Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Toni K Choueiri
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Susan Halabi
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC.,Department of Biostatistics and Bioinformatics, Duke University, Durham, NC
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC
| | - Alexander W Wyatt
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | | | - Shuang G Zhao
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI.,Department of Human Oncology, University of Wisconsin, Madison, WI.,William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Joshua M Lang
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, WI.,Department of Medicine, University of Wisconsin, Madison, WI
| |
Collapse
|
26
|
McKay RR, Kwak L, Crowdis JP, Sperger JM, Zhao SG, Xie W, Werner L, Lis RT, Zhang Z, Wei XX, Lang JM, Van Allen EM, Bhatt RS, Yu EY, Nelson PS, Bubley GJ, Montgomery RB, Taplin ME. Phase II Multicenter Study of Enzalutamide in Metastatic Castration-Resistant Prostate Cancer to Identify Mechanisms Driving Resistance. Clin Cancer Res 2021; 27:3610-3619. [PMID: 33849963 PMCID: PMC8254786 DOI: 10.1158/1078-0432.ccr-20-4616] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/23/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Enzalutamide is a second-generation androgen receptor (AR) inhibitor that has improved overall survival (OS) in metastatic castration-resistant prostate cancer (CRPC). However, nearly all patients develop resistance. We designed a phase II multicenter study of enzalutamide in metastatic CRPC incorporating tissue and blood biomarkers to dissect mechanisms driving resistance. PATIENTS AND METHODS Eligible patients with metastatic CRPC underwent a baseline metastasis biopsy and then initiated enzalutamide 160 mg daily. A repeat metastasis biopsy was obtained at radiographic progression from the same site when possible. Blood for circulating tumor cell (CTC) analysis was collected at baseline and progression. The primary objective was to analyze mechanisms of resistance in serial biopsies. Whole-exome sequencing was performed on tissue biopsies. CTC samples underwent RNA sequencing. RESULTS A total of 65 patients initiated treatment, of whom 22 (33.8%) had received prior abiraterone. Baseline biopsies were enriched for alterations in AR (mutations, amplifications) and tumor suppression genes (PTEN, RB1, and TP53), which were observed in 73.1% and 92.3% of baseline biopsies, respectively. Progression biopsies revealed increased AR amplifications (64.7% at progression vs. 53.9% at baseline) and BRCA2 alterations (64.7% at progression vs. 38.5% at baseline). Genomic analysis of baseline and progression CTC samples demonstrated increased AR splice variants, AR-regulated genes, and neuroendocrine markers at progression. CONCLUSIONS Our results demonstrate that a large proportion of enzalutamide-treated patients have baseline and progression alterations in the AR pathway and tumor suppressor genes. We demonstrate an increased number of BRCA2 alterations post-enzalutamide, highlighting the importance of serial tumor sampling in CRPC.
Collapse
Affiliation(s)
- Rana R McKay
- University of California San Diego, San Diego, California
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lucia Kwak
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jamie M Sperger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Shuang G Zhao
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wanling Xie
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Rosina T Lis
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Xiao X Wei
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Rupal S Bhatt
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Evan Y Yu
- University of Washington, Seattle, Washington
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Peter S Nelson
- University of Washington, Seattle, Washington
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Glenn J Bubley
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - R Bruce Montgomery
- University of Washington, Seattle, Washington
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | |
Collapse
|
27
|
Ayuso JM, Park KY, Virumbrales-Muñoz M, Beebe DJ. Toward improved in vitro models of human cancer. APL Bioeng 2021; 5:010902. [PMID: 33532672 PMCID: PMC7822630 DOI: 10.1063/5.0026857] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of death across the world and continues to increase in incidence. Despite years of research, multiple tumors (e.g., glioblastoma, pancreatic cancer) still have limited treatment options in the clinic. Additionally, the attrition rate and cost of drug development have continued to increase. This trend is partly explained by the poor predictive power of traditional in vitro tools and animal models. Moreover, multiple studies have highlighted that cell culture in traditional Petri dishes commonly fail to predict drug sensitivity. Conversely, animal models present differences in tumor biology compared with human pathologies, explaining why promising therapies tested in animal models often fail when tested in humans. The surging complexity of patient management with the advent of cancer vaccines, immunotherapy, and precision medicine demands more robust and patient-specific tools to better inform our understanding and treatment of human cancer. Advances in stem cell biology, microfluidics, and cell culture have led to the development of sophisticated bioengineered microscale organotypic models (BMOMs) that could fill this gap. In this Perspective, we discuss the advantages and limitations of patient-specific BMOMs to improve our understanding of cancer and how these tools can help to confer insight into predicting patient response to therapy.
Collapse
Affiliation(s)
| | - Keon-Young Park
- Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | | | | |
Collapse
|
28
|
Kyriakopoulos CE, Heath EI, Ferrari A, Sperger JM, Singh A, Perlman SB, Roth AR, Perk TG, Modelska K, Porcari A, Duggan W, Lang JM, Jeraj R, Liu G. Exploring Spatial-Temporal Changes in 18F-Sodium Fluoride PET/CT and Circulating Tumor Cells in Metastatic Castration-Resistant Prostate Cancer Treated With Enzalutamide. J Clin Oncol 2020; 38:3662-3671. [PMID: 32897830 DOI: 10.1200/jco.20.00348] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Intrapatient treatment response heterogeneity is under-recognized. Quantitative total bone imaging (QTBI) using 18F-NaF positron emission tomography/computed tomography (PET/CT) scans is a tool that allows characterization of interlesional treatment response heterogeneity in bone. Understanding spatial-temporal response is important to identify individuals who may benefit from treatment beyond progression. PATIENTS AND METHODS Men with progressive metastatic castration-resistant prostate cancer (mCRPC) with at least two lesions on bone scintigraphy were enrolled and treated with enzalutamide 160 mg daily (ClinicalTrials.gov identifier: NCT02384382). 18F-NaF PET/CT scans were obtained at baseline (PET1), week 13 (PET2), and at the time of prostate-specific antigen (PSA) progression, standard radiographic or clinical progression, or at 2 years without progression (PET3). QTBI was used to determine lesion-level response. The primary end point was the proportion of men with at least one responding bone lesion on PET3 using QTBI. RESULTS Twenty-three men were enrolled. Duration on treatment ranged from 1.4 to 34.1 months. In general, global standardized uptake value (SUV) metrics decreased while on enzalutamide (PET2) and increased at the time of progression (PET3). The most robust predictor of PSA progression was change in SUVhetero (PET1 to PET3; hazard ratio, 3.88; 95% CI, 1.24 to 12.1). Although overall functional disease burden improved during enzalutamide treatment, an increase in total burden (SUVtotal) was seen at the time of progression, as measured by 18F-NaF PET/CT. All (22/22) evaluable men had at least one responding bone lesion at PET3 using QTBI. CONCLUSION We found that the proportion of progressing lesions was low, indicating that a substantial number of lesions appear to continue to benefit from enzalutamide beyond progression. Selective targeting of nonresponding lesions may be a reasonable approach to extend benefit.
Collapse
Affiliation(s)
| | - Elisabeth I Heath
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI
| | - Anna Ferrari
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Jamie M Sperger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Anupama Singh
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Scott B Perlman
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI.,Department of Radiology, University of Wisconsin, Madison, WI
| | - Alison R Roth
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI.,Department of Medical Physics, University of Wisconsin, Madison, WI
| | - Timothy G Perk
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI.,Department of Medical Physics, University of Wisconsin, Madison, WI
| | | | | | | | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI
| | - Robert Jeraj
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI.,Department of Medical Physics, University of Wisconsin, Madison, WI.,AIQ Solutions, Madison, WI
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI.,AIQ Solutions, Madison, WI
| |
Collapse
|
29
|
Chen K, Amontree J, Varillas J, Zhang J, George TJ, Fan ZH. Incorporation of lateral microfiltration with immunoaffinity for enhancing the capture efficiency of rare cells. Sci Rep 2020; 10:14210. [PMID: 32848184 PMCID: PMC7450051 DOI: 10.1038/s41598-020-71041-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/22/2020] [Indexed: 02/03/2023] Open
Abstract
The methods for isolating rare cells such as circulating tumor cells (CTCs) can be generally classified into two categories: those based on physical properties (e.g., size) and methods based on biological properties (e.g., immunoaffinity). CellSearch, the only FDA-approved method for the CTC-based cancer prognosis, relies on immunoaffinity interactions between CTCs and antibodies immobilized on magnetic particles. Immunoaffinity-based CTC isolation has also been employed in microfluidic devices, which show higher capture efficiency than CellSearch. We report here our investigation of combining size-based microfiltration into a microfluidic device with immunoaffinity for enhanced capture efficiency of CTCs. The device consists of four serpentine main channels, and each channel contains an array of lateral filters that create a two-dimensional flow. The main flow is through the serpentine channel, allowing the majority of the sample to pass by while the secondary flow goes through the lateral filters. The device design is optimized to make all fluid particles interact with filters. The filter sizes range from 24 to 12 µm, being slightly larger than or having similar dimension of CTCs. These filters are immobilized with antibodies specific to CTCs and thus they function as gates, allowing normal blood cells to pass by while forcing the interactions between CTCs and antibodies on the filter surfaces. The hydrodynamic force experienced by a CTC was also studied for optimal experimental conditions to ensure immunoaffinity-enabled cell capture. The device was evaluated by capturing two types of tumor cells spiked in healthy blood or a buffer, and we found that their capture efficiency was between 87.2 and 93.5%. The platform was further validated by isolating CTCs from blood samples of patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Kangfu Chen
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jacob Amontree
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jose Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, P.O. Box 100278, Gainesville, FL, 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL, 32611, USA.
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA.
| |
Collapse
|
30
|
Carrozzo M, Eriksen JG, Bensadoun RJ, Boers-Doets CB, Lalla RV, Peterson DE. Oral Mucosal Injury Caused by Targeted Cancer Therapies. J Natl Cancer Inst Monogr 2020; 2019:5551364. [PMID: 31425602 DOI: 10.1093/jncimonographs/lgz012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/14/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023] Open
Abstract
Targeted cancer therapies have fundamentally transformed the treatment of many types of cancers over the past decade, including breast, colorectal, lung, and pancreatic cancers, as well as lymphoma, leukemia, and multiple myeloma. The unique mechanisms of action of these agents have resulted in many patients experiencing enhanced tumor response together with a reduced adverse event profile as well. Toxicities do continue to occur, however, and in selected cases can be clinically challenging to manage. Of particular importance in the context of this monograph is that the pathobiology for oral mucosal lesions caused by targeted cancer therapies has only been preliminarily investigated. There is distinct need for novel basic, translational, and clinical research strategies to enhance design of preventive and therapeutic approaches for patients at risk for development of these lesions. The research modeling can be conceptually enhanced by extrapolating "lessons learned" from selected oral mucosal conditions in patients without cancer as well. This approach may permit determination of the extent to which pathobiology and clinical management are either similar to or uniquely distinct from oral mucosal lesions caused by targeted cancer therapies. Modeling associated with oral mucosal disease in non-oncology patients is thus presented in this context as well. This article addresses this emerging paradigm, with emphasis on current mechanistic modeling and clinical treatment. This approach is in turn designed to foster delineation of new research strategies, with the goal of enhancing cancer patient treatment in the future.
Collapse
Affiliation(s)
- M Carrozzo
- Center for Oral Health Research, Oral Medicine Department, School of Dental Sciences, Newcastle University, UK
| | - J Grau Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - R-J Bensadoun
- Institut Niçois de Cancérologie (INC), Centre de Haute Energie, Nice, France
| | - C B Boers-Doets
- CancerMed, Department of Medical Strategy, Wormer, The Netherlands.,Impaqtt Foundation, Department of Adverse Event Research & Valorisation, Wormer, The Netherlands
| | - R V Lalla
- Section of Oral Medicine, Department of Oral Health & Diagnostic Sciences, School of Dental Medicine, UConn Health, Farmington, CT
| | - D E Peterson
- Section of Oral Medicine, Department of Oral Health & Diagnostic Sciences, School of Dental Medicine & Neag Comprehensive Cancer Center, UConn Health, Farmington, CT
| |
Collapse
|
31
|
Chen Y, Liu Y, Shi Y, Ping J, Wu J, Chen H. Magnetic particles for integrated nucleic acid purification, amplification and detection without pipetting. Trends Analyt Chem 2020; 127:115912. [PMID: 32382202 PMCID: PMC7202819 DOI: 10.1016/j.trac.2020.115912] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nucleic acid amplification based detection plays an important role in food safety, environmental monitoring and clinical diagnosis. However, traditional nucleic acid detection process involves transferring liquid from one tube to another by pipetting. It requires trained persons, equipped labs and consumes lots of time. The ideal nucleic acid detection is integrated, closed, simplified and automated. Magnetic particles actuated by magnetic fields can efficiently adsorb nucleic acids and promote integrated nucleic acid assays without pipetting driven by pumps and centrifuges. We will comprehensively review magnetic particles assisted integrated system for nucleic acid detection and hope it can inspire further related study.
Collapse
Key Words
- ATP, adenosine triphosphate
- DLS, dynamic light scattering
- FMR, ferromagnetic resonance
- GTC, guanidinium thiocyanate
- ICP-AES, inductively coupled plasma atomic emission spectroscopy
- IFAST, immiscible filtration assisted by surface tension
- Immiscible interface
- Integrated detection
- LAMP, loop-mediated isothermal amplification
- Magnetic particles
- Nucleic acid
- PCR, polymerase chain reaction
- PEG, polyethylene glycol
- POCT, point-of-care testing
- RPA, recombinase polymerase amplification
- SQUID, superconducting quantum interference device magnetometer
- TEM, transmission electron microscopy
- XRD, X-Ray diffraction
- qPCR, quantitative PCR
Collapse
Affiliation(s)
- Yanju Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yang Liu
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, 310012, China
| | - Ya Shi
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, 310012, China
| | - Jianfeng Ping
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jian Wu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of on Site Processing Equipment for Agricultural Products, Ministry of Agriculture, Hangzhou, 310058, China
| | - Huan Chen
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, 310012, China
| |
Collapse
|
32
|
Singh A, Denu RA, Wolfe SK, Sperger JM, Schehr J, Witkowsky T, Esbona K, Chappell RJ, Weaver BA, Burkard ME, Lang JM. Centrosome amplification is a frequent event in circulating tumor cells from subjects with metastatic breast cancer. Mol Oncol 2020; 14:1898-1909. [PMID: 32255253 PMCID: PMC7400789 DOI: 10.1002/1878-0261.12687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 01/05/2023] Open
Abstract
Centrosome amplification (CA) is a common phenomenon in cancer, promotes genomic stability and cancer evolution, and has been reported to promote metastasis. CA promotes a stochastic gain/loss of chromosomes during cell division, known as chromosomal instability (CIN). However, it is unclear whether CA is present in circulating tumor cells (CTCs), the seeds for metastasis. Here, we surveyed CA in CTCs from human subjects with metastatic breast cancer. CTCs were captured by CD45 exclusion and selection of EpCAM‐positive cells using an exclusion‐based sample preparation technology platform known as VERSA (versatile exclusion‐based rare sample analysis). Centriole amplification (centrin foci> 4) is the definitive assay for CA. However, determination of centrin foci is technically challenging and incompatible with automated analysis. To test if the more technically accessible centrosome marker pericentrin could serve as a surrogate for centriole amplification in CTCs, cells were stained with pericentrin and centrin antibodies to evaluate CA. This assay was first validated using breast cancer cell lines and a nontransformed epithelial cell line model of inducible CA, then translated to CTCs. Pericentrin area and pericentrin area x intensity correlate well with centrin foci, validating pericentrin as a surrogate marker of CA. CA is found in CTCs from 75% of subjects, with variability in the percentage and extent of CA in individual circulating cells in a given subject, similar to the variability previously seen in primary tumors and cell lines. In summary, we created, validated, and implemented a novel method to assess CA in CTCs from subjects with metastatic breast cancer. Such an assay will be useful for longitudinal monitoring of CA in cancer patients and in prospective clinical trials for assessing the impact of CA on response to therapy.
Collapse
Affiliation(s)
- Ashok Singh
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Ryan A Denu
- Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison, WI, USA
| | - Serena K Wolfe
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Jamie M Sperger
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Jennifer Schehr
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Tessa Witkowsky
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Karla Esbona
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA
| | - Richard J Chappell
- Departments of Statistics and of Biostatistics & Medical Informatics, University of Wisconsin-Madison, WI, USA
| | - Beth A Weaver
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA.,Department of Cell and Regenerative Biology and Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, WI, USA
| | - Mark E Burkard
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA.,Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison, WI, USA
| | - Joshua M Lang
- Carbone Cancer Center, University of Wisconsin-Madison, WI, USA.,Department of Medicine, Division of Hematology/Oncology, University of Wisconsin-Madison, WI, USA
| |
Collapse
|
33
|
Hamid AB, Petreaca RC. Secondary Resistant Mutations to Small Molecule Inhibitors in Cancer Cells. Cancers (Basel) 2020; 12:cancers12040927. [PMID: 32283832 PMCID: PMC7226513 DOI: 10.3390/cancers12040927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Secondary resistant mutations in cancer cells arise in response to certain small molecule inhibitors. These mutations inevitably cause recurrence and often progression to a more aggressive form. Resistant mutations may manifest in various forms. For example, some mutations decrease or abrogate the affinity of the drug for the protein. Others restore the function of the enzyme even in the presence of the inhibitor. In some cases, resistance is acquired through activation of a parallel pathway which bypasses the function of the drug targeted pathway. The Catalogue of Somatic Mutations in Cancer (COSMIC) produced a compendium of resistant mutations to small molecule inhibitors reported in the literature. Here, we build on these data and provide a comprehensive review of resistant mutations in cancers. We also discuss mechanistic parallels of resistance.
Collapse
|
34
|
Tokar JJ, Stahlfeld CN, Sperger JM, Niles DJ, Beebe DJ, Lang JM, Warrick JW. Pairing Microwell Arrays with an Affordable, Semiautomated Single-Cell Aspirator for the Interrogation of Circulating Tumor Cell Heterogeneity. SLAS Technol 2020; 25:162-176. [PMID: 31983266 PMCID: PMC8879417 DOI: 10.1177/2472630319898146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Comprehensive analysis of tumor heterogeneity requires robust methods for the isolation and analysis of single cells from patient samples. An ideal approach would be fully compatible with downstream analytic methods, such as advanced genomic testing. These endpoints necessitate the use of live cells at high purity. A multitude of microfluidic circulating tumor cell (CTC) enrichment technologies exist, but many of those perform bulk sample enrichment and are not, on their own, capable of single-cell interrogation. To address this, we developed an affordable semiautomated single-cell aspirator (SASCA) to further enrich rare-cell populations from a specialized microwell array, per their phenotypic markers. Immobilization of cells within microwells, integrated with a real-time image processing software, facilitates the detection and precise isolation of targeted cells that have been optimally seeded into the microwells. Here, we demonstrate the platform capabilities through the aspiration of target cells from an impure background population, where we obtain purity levels of 90%-100% and demonstrate the enrichment of the target population with high-quality RNA extraction. A range of low cell numbers were aspirated using SASCA before undergoing whole transcriptome and genome analysis, exhibiting the ability to obtain endpoints from low-template inputs. Lastly, CTCs from patients with castration-resistant prostate cancer were isolated with this platform and the utility of this method was confirmed for rare-cell isolation. SASCA satisfies a need for an affordable option to isolate single cells or highly purified subpopulations of cells to probe complex mechanisms driving disease progression and resistance in patients with cancer.
Collapse
Affiliation(s)
- Jacob J Tokar
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
| | | | - Jamie M Sperger
- Dept. of Medicine – Univ. of Wisconsin, Madison - Madison, USA
| | - David J Niles
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
| | - David J Beebe
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
- UW Carbone Cancer Center. – Univ. of Wisconsin, Madison - Madison, USA
| | - Joshua M Lang
- UW Carbone Cancer Center. – Univ. of Wisconsin, Madison - Madison, USA
- Dept. of Medicine – Univ. of Wisconsin, Madison - Madison, USA
| | - Jay W Warrick
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
| |
Collapse
|
35
|
Batth IS, Mitra A, Rood S, Kopetz S, Menter D, Li S. CTC analysis: an update on technological progress. Transl Res 2019; 212:14-25. [PMID: 31348892 PMCID: PMC6755047 DOI: 10.1016/j.trsl.2019.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/21/2019] [Accepted: 07/09/2019] [Indexed: 12/28/2022]
Abstract
There is a growing need for a more accurate, real-time assessment of tumor status and the probability of metastasis, relapse, or response to treatment. Conventional means of assessment include imaging and tissue biopsies that can be highly invasive, may not provide complete information of the disease's heterogeneity, and not ideal for repeat analysis. Therefore, a less-invasive means of acquiring similar information at greater time points is necessary. Liquid biopsies are samples of a patients' peripheral blood and hold potential of addressing these criteria. Ongoing research has revealed that a tumor can release circulating cells, genetic materials (DNA or RNA), and exosomes into circulation. These potential biomarkers can be captured in a liquid biopsy and analyzed to determine disease status. To achieve these goals, numerous technologies have been developed. In this review, we discuss both prominent and newly developed technologies for circulating tumor cell capture and analysis and their clinical impact.
Collapse
Affiliation(s)
- Izhar S Batth
- Department of Pediatrics - Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abhisek Mitra
- Department of Pediatrics - Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Scott Kopetz
- Department of Gastrointestinal (GI) Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - David Menter
- Department of Gastrointestinal (GI) Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Shulin Li
- Department of Pediatrics - Research, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
36
|
Juang DS, Berry SM, Li C, Lang JM, Beebe DJ. Centrifugation-Assisted Immiscible Fluid Filtration for Dual-Bioanalyte Extraction. Anal Chem 2019; 91:11848-11855. [PMID: 31411020 DOI: 10.1021/acs.analchem.9b02572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The extraction of bioanalytes is the first step in many diagnostic and analytical assays. However, most bioanalyte extraction methods require extensive dilution-based washing processes that are not only time-consuming and laborious but can also result in significant sample loss, limiting their applications in rare sample analyses. Here, we present a method that enables the efficient extraction of multiple different bioanalytes from rare samples (down to 10 cells) without washing-centrifugation-assisted immiscible fluid filtration (CIFF). CIFF utilizes centrifugal force to drive the movement of analyte-bound glass microbeads from an aqueous sample into an immiscible hydrophobic solution to perform an efficient, simple, and nondilutive extraction. The method can be performed using conventional polymerase chain reaction (PCR) tubes with no requirement of specialized devices, columns, or instruments, making it broadly accessible and cost-effective. The CIFF process can effectively remove approximately 99.5% of the aqueous sample in one extraction with only 0.5% residual carryover, whereas a traditional "spin-down and aspirate" operation results in a higher 3.6% carryover. Another unique aspect of CIFF is its ability to perform two different solid-phase bioanalytes extractions simultaneously within a single vessel without fractionating the sample or performing serial extractions. Here we demonstrate efficient mRNA and DNA extraction from low-input samples (down to 10 cells) with slightly higher to comparable recovery compared to a traditional column-based extraction technique and the simultaneous extraction of two different proteins in the same tube using CIFF.
Collapse
|
37
|
Li C, Niles DJ, Juang DS, Lang JM, Beebe DJ. Automated System for Small-Population Single-Particle Processing Enabled by Exclusive Liquid Repellency. SLAS Technol 2019; 24:535-542. [PMID: 31180792 DOI: 10.1177/2472630319853219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Exclusive liquid repellency (ELR) describes an extreme wettability phenomenon in which a liquid phase droplet is completely repelled from a solid phase when exposed to a secondary immiscible liquid phase. Earlier, we developed a multi-liquid-phase open microfluidic (or underoil) system based on ELR to facilitate rare-cell culture and single-cell processing. The ELR system can allow for the handling of small volumes of liquid droplets with ultra-low sample loss and biofouling, which makes it an attractive platform for biological applications that require lossless manipulation of rare cellular samples (especially for a limited sample size in the range of a few hundred to a few thousand cells). Here, we report an automated platform using ELR microdrops for single-particle (or single-cell) isolation, identification, and retrieval. This was accomplished via the combined use of a robotic liquid handler, an automated microscopic imaging system, and real-time image-processing software for single-particle identification. The automated ELR technique enables rapid, hands-free, and robust isolation of microdrop-encapsulated rare cellular samples.
Collapse
Affiliation(s)
- Chao Li
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Niles
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Duane S Juang
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
38
|
Lu YT, Delijani K, Mecum A, Goldkorn A. Current status of liquid biopsies for the detection and management of prostate cancer. Cancer Manag Res 2019; 11:5271-5291. [PMID: 31239778 PMCID: PMC6559244 DOI: 10.2147/cmar.s170380] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
In recent years, new therapeutic options have become available for prostate cancer (PC) patients, generating an urgent need for better biomarkers to guide the choice of therapy and monitor treatment response. Liquid biopsies, including circulating tumor cells (CTCs), circulating nucleic acids, and exosomes, have been developed as minimally invasive assays allowing oncologists to monitor PC patients with real-time cellular or molecular information. While CTC counts remain the most extensively validated prognostic biomarker to monitor treatment response, recent advances demonstrate that CTC morphology and androgen receptor characterization can provide additional information to guide the choice of treatment. Characterization of cell-free DNA (cfDNA) is another rapidly emerging field with novel technologies capable of monitoring the evolution of treatment relevant alterations such as those in DNA damage repair genes for poly (ADP-ribose) polymerase (PARP) inhibition. In addition, several new liquid biopsy fields are emerging, including the characterization of heterogeneity, CTC RNA sequencing, the culture and xenografting of CTCs, and the characterization of extracellular vesicles (EVs) and circulating microRNAs. This review describes the clinical utilization of liquid biopsies in the management of PC patients and emerging liquid biopsy technologies with the potential to advance personalized cancer therapy.
Collapse
Affiliation(s)
- Yi-Tsung Lu
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Kevin Delijani
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Andrew Mecum
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
39
|
Worroll D, Galletti G, Gjyrezi A, Nanus DM, Tagawa ST, Giannakakou P. Androgen receptor nuclear localization correlates with AR-V7 mRNA expression in circulating tumor cells (CTCs) from metastatic castration resistance prostate cancer patients. Phys Biol 2019; 16:036003. [PMID: 30763921 DOI: 10.1088/1478-3975/ab073a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Androgen receptor (AR) signaling drives prostate cancer (PC) progression and remains active upon transition to castration resistant prostate cancer (CRPC). Active AR signaling is achieved through the nuclear accumulation of AR following ligand binding and through expression of ligand-independent, constitutively active AR splice variants, such as AR-V7, which is the most commonly expressed variant in metastatic CRPC (mCRPC) patients. Most currently approved PC therapies aim to abrogate AR signaling and activity by inhibiting this ligand-mediated nuclear translocation. In a prospective multi-institutional clinical study, we recently showed that taxane based chemotherapy is also capable of impairing AR nuclear localization (ARNL) in circulating tumor cells (CTCs) from CRPC patients, whereas taxane induced decreases in ARNL were associated with response. Thus, quantitative assessment of ARNL in CTCs can be used to monitor therapeutic response in patients and help guide clinical decisions. Here, we describe the development and implementation of quantitative high throughput (QHT) image analysis algorithms to aid in CTC identification and quantitative assessment of percent ARNL (%ARNL). We applied this algorithm to fifteen CRPC patients at the start of taxane chemotherapy, quantified %ARNL in CTCs, and correlated with expression of AR-V7 mRNA (from CTCs enriched via negative, CD45+ depletion of peripheral blood) and with biochemical (prostate specific antigen; PSA) response to taxane chemotherapy. We found that CTCs from AR-V7 positive patients had higher baseline %ARNL compared to CTCs from AR-V7 negative patients, consistent with the constitutive nuclear localization of AR-V7. In addition, lower %ARNL in CTCs at baseline was associated with biochemical response to taxane chemotherapy. High inter- and intra-patient heterogeneity was also observed. As ARNL is required for active AR signaling, the QHT algorithms described herein can provide prognostic and/or predictive value in future clinical studies.
Collapse
Affiliation(s)
- Daniel Worroll
- Department of Medicine, Hematology/Oncology, Weill Cornell Medicine, New York, NY, United States of America. Author to whom any correspondence should be addressed
| | | | | | | | | | | |
Collapse
|
40
|
Kozminsky M, Fouladdel S, Chung J, Wang Y, Smith DC, Alva A, Azizi E, Morgan T, Nagrath S. Detection of CTC Clusters and a Dedifferentiated RNA-Expression Survival Signature in Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801254. [PMID: 30693182 PMCID: PMC6343066 DOI: 10.1002/advs.201801254] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/19/2018] [Indexed: 05/20/2023]
Abstract
Rates of progression and treatment response in advanced prostate cancer are highly variable, necessitating non-invasive methods to assess the molecular characteristics of these tumors in real time. The unique potential of circulating tumor cells (CTCs) to serve as a clinically useful liquid biomarker is due to their ability to inform via both enumeration and RNA expression. A microfluidic graphene oxide-based device (GO Chip) is used to isolate CTCs and CTC clusters from the whole blood of 41 men with metastatic castration-resistant prostate cancer. Additionally, the expression of 96 genes of interest is determined by RT-qPCR. Multivariate analyses are conducted to determine the genes most closely associated with overall survival, PSA progression, and radioclinical progression. A preliminary signature, comprising high expression of stemness genes and low expression of epithelial and mesenchymal genes, potentially implicates an undifferentiated CTC phenotype as a marker of poor prognosis in this setting.
Collapse
Affiliation(s)
- Molly Kozminsky
- Department of Chemical EngineeringUniversity of Michigan2300 Hayward StreetAnn ArborMI48109USA
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
| | - Shamileh Fouladdel
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
- Department of Internal MedicineUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Jae‐Seung Chung
- Department of UrologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Yugang Wang
- Department of UrologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - David C. Smith
- Department of Internal MedicineDivision of Hematology/OncologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Ajjai Alva
- Department of Internal MedicineDivision of Hematology/OncologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Ebrahim Azizi
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
- Department of Internal MedicineUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Todd Morgan
- Department of UrologyUniversity of Michigan1500 E. Medical Center DriveAnn ArborMI48109USA
| | - Sunitha Nagrath
- Department of Chemical EngineeringUniversity of Michigan2300 Hayward StreetAnn ArborMI48109USA
- Translational Oncology ProgramUniversity of Michigan1600 Huron PkwyAnn ArborMI48109USA
- Biointerfaces InstituteUniversity of Michigan2800 Plymouth RoadAnn ArborMI48109USA
| |
Collapse
|
41
|
Pezzi HM, Guckenberger DJ, Schehr JL, Rothbauer J, Stahlfeld C, Singh A, Horn S, Schultz ZD, Bade RM, Sperger JM, Berry SM, Lang JM, Beebe DJ. Versatile exclusion-based sample preparation platform for integrated rare cell isolation and analyte extraction. LAB ON A CHIP 2018; 18:3446-3458. [PMID: 30334061 PMCID: PMC6402328 DOI: 10.1039/c8lc00620b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Rare cell populations provide a patient-centric tool to monitor disease treatment, response, and resistance. However, understanding rare cells is a complex problem, which requires cell isolation/purification and downstream molecular interrogation - processes challenged by non-target populations, which vary patient-to-patient and change with disease. As such, cell isolation platforms must be amenable to a range of sample types while maintaining high efficiency and purity. The multiplexed technology for automated extraction (mTAE) is a versatile magnetic bead-based isolation platform that facilitates positive, negative, and combinatorial selection with integrated protein staining and nucleic acid isolation. mTAE is validated by isolating circulating tumor cells (CTCs) - a model rare cell population - from breast and prostate cancer patient samples. Negative selection yielded high efficiency capture of CTCs while positive selection yielded higher purity with an average of only 95 contaminant cells captured per milliliter of processed whole blood. With combinatorial selection, an overall increase in capture efficiency was observed, highlighting the potential significance of integrating multiple capture approaches on a single platform. Following capture (and staining), on platform nucleic acid extraction enabled the detection of androgen receptor-related transcripts from CTCs isolated from prostate cancer patients. The flexibility (e.g. negative, positive, combinatorial selection) and capabilities (e.g. isolation, protein staining, and nucleic acid extraction) of mTAE enable users to freely interrogate specific cell populations, a capability required to understand the potential of emerging rare cell populations and readily adapt to the heterogeneity presented across clinical samples.
Collapse
Affiliation(s)
- Hannah M Pezzi
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Seenivasan R, Warrick JW, Rodriguez CI, Mattison W, Beebe DJ, Setaluri V, Gunasekaran S. Integrating Electrochemical Immunosensing and Cell Adhesion Technologies for Cancer Cell Detection and Enumeration. Electrochim Acta 2018; 286:205-211. [PMID: 31130739 PMCID: PMC6530932 DOI: 10.1016/j.electacta.2018.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We have successfully integrated techniques for controlling cell adhesion and performing electrochemical differential pulse voltammetry (DPV) through the use of digitally controlled microfluidics and patterned transparent indium tin oxide electrode arrays to enable rapid and sensitive enumeration of cancer cells in a scalable microscale format. This integrated approach leverages a dual-working electrode (WE) surface to improve the specificity of the detection system. Here, one of the WE surfaces is functionalized with anti-Melanocortin 1 Receptor antibodies specific to melanoma cancer cells, while the other WE acts as a control (i.e., without antibody), for detecting non-specific interactions between cells and the electrode. The method is described and shown to provide effective detection of melanoma cells at concentrations ranging between 25 to 300 cells per 20 μL sample volume after a 5 min incubation and 15 s of DPV measurements. The estimated limit of detection was ~17 cells. The sensitivity and specificity of the assay were quantified using addition of large fractions of non-target cells and resulted in a detection reproducibility of ~97%. The proposed approach demonstrates a unique integration of electrochemical sensing and microfluidic cell adhesion technologies with multiple advantages such as label-free detection, short detection times, and low sample volumes. Next steps for this platform include testing with patient samples and use of other cell-surface biomarkers for detection and enumeration of circulating tumor cells in prostate, breast, and colon cancer.
Collapse
Affiliation(s)
- Rajesh Seenivasan
- Department of Biological Systems Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jay W. Warrick
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Carlos I. Rodriguez
- Department of Dermatology, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - William Mattison
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Vijayasaradhi Setaluri
- Department of Dermatology, UW School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sundaram Gunasekaran
- Department of Biological Systems Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
43
|
Li C, Yu J, Schehr J, Berry SM, Leal TA, Lang JM, Beebe DJ. Exclusive Liquid Repellency: An Open Multi-Liquid-Phase Technology for Rare Cell Culture and Single-Cell Processing. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17065-17070. [PMID: 29738227 PMCID: PMC9703972 DOI: 10.1021/acsami.8b03627] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The concept of high liquid repellency in multi-liquid-phase systems (e.g., aqueous droplets in an oil background) has been applied to areas of biomedical research to realize intrinsic advantages not available in single-liquid-phase systems. Such advantages have included minimizing analyte loss, facile manipulation of single-cell samples, elimination of biofouling, and ease of use regarding loading and retrieving of the sample. In this paper, we present generalized design rules for predicting the wettability of solid-liquid-liquid systems (especially for discrimination between exclusive liquid repellency (ELR) and finite liquid repellency) to extend the applications of ELR. We then apply ELR to two model systems with open microfluidic design in cell biology: (1) in situ underoil culture and combinatorial coculture of mammalian cells in order to demonstrate directed single-cell multiencapsulation with minimal waste of samples as compared to stochastic cell seeding and (2) isolation of a pure population of circulating tumor cells, which is required for certain downstream analyses including sequencing and gene expression profiling.
Collapse
Affiliation(s)
- Chao Li
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin, WI 53705 (United States)
| | - Jiaquan Yu
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin, WI 53705 (United States)
| | - Jennifer Schehr
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 (United States)
| | - Scott M. Berry
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin, WI 53705 (United States)
| | - Ticiana A. Leal
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 (United States)
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53792 (United States)
| | - Joshua M. Lang
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin, WI 53705 (United States)
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 (United States)
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53792 (United States)
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Wisconsin, WI 53705 (United States)
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705 (United States)
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53792 (United States)
- Corresponding Author:
| |
Collapse
|
44
|
Pezzi H, Niles DJ, Schehr JL, Beebe DJ, Lang JM. Integration of Magnetic Bead-Based Cell Selection into Complex Isolations. ACS OMEGA 2018; 3:3908-3917. [PMID: 29732449 PMCID: PMC5928489 DOI: 10.1021/acsomega.7b01427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/22/2018] [Indexed: 05/12/2023]
Abstract
Magnetic bead-based analyte capture has emerged as a ubiquitous method in cell isolation, enabling the highly specific capture of target populations through simple magnetic manipulation. To date, no "one-size fits all" magnetic bead has been widely adopted leading to an overwhelming number of commercial beads. Ultimately, the ideal bead is one that not only facilitates cell isolation but also proves compatible with the widest range of downstream applications and analytic endpoints. Despite the diverse offering of sizes, coatings, and conjugation chemistries, few studies exist to benchmark the performance characteristics of different commercially available beads; importantly, these bead characteristics ultimately determine the ability of a bead to integrate into the user's assay. In this report, we evaluate bead-based cell isolation considerations, approaches, and results across a subset of commercially available magnetic beads (Dynabeads FlowComps, Dynabeads CELLection, GE Healthcare Sera-Mag SpeedBeads streptavidin-blocked magnetic particles, Dynabeads M-270s, Dynabeads M-280s) to compare and contrast both capture-specific traits (i.e., purity, capture efficacy, and contaminant isolations) and endpoint compatibility (i.e., protein localization, fluorescence imaging, and nucleic acid extraction). We identify specific advantages and contexts of use in which distinct bead products may facilitate experimental goals and integrate into downstream applications.
Collapse
Affiliation(s)
- Hannah
M. Pezzi
- Department
of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin−Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - David J. Niles
- Department
of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin−Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Jennifer L. Schehr
- Department of Medicine and Carbone Cancer Center, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
| | - David J. Beebe
- Department
of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin−Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- Department of Medicine and Carbone Cancer Center, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
- E-mail: (D.J.B.)
| | - Joshua M. Lang
- Department
of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin−Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- Department of Medicine and Carbone Cancer Center, University
of Wisconsin−Madison, Madison, Wisconsin 53705, United States
- E-mail: (J.M.L.)
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Metastatic prostate cancer is a lethal and highly heterogeneous malignancy, associated with a broad spectrum of potentially actionable molecular alterations. In the past decade, disease profiling has expanded to include not only traditional tumor tissue, but also liquid biopsies of cells and genetic material circulating in the blood. These liquid biopsies offer a minimally invasive, repeatable source of tumor material for longitudinal disease profiling but also raise new technical and biological challenges. Here we will summarize recent advances in liquid biopsy strategies and the role they have played in biomarker development and disease management. RECENT FINDINGS Technologies for analysis of circulating tumor cells (CTCs) continue to evolve rapidly, and the latest high content scanning platforms have underscored the phenotypic heterogeneity of CTC populations. Among liquid biopsies, CTC enumeration remains the most extensively validated prognostic marker to date, but other clinically relevant phenotypes like androgen receptor (AR) localization or presence of AR-V7 splice variant are important new predictors of therapy response. Serial genomic profiling of CTCs or circulating tumor DNA (ctDNA) is helping to define primary and acquired resistance mechanisms and helping to guide patient selection for targeted therapies such as poly(adenosine diphosphate [ADP] ribose) polymerase (PARP) inhibition. The era of liquid biopsy-based biomarkers has arrived, driven by powerful new enrichment and analysis techniques. As new blood-based markers are identified, their biological significance as disease drivers must be elucidated to advance new therapeutic strategies, and their clinical impact must be translated through assay standardization, followed by analytical and clinical validation. These efforts, already ongoing on multiple fronts, constitute the critical steps toward more effective precision management of advanced prostate cancer.
Collapse
Affiliation(s)
- Gareth J Morrison
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Amir Goldkorn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine and Translational and Clinical Science Program, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
46
|
Regulation of inside-out β1-integrin activation by CDCP1. Oncogene 2018; 37:2817-2836. [PMID: 29511352 DOI: 10.1038/s41388-018-0142-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 12/21/2022]
Abstract
Tumor metastasis depends on the dynamic regulation of cell adhesion through β1-integrin. The Cub-Domain Containing Protein-1, CDCP1, is a transmembrane glycoprotein which regulates cell adhesion. Overexpression and loss of CDCP1 have been observed in the same cancer types to promote metastatic progression. Here, we demonstrate reduced CDCP1 expression in high-grade, primary prostate cancers, circulating tumor cells and tumor metastases of patients with castrate-resistant prostate cancer. CDCP1 is expressed in epithelial and not mesenchymal cells, and its cell surface and mRNA expression declines upon stimulation with TGFβ1 and epithelial-to-mesenchymal transition. Silencing of CDCP1 in DU145 and PC3 cells resulted in 3.4-fold higher proliferation of non-adherent cells and 4.4-fold greater anchorage independent growth. CDCP1-silenced tumors grew in 100% of mice, compared to 30% growth of CDCP1-expressing tumors. After CDCP1 silencing, cell adhesion and migration diminished 2.1-fold, caused by loss of inside-out activation of β1-integrin. We determined that the loss of CDCP1 reduces CDK5 kinase activity due to the phosphorylation of its regulatory subunit, CDK5R1/p35, by c-SRC on Y234. This generates a binding site for the C2 domain of PKCδ, which in turn phosphorylates CDK5 on T77. The resulting dissociation of the CDK5R1/CDK5 complex abolishes the activity of CDK5. Mutations of CDK5-T77 and CDK5R1-Y234 phosphorylation sites re-establish the CDK5/CDKR1 complex and the inside-out activity of β1-integrin. Altogether, we discovered a new mechanism of regulation of CDK5 through loss of CDCP1, which dynamically regulates β1-integrin in non-adherent cells and which may promote vascular dissemination in patients with advanced prostate cancer.
Collapse
|
47
|
Heninger E, Krueger TEG, Thiede SM, Sperger JM, Byers BL, Kircher MR, Kosoff D, Yang B, Jarrard DF, McNeel DG, Lang JM. Inducible expression of cancer-testis antigens in human prostate cancer. Oncotarget 2018; 7:84359-84374. [PMID: 27769045 PMCID: PMC5341296 DOI: 10.18632/oncotarget.12711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022] Open
Abstract
Immune tolerance to self-antigens can limit robust anti-tumor immune responses in the use of tumor vaccines. Expression of novel tumor associated antigens can improve immune recognition and lysis of tumor cells. The cancer-testis antigen (CTA) family of proteins has been hypothesized to be an ideal class of antigens due to tumor-restricted expression, a subset of which have been found to induce antibody responses in patients with prostate disease. We demonstrate that CTA expression is highly inducible in five different Prostate Cancer (PC) cell lines using a hypomethylating agent 5-Aza-2′-deoxycytidine (5AZA) and/or a histone deacetylase inhibitor LBH589. These CTAs include NY-ESO1, multiple members of the MAGE and SSX families and NY-SAR35. A subset of CTAs is synergistically induced by the combination of 5AZA and LBH589. We developed an ex vivo organ culture using human PC biopsies for ex vivo drug treatments to evaluate these agents in clinical samples. These assays found significant induction of SSX2 in 9/9 distinct patient samples and NY-SAR35 in 7/9 samples. Further, we identify expression of SSX2 in circulating tumor cells (CTC) from patients with advanced PC. These results indicate that epigenetic modifying agents can induce expression of a broad range of neoantigens in human PC and may serve as a useful adjunctive therapy with novel tumor vaccines and checkpoint inhibitors.
Collapse
Affiliation(s)
- Erika Heninger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Timothy E G Krueger
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Stephanie M Thiede
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Jamie M Sperger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Brianna L Byers
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Madison R Kircher
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Bing Yang
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| |
Collapse
|
48
|
Chalfin HJ, Glavaris SA, Malihi PD, Sperger JM, Gorin MA, Lu C, Goodwin CR, Chen Y, Caruso EA, Dumpit R, Kuhn P, Lang JM, Nelson PS, Luo J, Pienta KJ. Prostate Cancer Disseminated Tumor Cells are Rarely Detected in the Bone Marrow of Patients with Localized Disease Undergoing Radical Prostatectomy across Multiple Rare Cell Detection Platforms. J Urol 2018; 199:1494-1501. [PMID: 29339080 DOI: 10.1016/j.juro.2018.01.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2018] [Indexed: 01/04/2023]
Abstract
PURPOSE Prostate circulating tumor cells escape into peripheral blood and enter bone marrow as disseminated tumor cells, representing an early step before conventionally detectable metastasis. It is unclear how frequently this occurs in localized disease and existing detection methods rely on epithelial markers with low specificity and sensitivity. We used multiple methodologies of disseminated tumor cell detection in bone marrow harvested at radical prostatectomy. MATERIALS AND METHODS Bone marrow was harvested from 208 clinically localized cases, 16 controls and 5 metastatic cases with peripheral blood obtained from 37 metastatic cases. Samples were evaluated at 4 centers with 4 distinct platforms using antibody enrichment with the AdnaTest (Qiagen®) or VERSA (versatile exclusion based rare sample analysis), or whole sample interrogation with the RareCyte platform (Seattle, Washington) or HD-SCA (high definition single cell assay) using traditional epithelial markers and prostate specific markers. We investigated the sensitivity and specificity of these markers by evaluating expression levels in control and metastatic cases. RESULTS EpCAM, NKX3.1 and AR were nonspecifically expressed in controls and in most samples using AdnaTest with no relation to perioperative variables. Only 1 patient with localized disease showed positive results for the prostate specific marker PSA. With the VERSA platform no localized case demonstrated disseminated tumor cells. With the RareCyte and HD-SCA platforms only a single patient had 1 disseminated tumor cell. CONCLUSIONS Evaluation across multiple platforms revealed that epithelial markers are nonspecific in bone marrow and, thus, not suitable for disseminated tumor cell detection. Using prostate specific markers disseminated tumor cells were typically not detected in patients with localized prostate cancer.
Collapse
Affiliation(s)
- Heather J Chalfin
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Stephanie A Glavaris
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Paymaneh D Malihi
- Bridge Institute, University of Southern California, Los Angeles, California
| | - Jamie M Sperger
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Michael A Gorin
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Changxue Lu
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - C Rory Goodwin
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yan Chen
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Emily A Caruso
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ruth Dumpit
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Peter Kuhn
- Bridge Institute, University of Southern California, Los Angeles, California
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jun Luo
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
49
|
Galletti G, Worroll D, Nanus DM, Giannakakou P. Using circulating tumor cells to advance precision medicine in prostate cancer. JOURNAL OF CANCER METASTASIS AND TREATMENT 2017; 3:190-205. [PMID: 29707651 PMCID: PMC5913755 DOI: 10.20517/2394-4722.2017.45] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The field of CTC enrichment has seen many emerging technologies in recent years, which have resulted in the identification and monitoring of clinically relevant, CTC-based biomarkers that can be analyzed routinely without invasive procedures. Several molecular platforms have been used to investigate the molecular profile of the disease, from high throughput gene expression analyses down to single cell biological dissection. The established presence of CTC heterogeneity nevertheless constitutes a challenge for cell isolation as the several subpopulations can potentially display different molecular characteristics; in this scenario, careful consideration must be given to the isolation approach, whereas methods that discriminate against certain subpopulations may result in the exclusion of CTCs that carry biological relevance. In the context of prostate cancer (PC), CTC molecular interrogation can enable longitudinal monitoring of key biological features during treatment with substantial clinical impact, as several biomarkers could predict tumor response to AR signaling inhibitors (abiraterone, enzalutamide) or standard chemotherapy (taxanes). Thus, CTCs represent a valuable opportunity to personalize medicine in current clinical practice.
Collapse
Affiliation(s)
- Giuseppe Galletti
- Department of Medicine, Hematology/Oncology, Weill Cornell Medicine, New York, NY
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Daniel Worroll
- Department of Medicine, Hematology/Oncology, Weill Cornell Medicine, New York, NY
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - David M Nanus
- Department of Medicine, Hematology/Oncology, Weill Cornell Medicine, New York, NY
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Paraskevi Giannakakou
- Department of Medicine, Hematology/Oncology, Weill Cornell Medicine, New York, NY
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
50
|
Clinical utility of emerging liquid biomarkers in advanced prostate cancer. Cancer Genet 2017; 228-229:151-158. [PMID: 28958406 DOI: 10.1016/j.cancergen.2017.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 08/21/2017] [Indexed: 01/04/2023]
Abstract
The therapeutic landscape of advanced prostate cancer (PCa) has rapidly expanded in recent years. Despite significant improvements in patient overall survival, it remains challenging to determine the optimal therapy and sequence of therapies for individual patients. The development of molecular biomarkers will be key for patient stratification, and for monitoring response and resistance to therapy. In this context, minimally-invasive blood-based "liquid" biopsies are attractive as a practical surrogate for solid tumor tissue, providing a window into metastatic disease. Circulating tumor cells and circulating cell-free tumor DNA in particular have demonstrated remarkable potential to inform on PCa patient outcomes through the detection of specific genomic and transcriptomic alterations. This review covers recent advances in the development of clinically-informative liquid biomarkers for advanced PCa.
Collapse
|