1
|
Park J, Lee YT, Agopian VG, Liu JS, Koltsova EK, You S, Zhu Y, Tseng HR, Yang JD. Liquid biopsy in hepatocellular carcinoma: Challenges, advances, and clinical implications. Clin Mol Hepatol 2025; 31:S255-S284. [PMID: 39604328 PMCID: PMC11925447 DOI: 10.3350/cmh.2024.0541] [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: 07/10/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive primary liver malignancy often diagnosed at an advanced stage, resulting in a poor prognosis. Accurate risk stratification and early detection of HCC are critical unmet needs for improving outcomes. Several blood-based biomarkers and imaging tests are available for early detection, prediction, and monitoring of HCC. However, serum protein biomarkers such as alpha-fetoprotein have shown relatively low sensitivity, leading to inaccurate performance. Imaging studies also face limitations related to suboptimal accuracy, high cost, and limited implementation. Recently, liquid biopsy techniques have gained attention for addressing these unmet needs. Liquid biopsy is non-invasive and provides more objective readouts, requiring less reliance on healthcare professional's skills compared to imaging. Circulating tumor cells, cell-free DNA, and extracellular vesicles are targeted in liquid biopsies as novel biomarkers for HCC. Despite their potential, there are debates regarding the role of these novel biomarkers in the HCC care continuum. This review article aims to discuss the technical challenges, recent technical advancements, advantages and disadvantages of these liquid biopsies, as well as their current clinical application and future directions of liquid biopsy in HCC.
Collapse
Affiliation(s)
- Jaeho Park
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yi-Te Lee
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Vatche G. Agopian
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Jessica S Liu
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Ekaterina K. Koltsova
- Smidt Heart Institute, Department of Medicine, Department of Biomedical Sciences, 8700 Beverly Blvd, Los Angeles, CA, USA
| | - Sungyong You
- Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
2
|
Ou Y, Chu GCY, Lyu J, Yin L, Lim A, Zhai N, Cui X, Lewis MS, Edderkaoui M, Pandol SJ, Wang R, Zhang Y. Overcoming Resistance in Prostate Cancer Therapy Using a DZ-Simvastatin Conjugate. Mol Pharm 2024; 21:873-882. [PMID: 38229228 PMCID: PMC11025579 DOI: 10.1021/acs.molpharmaceut.3c00993] [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] [Indexed: 01/18/2024]
Abstract
Prostate cancer (PC), particularly its metastatic castration-resistant form (mCRPC), is a leading cause of cancer-related deaths among men in the Western world. Traditional systemic treatments, including hormonal therapy and chemotherapy, offer limited effectiveness due to tumors' inherent resistance to these therapies. Moreover, they often come with significant side effects. We have developed a delivery method using a tumor-cell-specific heptamethine carbocyanine dye (DZ) designed to transport therapeutic agents directly to tumor cells. This research evaluated simvastatin (SIM) as the antitumor payload because of its demonstrated chemopreventive effects on human cancers and its well-documented safety profile. We designed and synthesized a DZ-SIM conjugate for tumor cell targeting. PC cell lines and xenograft tumor models were used to assess tumor-cell targeting specificity and killing activity and to investigate the corresponding mechanisms. DZ-SIM treatment effectively killed PC cells regardless of their androgen receptor status or inherent therapeutic resistance. The conjugate targeted and suppressed xenograft tumor formation without harming normal cells of the host. In cancer cells, DZ-SIM was enriched in subcellular organelles, including mitochondria, where the conjugate formed adducts with multiple proteins and caused the loss of transmembrane potential, promoting cell death. The DZ-SIM specifically targets PC cells and their mitochondria, resulting in a loss of mitochondrial function and cell death. With a unique subcellular targeting strategy, the conjugate holds the potential to outperform existing chemotherapeutic drugs. It presents a novel strategy to circumvent therapeutic resistance, offering a more potent treatment for mCRPC.
Collapse
Affiliation(s)
- Yan Ou
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Gina Chia-Yi Chu
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ji Lyu
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Liyuan Yin
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Adrian Lim
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ning Zhai
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Xiaojiang Cui
- Department of Surgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Michael S. Lewis
- Department of Pathology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- VA Greater Los Angeles Healthcare System, 11301 Wilshire Blvd, Los Angeles, CA 90073, United States
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Ruoxiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| | - Yi Zhang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, United States
| |
Collapse
|
3
|
Lu L, Hu W, Liu B, Yang T. Insights into Circulating Tumor Cell Clusters: A Barometer for Treatment Effects and Prognosis for Prostate Cancer Patients. Cancers (Basel) 2022; 14:cancers14163985. [PMID: 36010983 PMCID: PMC9406494 DOI: 10.3390/cancers14163985] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Circulating tumor cells (CTCs) are a promising biomarker for the risk of prostate cancer aggressiveness and metastasis and play a role in the processes of tumor migration and metastasis. CTC clusters, which have different physical and biological properties from individual CTCs, are collections of tumor cells and non-malignant cells, resulting in greater metastatic potential. Therefore, this review aims to summarize the current knowledge of CTC clusters in metastasis as well as related biological properties and to suggest possibilities for their usage in diagnostic and therapeutic practice. Abstract Prostate cancer (PCa) exhibits high cellular heterogeneity across patients. Therefore, there is an urgent need for more real-time and accurate detection methods, in both prognosis and treatment in clinical settings. Circulating tumor cell (CTC) clusters, a population of tumor cells and non-malignant cells in the blood of patients with tumors, are a promising non-invasive tool for screening PCa progression and identifying potential benefit groups. CTC clusters are associated with tumor metastasis and possess stem-like characteristics, which are likely attributable to epithelial–mesenchymal transition (EMT). Additionally, these biological properties of CTC clusters, particularly androgen receptor V7, have indicated the potential to reflect curative effects, guide treatment modalities, and predict prognosis in PCa patients. Here, we discuss the role of CTC clusters in the mechanisms underlying PCa metastasis and clinical applications, with the aim of informing more appropriate clinical decisions, and ultimately, improving the overall survival of PCa patients.
Collapse
Affiliation(s)
- Linyao Lu
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Wei Hu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Bingli Liu
- Department of Orthopedics, Shanghai Pudong New Area People’s Hospital, Shanghai 201299, China
- Correspondence: (B.L.); (T.Y.); Tel./Fax: +86-21-2050-9000 (B.L.); +86-21-6803-6506 (T.Y.)
| | - Tao Yang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- Correspondence: (B.L.); (T.Y.); Tel./Fax: +86-21-2050-9000 (B.L.); +86-21-6803-6506 (T.Y.)
| |
Collapse
|
4
|
Teng PC, Agopian VG, Lin TY, You S, Zhu Y, Tseng HR, Yang JD. Circulating tumor cells: A step toward precision medicine in hepatocellular carcinoma. J Gastroenterol Hepatol 2022; 37:1179-1190. [PMID: 35543075 PMCID: PMC9271591 DOI: 10.1111/jgh.15886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/02/2022] [Indexed: 12/09/2022]
Abstract
Serum alpha-fetoprotein and radiologic imaging are the most commonly used tests for early diagnosis and dynamic monitoring of treatment response in hepatocellular carcinoma (HCC). However, the accuracy of these tests is limited, and they may not reflect the underlying biology of the tumor. Thus, developing highly accurate novel HCC biomarkers reflecting tumor biology is a clinically unmet need. Circulating tumor cells (CTCs) have long been proposed as a noninvasive biomarker in clinical oncology. Most CTC assays utilize immunoaffinity-based, size-based, and/or enrichment-free mechanisms followed by immunocytochemical staining to characterize CTCs. The prognostic value of HCC CTC enumeration has been extensively validated. Subsets of CTCs expressing mesenchymal markers are also reported to have clinical significance. In addition, researchers have been devoting their efforts to molecular characterizations of CTCs (e.g. genetics and transcriptomics) as molecular profiling can offer a more accurate readout and provide biological insights. As new molecular profiling techniques, such as digital polymerase chain reaction, are developed to detect minimal amounts of DNA/RNA, several research groups have established HCC CTC digital scoring systems to quantify clinically relevant gene panels. Given the versatility of CTCs to provide intact molecular and functional data that reflects the underlying tumor, CTCs have great potential as a noninvasive biomarker in HCC. Large-scale, prospective studies for HCC CTCs with a standardized protocol are necessary for successful clinical translation.
Collapse
Affiliation(s)
- Pai-Chi Teng
- Department of Education and Research, Taipei City Hospital Renai Branch, Taipei, Taiwan,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA,California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Vatche G. Agopian
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Ting-Yi Lin
- Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taiwan,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Ju Dong Yang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA,Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA,Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA,corresponding author (Dr. Ju Dong Yang):
| |
Collapse
|
5
|
Sun N, Yang Y, Miao H, Redublo P, Liu H, Liu W, Huang YW, Teng PC, Zhang C, Zhang RY, Smalley M, Yang P, Chou SJ, Huai K, Zhang Z, Lee YT, Wang JJ, Wang J, Liang IY, Zhang TX, Zhang D, Liang L, Weiss PS, Posadas EM, Donahue T, Hecht JR, Allen-Auerbach MS, Bergsland EK, Hope TA, Pei R, Zhu Y, Tseng HR, Heaney AP. Discovery and characterization of circulating tumor cell clusters in neuroendocrine tumor patients using nanosubstrate-embedded microchips. Biosens Bioelectron 2022; 199:113854. [PMID: 34896918 PMCID: PMC8900541 DOI: 10.1016/j.bios.2021.113854] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/25/2021] [Accepted: 11/27/2021] [Indexed: 01/19/2023]
Abstract
Circulating tumor cell (CTC) clusters are present in cancer patients with severe metastasis, resulting in poor clinical outcomes. However, CTC clusters have not been studied as extensively as single CTCs, and the clinical utility of CTC clusters remains largely unknown. In this study, we aim sought to explore the feasibility of NanoVelcro Chips to simultaneously detect both single CTCs and CTC clusters with negligible perturbation to their intrinsic properties in neuroendocrine tumors (NETs). We discovered frequent CTC clusters in patients with advanced NETs and examined their potential roles, together with single NET CTCs, as novel biomarkers of patient response following peptide receptor radionuclide therapy (PRRT). We observed dynamic changes in both total NET CTCs and NET CTC cluster counts in NET patients undergoing PRRT which correlated with clinical outcome. These preliminary findings suggest that CTC clusters, along with single CTCs, offer a potential non-invasive option to monitor the treatment response in NET patients undergoing PRRT.
Collapse
Affiliation(s)
- Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States; Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215123, PR China
| | - Yingying Yang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States; Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Hui Miao
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States; Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, PR China
| | - Peter Redublo
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Hongtao Liu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Wenfei Liu
- Department of Chemistry and Biochemistry, Department of Bioengineering, Department of Materials Science and Engineering, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, United States
| | - Yen-Wen Huang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Pai-Chi Teng
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, United States
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States; Department of Pathology, Southern Medical University, Guangzhou, 510515, Guangdong Province, PR China
| | - Ryan Y Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Peng Yang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Shih-Jie Chou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Kevin Huai
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Zhicheng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Jasmine J Wang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, United States
| | - Jing Wang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Icy Y Liang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Tiffany X Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Dongyun Zhang
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Li Liang
- Department of Pathology, Southern Medical University, Guangzhou, 510515, Guangdong Province, PR China
| | - Paul S Weiss
- Department of Chemistry and Biochemistry, Department of Bioengineering, Department of Materials Science and Engineering, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, United States
| | - Edwin M Posadas
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, United States
| | - Timothy Donahue
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - J Randolph Hecht
- Department of Medicine, Division of Hematology Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Martin S Allen-Auerbach
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States
| | - Emily K Bergsland
- Department of Clinical Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, 94158, United States
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, United States
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215123, PR China.
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States.
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States.
| | - Anthony P Heaney
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, United States.
| |
Collapse
|
6
|
Mundekkad D, Cho WC. Nanoparticles in Clinical Translation for Cancer Therapy. Int J Mol Sci 2022; 23:1685. [PMID: 35163607 PMCID: PMC8835852 DOI: 10.3390/ijms23031685] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
The advent of cancer therapeutics brought a paradigm shift from conventional therapy to precision medicine. The new therapeutic modalities accomplished through the properties of nanomaterials have extended their scope in cancer therapy beyond conventional drug delivery. Nanoparticles can be channeled in cancer therapy to encapsulate active pharmaceutical ingredients and deliver them to the tumor site in a more efficient manner. This review enumerates various types of nanoparticles that have entered clinical trials for cancer treatment. The obstacles in the journey of nanodrug from clinic to market are reviewed. Furthermore, the latest developments in using nanoparticles in cancer therapy are also highlighted.
Collapse
Affiliation(s)
- Deepa Mundekkad
- Centre for NanoBioTechnology (CNBT), Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India;
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| |
Collapse
|
7
|
Lee YT, Sun N, Kim M, Wang JJ, Tran BV, Zhang RY, Qi D, Zhang C, Chen PJ, Sadeghi S, Finn RS, Saab S, Han SHB, Busuttil RW, Pei R, Zhu Y, Tseng HR, You S, Yang JD, Agopian VG. Circulating Tumor Cell-Based Messenger RNA Scoring System for Prognostication of Hepatocellular Carcinoma: Translating Tissue-Based Messenger RNA Profiling Into a Noninvasive Setting. Liver Transpl 2022; 28:200-214. [PMID: 34664394 PMCID: PMC8820407 DOI: 10.1002/lt.26337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/15/2021] [Accepted: 10/08/2021] [Indexed: 02/03/2023]
Abstract
Numerous studies in hepatocellular carcinoma (HCC) have proposed tissue-based gene signatures for individualized prognostic assessments. Here, we develop a novel circulating tumor cell (CTC)-based transcriptomic profiling assay to translate tissue-based messenger RNA (mRNA) signatures into a liquid biopsy setting for noninvasive HCC prognostication. The HCC-CTC mRNA scoring system combines the NanoVelcro CTC Assay for enriching HCC CTCs and the NanoString nCounter platform for quantifying the HCC-CTC Risk Score (RS) panel in enriched HCC CTCs. The prognostic role of the HCC-CTC RS was assessed in The Cancer Genome Atlas (TCGA) HCC cohort (n = 362) and validated in an independent clinical CTC cohort (n = 40). The HCC-CTC RS panel was developed through our integrated data analysis framework of 8 HCC tissue-based gene signatures and identified the top 10 prognostic genes (discoidin domain receptor tyrosine kinase 1 [DDR1], enoyl-CoA hydratase and 3-hydroxyacyl CoA dehydrogenase [EHHADH], androgen receptor [AR], lumican [LUM], hydroxysteroid 17-beta dehydrogenase 6[HSD17B6], prostate transmembrane protein, androgen induced 1 [PMEPA1], tsukushi, small leucine rich proteoglycan [TSKU], N-terminal EF-hand calcium binding protein 2 [NECAB2], ladinin 1 [LAD1], solute carrier family 27 member 5 [SLC27A5]) highly expressed in HCC with low expressions in white blood cells. The panel accurately discriminated overall survival in TCGA HCC cohort (hazard ratio [HR], 2.0; 95% confidence interval [CI], 1.4-2.9). The combined use of the scoring system and HCC-CTC RS panel successfully distinguished artificial blood samples spiked with an aggressive HCC cell type, SNU-387, from those spiked with PLC/PRF/5 cells (P = 0.02). In the CTC validation cohort (n = 40), HCC-CTC RS remained an independent predictor of survival (HR, 5.7; 95% CI, 1.5-21.3; P = 0.009) after controlling for Model for End-Stage Liver Disease score, Barcelona Clinic Liver Cancer stage, and CTC enumeration count. Our study demonstrates a novel interdisciplinary approach to translate tissue-based gene signatures into a liquid biopsy setting. This noninvasive approach will allow real-time disease profiling and dynamic prognostication of HCC.
Collapse
Affiliation(s)
- Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA,Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, P.R. China
| | - Minhyung Kim
- Division of Cancer Biology and Therapeutics, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jasmine J. Wang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Benjamin V. Tran
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA
| | - Ryan Y. Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Pin-Jung Chen
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Saeed Sadeghi
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA,Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Richard S. Finn
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA,Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Sammy Saab
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Steven-Huy B. Han
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Ronald W. Busuttil
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, P.R. China
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Ju Dong Yang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA,Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA,Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Vatche G. Agopian
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
8
|
Identification of DNA Damage Repair-Associated Prognostic Biomarkers for Prostate Cancer Using Transcriptomic Data Analysis. Int J Mol Sci 2021; 22:ijms222111771. [PMID: 34769200 PMCID: PMC8584064 DOI: 10.3390/ijms222111771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 02/08/2023] Open
Abstract
In the recent decade, the importance of DNA damage repair (DDR) and its clinical application have been firmly recognized in prostate cancer (PC). For example, olaparib was just approved in May 2020 to treat metastatic castration-resistant PC with homologous recombination repair-mutated genes; however, not all patients can benefit from olaparib, and the treatment response depends on patient-specific mutations. This highlights the need to understand the detailed DDR biology further and develop DDR-based biomarkers. In this study, we establish a four-gene panel of which the expression is significantly associated with overall survival (OS) and progression-free survival (PFS) in PC patients from the TCGA-PRAD database. This panel includes DNTT, EXO1, NEIL3, and EME2 genes. Patients with higher expression of the four identified genes have significantly worse OS and PFS. This significance also exists in a multivariate Cox regression model adjusting for age, PSA, TNM stages, and Gleason scores. Moreover, the expression of the four-gene panel is highly correlated with aggressiveness based on well-known PAM50 and PCS subtyping classifiers. Using publicly available databases, we successfully validate the four-gene panel as having the potential to serve as a prognostic and predictive biomarker for PC specifically based on DDR biology.
Collapse
|
9
|
Yoon J, Kim M, Posadas EM, Freedland SJ, Liu Y, Davicioni E, Den RB, Trock BJ, Karnes RJ, Klein EA, Freeman MR, You S. A comparative study of PCS and PAM50 prostate cancer classification schemes. Prostate Cancer Prostatic Dis 2021; 24:733-742. [PMID: 33531653 PMCID: PMC8326303 DOI: 10.1038/s41391-021-00325-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/20/2020] [Accepted: 01/15/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Two prostate cancer (PC) classification methods based on transcriptome profiles, a de novo method referred to as the "Prostate Cancer Classification System" (PCS) and a variation of the established PAM50 breast cancer algorithm, were recently proposed. Both studies concluded that most human PC can be assigned to one of three tumor subtypes, two categorized as luminal and one as basal, suggesting the two methods reflect consistency in underlying biology. Despite the similarity, differences and commonalities between the two classification methods have not yet been reported. METHODS Here, we describe a comparison of the PCS and PAM50 classification systems. PCS and PAM50 signatures consisting of 37 (PCS37) and 50 genes, respectively, were used to categorize 9,947 PC patients into PCS and PAM50 classes. Enrichment of hallmark gene sets and luminal and basal marker gene expression were assessed in the same datasets. Finally, survival analysis was performed to compare PCS and PAM50 subtypes in terms of clinical outcomes. RESULTS PCS and PAM50 subtypes show clear differential expression of PCS37 and PAM50 genes. While only three genes are shared in common between the two systems, there is some consensus between three subtype pairs (PCS1 versus Luminal B, PCS2 versus Luminal A, and PCS3 versus Basal) with respect to gene expression, cellular processes, and clinical outcomes. PCS categories displayed better separation of cellular processes and luminal and basal marker gene expression compared to PAM50. Although both PCS1 and Luminal B tumors exhibited the worst clinical outcomes, outcomes between aggressive and less aggressive subtypes were better defined in the PCS system, based on larger hazard ratios observed. CONCLUSION The PCS and PAM50 classification systems are similar in terms of molecular profiles and clinical outcomes. However, the PCS system exhibits greater separation in multiple clinical outcomes and provides better separation of prostate luminal and basal characteristics.
Collapse
Affiliation(s)
- Junhee Yoon
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Minhyung Kim
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Edwin M Posadas
- Urologic Oncology Program & Uro-Oncology Research Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen J Freedland
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Urology, Department of Surgery, Veteran Affairs Healthcare System, Durham, NC, USA
| | - Yang Liu
- Decipher Biosciences Inc., San Diego, CA, USA
| | | | - Robert B Den
- Department of Radiation Oncology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA, USA
| | - Bruce J Trock
- James Buchanan Brady Urological Institute, Johns Hopkins Hospital, Baltimore, MD, USA
| | | | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Michael R Freeman
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Pollan SG, Teng PC, Jan YJ, Livingstone J, Huang C, Kim M, Mariscal J, Rodriguez M, Chen JF, You S, DiVizio D, Boutros PC, Chan KS, Rasorenova O, Cress A, Spassov D, Moasser M, Posadas EM, Freedland SJ, Freeman MR, Zheng JJ, Knudsen BS. Loss of CDCP1 triggers FAK activation in detached prostate cancer cells. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2021; 9:350-366. [PMID: 34541033 PMCID: PMC8446766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
A major metastasis suppressing mechanism is the rapid apoptotic death of cancer cells upon detachment from extracellular matrix, a process called anoikis. Focal adhesion kinase (PTK2/FAK) is a key enzyme involved in evasion of anoikis. We show that loss of the Cub-domain containing protein-1 (CDCP1), paradoxically stimulates FAK activation in the detached state of prostate cancer cells. In CDCP1low DU145 and PC3 prostate cancer cells, detachment-activation of FAK occurs through local production of PI(4,5)P2. PI(4,5)P2 is generated by the PIP5K1c-201 splicing isoform of PIP5K1c, which contains a unique SRC phosphorylation site. In the detached state, reduced expression of CDCP1 and an alternative CDCP1-independent SRC activation mechanism triggers PIP5K1c-pY644 phosphorylation by SRC. This causes a switch of Talin binding from β1-integrin to PIP5K1c-pY644 and leads to activation of PIP5K1c-FAK. Reduced CDCP1 expression also inactivates CDK5, a negative regulator of PIP5K1c. Furthermore, immersion of prostate cancer cells in 10% human plasma or fetal bovine serum is required for activation of PIP5K1c-FAK. The PIP5K1c induced detachment-activation of FAK in preclinical models sensitizes CDCP1low prostate cancer cells to FAK inhibitors. In patients, CDCP1High versus CDCP1low circulating tumor cells differ in expression of AR-v7, ONECUT2 and HOXB13 oncogenes and TMPRSS2 and display intra-patient heterogeneity of FAK-pY397 expression. Taken together, CDCP1low and CDCP1high detached prostate cancer cells activate distinct cytoplasmic kinase complexes and targetable transcription factors, which has important therapeutic implications.
Collapse
Affiliation(s)
- Sara G Pollan
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Pai-Chi Teng
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Yu Jen Jan
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Julie Livingstone
- Department of Informatics and Biocomputing, Ontario Institute for Cancer ResearchToronto, ON M5G 1L7, Canada
| | - Cai Huang
- Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky789 South Limestone St, Lexington, KY 40536, USA
| | - Minhyung Kim
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Javier Mariscal
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Maria Rodriguez
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Jie-Fu Chen
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Dolores DiVizio
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Paul C Boutros
- Department of Human Genetics and Urology, Jonsson Comprehensive Cancer Centre, University of CaliforniaLos Angeles, CA, USA
| | - Keith Syson Chan
- Department of Pathology, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Olga Rasorenova
- Department of Molecular Biology and Biochemistry, University of California IrvineIrvine, CA 92697, USA
| | - Anne Cress
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine1501 N, Campbell Avenue, Tucson, AZ 85724, USA
| | - Danislav Spassov
- Department of Medicine, University of California San FranciscoSan Francisco, CA 94143, USA
| | - Mark Moasser
- Department of Medicine, University of California San FranciscoSan Francisco, CA 94143, USA
| | - Edwin M Posadas
- Department of Medicine, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Stephen J Freedland
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Michael R Freeman
- Department of Surgery, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Jie J Zheng
- Department of Cell & Developmental Biology, University of California Los AngelesCHS BH-973B, Los Angeles, CA 90095, USA
| | - Beatrice S Knudsen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center8700 Beverly Blvd, Los Angeles, CA 90048, USA
- Department of Pathology, University of UtahSalt Lake City, UT 84112, USA
| |
Collapse
|
11
|
Sun N, Lee YT, Kim M, Wang JJ, Zhang C, Teng PC, Qi D, Zhang RY, Tran BV, Lee YT, Ye J, Palomique J, Nissen NN, Han SHB, Sadeghi S, Finn RS, Saab S, Busuttil RW, Posadas EM, Liang L, Pei R, Yang JD, You S, Agopian VG, Tseng HR, Zhu Y. Covalent Chemistry-Mediated Multimarker Purification of Circulating Tumor Cells Enables Noninvasive Detection of Molecular Signatures of Hepatocellular Carcinoma. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2001056. [PMID: 34212072 PMCID: PMC8240468 DOI: 10.1002/admt.202001056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Indexed: 05/02/2023]
Abstract
Transcriptomic profiling of tumor tissues introduces a large database, which has led to improvements in the ability of cancer diagnosis, treatment, and prevention. However, performing tumor transcriptomic profiling in the clinical setting is very challenging since the procurement of tumor tissues is inherently limited by invasive sampling procedures. Here, we demonstrated the feasibility of purifying hepatocellular carcinoma (HCC) circulating tumor cells (CTCs) from clinical patient samples with improved molecular integrity using Click Chips in conjunction with a multimarker antibody cocktail. The purified CTCs were then subjected to mRNA profiling by NanoString nCounter platform, targeting 64 HCC-specific genes, which were generated from an integrated data analysis framework with 8 tissue-based prognostic gene signatures from 7 publicly available HCC transcriptomic studies. After bioinformatics analysis and comparison, the HCC CTC-derived gene signatures showed high concordance with HCC tissue-derived gene signatures from TCGA database, suggesting that HCC CTCs purified by Click Chips could enable the translation of HCC tissue molecular profiling into a noninvasive setting.
Collapse
Affiliation(s)
- Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Minhyung Kim
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jasmine J Wang
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ceng Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Pai-Chi Teng
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dongping Qi
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Ryan Y Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Benjamin V Tran
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Yue Tung Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Jinglei Ye
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Juvelyn Palomique
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nicholas N Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Steven-Huy B Han
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Saeed Sadeghi
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Richard S Finn
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Sammy Saab
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Ronald W Busuttil
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Edwin M Posadas
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, P.R. China
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, P.R. China
| | - Ju Dong Yang
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sungyong You
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vatche G Agopian
- Department of Surgery, UCLA, 200 Medical Plaza, Los Angeles, CA, 90024, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), 570 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
12
|
Li Y, Zheng Y, Wu L, Li J, Ji J, Yu Q, Dai W, Feng J, Wu J, Guo C. Current status of ctDNA in precision oncology for hepatocellular carcinoma. J Exp Clin Cancer Res 2021; 40:140. [PMID: 33902698 PMCID: PMC8074474 DOI: 10.1186/s13046-021-01940-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/06/2021] [Indexed: 01/12/2023] Open
Abstract
The conventional method used to obtain a tumor biopsy for hepatocellular carcinoma (HCC) is invasive and does not evaluate dynamic cancer progression or assess tumor heterogeneity. It is thus imperative to create a novel non-invasive diagnostic technique for improvement in cancer screening, diagnosis, treatment selection, response assessment, and predicting prognosis for HCC. Circulating tumor DNA (ctDNA) is a non-invasive liquid biopsy method that reveals cancer-specific genetic and epigenetic aberrations. Owing to the development of technology in next-generation sequencing and PCR-based assays, the detection and quantification of ctDNA have greatly improved. In this publication, we provide an overview of current technologies used to detect ctDNA, the ctDNA markers utilized, and recent advances regarding the multiple clinical applications in the field of precision medicine for HCC.
Collapse
Affiliation(s)
- Yan Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, number 1291, Jiangning road, Putuo, Shanghai, 200060, China.
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang road, Jing'an, Shanghai, 200072, China.
| |
Collapse
|
13
|
Giménez-Capitán A, Bracht J, García JJ, Jordana-Ariza N, García B, Garzón M, Mayo-de-Las-Casas C, Viteri-Ramirez S, Martinez-Bueno A, Aguilar A, Sullivan IG, Johnson E, Huang CY, Gerlach JL, Warren S, Beechem JM, Teixidó C, Rosell R, Reguart N, Molina-Vila MA. Multiplex Detection of Clinically Relevant Mutations in Liquid Biopsies of Cancer Patients Using a Hybridization-Based Platform. Clin Chem 2021; 67:554-563. [PMID: 33439966 DOI: 10.1093/clinchem/hvaa248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/30/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND With the advent of precision oncology, liquid biopsies are quickly gaining acceptance in the clinical setting. However, in some cases, the amount of DNA isolated is insufficient for Next-Generation Sequencing (NGS) analysis. The nCounter platform could be an alternative, but it has never been explored for detection of clinically relevant alterations in fluids. METHODS Circulating-free DNA (cfDNA) was purified from blood, cerebrospinal fluid, and ascites of patients with cancer and analyzed with the nCounter 3 D Single Nucleotide Variant (SNV) Solid Tumor Panel, which allows for detection of 97 driver mutations in 24 genes. RESULTS Validation experiments revealed that the nCounter SNV panel could detect mutations at allelic fractions of 0.02-2% in samples with ≥5 pg mutant DNA/µL. In a retrospective analysis of 70 cfDNAs from patients with cancer, the panel successfully detected EGFR, KRAS, BRAF, PIK3CA, and NRAS mutations when compared with previous genotyping in the same liquid biopsies and paired tumor tissues [Cohen kappa of 0.96 (CI = 0.92-1.00) and 0.90 (CI = 0.74-1.00), respectively]. In a prospective study including 91 liquid biopsies from patients with different malignancies, 90 yielded valid results with the SNV panel and mutations in EGFR, KRAS, BRAF, PIK3CA, TP53, NFE2L2, CTNNB1, ALK, FBXW7, and PTEN were found. Finally, serial liquid biopsies from a patient with NSCLC revealed that the semiquantitative results of the mutation analysis by the SNV panel correlated with the evolution of the disease. CONCLUSIONS The nCounter platform requires less DNA than NGS and can be employed for routine mutation testing in liquid biopsies of patients with cancer.
Collapse
Affiliation(s)
- Ana Giménez-Capitán
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Jillian Bracht
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain.,Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Juan José García
- Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Núria Jordana-Ariza
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Beatriz García
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Mónica Garzón
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Clara Mayo-de-Las-Casas
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | | | | | - Andrés Aguilar
- Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain
| | | | | | | | | | | | | | - Cristina Teixidó
- Department of Pathology, Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Rafael Rosell
- Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain.,Cancer Biology and Precision Medicine Program, Catalán Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Badalona, Barcelona, Spain
| | - Noemí Reguart
- Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Medical Oncology, Thoracic Oncology Unit, Hospital Clínic, Barcelona, Spain
| | - Miguel A Molina-Vila
- Pangaea Oncology, Laboratory of Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| |
Collapse
|
14
|
Research Progress for the Clinical Application of Circulating Tumor Cells in Prostate Cancer Diagnosis and Treatment. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6230826. [PMID: 33506020 PMCID: PMC7814947 DOI: 10.1155/2021/6230826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Prostate cancer is a life-threatening and highly heterogeneous malignancy. In the past decade, circulating tumor cells (CTCs) have been suggested to play a critical role in the occurrence and progression of prostate cancer. In particular, as the “seed” of the cancer metastasis cascade, CTCs determine numerous biological behaviors, such as tumor invasion into adjacent tissues and migration to distant organs. Many studies have shown that CTCs are necessary in the processes of tumor progression, including tumorigenesis, invasion, metastasis, and colonization. Furthermore, CTCs express various biomarkers relevant to prostate cancer and thus can be applied clinically in noninvasive tests. Moreover, CTCs can serve as potential prognostic targets in prostate cancer due to their roles in regulating many processes associated with cancer metastasis. In this review, we discuss the isolation and detection of CTCs as predictive markers of prostate cancer, and we discuss their clinical application in the diagnosis and prognosis of prostate cancer and in monitoring the response to treatment and the prediction of metastasis.
Collapse
|
15
|
Wang J, Sun N, Lee YT, Ni Y, Koochekpour R, Zhu Y, Tseng HR, Wang S, Jiang L, Zhu H. A circulating tumor cell-based digital assay for the detection of EGFR T790M mutation in advanced non-small cell lung cancer. J Mater Chem B 2020; 8:5636-5644. [PMID: 32525199 PMCID: PMC8136811 DOI: 10.1039/d0tb00589d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Determining the status of epidermal growth factor receptor (EGFR) T790M mutation is crucial for guiding further treatment intervention in advanced non-small cell lung cancer (NSCLC) patients who develop acquired resistance to initial EGFR tyrosine kinase inhibitor (TKI) treatment. Circulating tumor cells (CTCs) which contain plentiful copies of well-preserved RNA offer an ideal source for noninvasive detection of T790M mutation in NSCLC. We developed a CTC-based digital assay which synergistically integrates NanoVelcro Chips for enriching NSCLC CTCs and reverse-transcription droplet digital PCR (RT-ddPCR) for quantifying T790M transcripts in the enriched CTCs. We collected 46 peripheral arterial and venous blood samples from 27 advanced NSCLC patients for testing this CTC-based digital assay. The results showed that the T790M mutational status observed by the CTC-based digital assay matched with those observed by tissue-based diagnostic methods. Furthermore, higher copy numbers of T790M transcripts were observed in peripheral arterial blood than those detected in the matched peripheral venous blood. In short, our results demonstrated the potential of the NanoVelcro CTC-digital assay for noninvasive detection of the T790M mutation in NSCLC, and suggested that peripheral arterial blood sampling may offer a more abundant CTC source than peripheral venous blood in advanced NSCLC patients.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, P. R. China. and California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Na Sun
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA and Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou 215123, P. R. China
| | - Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yiqian Ni
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China.
| | - Rose Koochekpour
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Shuyang Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, P. R. China.
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China.
| | - Hongguang Zhu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
16
|
Morrison GJ, Cunha AT, Jojo N, Xu Y, Xu Y, Kwok E, Robinson P, Dorff T, Quinn D, Carpten J, Manojlovic Z, Goldkorn A. Cancer transcriptomic profiling from rapidly enriched circulating tumor cells. Int J Cancer 2020; 146:2845-2854. [PMID: 32037533 DOI: 10.1002/ijc.32915] [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] [Received: 01/08/2020] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/26/2022]
Abstract
Transcriptomic profiling of metastatic cancer can illuminate mechanisms of progression and lead to new therapies, but standard biopsy is invasive and reflects only a single metastatic site. In contrast, circulating tumor cell (CTC) profiling is noninvasive and repeatable, reflecting the dynamic and systemic nature of advanced disease. To date, transcriptomic profiling of CTCs has not delivered on its full potential, because white blood cells (WBCs) vastly outnumber CTCs. Current profiling strategies either lack cancer sensitivity and specificity or require specialized CTC capture protocols that are not readily scalable to large patient cohorts. Here, we describe a new strategy for rapid CTC enrichment and transcriptomic profiling using commercially available WBC depletion, microfluidic enrichment and RNA sequencing. When applied to blood samples from patients with advanced prostate cancer (PC), transcriptomes from enriched samples cluster with cancer positive controls and previously undetectable prostate-specific transcripts become readily measurable. Gene set enrichment analysis reveals multiple significantly enriched signaling pathways associated with PC, as well as novel pathways that merit further study. This accessible and scalable approach yields cancer-specific transcriptomic data and can be applied repeatedly and noninvasively in large cancer patient cohorts to discover new therapeutic targets in advanced disease.
Collapse
Affiliation(s)
- Gareth J Morrison
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
| | - Alexander T Cunha
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
| | - Nita Jojo
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
| | - Yucheng Xu
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
| | - Yili Xu
- Department of Translational Genomics, USC Keck School of Medicine and NCCC, California, Los Angeles
| | - Eric Kwok
- Department of Translational Genomics, USC Keck School of Medicine and NCCC, California, Los Angeles
| | - Peggy Robinson
- Angle PLC, Surrey, United Kingdom
- Caza Health LLC, Earlysville, Virginia
| | - Tanya Dorff
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - David Quinn
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
| | - John Carpten
- Department of Translational Genomics, USC Keck School of Medicine and NCCC, California, Los Angeles
| | - Zarko Manojlovic
- Department of Translational Genomics, USC Keck School of Medicine and NCCC, California, Los Angeles
| | - Amir Goldkorn
- Department of Medicine, University of Southern California (USC), Keck School of Medicine and Norris Comprehensive Cancer Center (NCCC), California, Los Angeles
| |
Collapse
|
17
|
Velez EM, Desai B, Ji L, Quinn DI, Colletti PM, Jadvar H. Comparative prognostic implication of treatment response assessments in mCRPC: PERCIST 1.0, RECIST 1.1, and PSA response criteria. Am J Cancer Res 2020; 10:3254-3262. [PMID: 32194866 PMCID: PMC7053201 DOI: 10.7150/thno.39838] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Accurate appraisal of treatment response in metastatic castrate-resistant prostate cancer (mCRPC) is challenging in view of remarkable tumor heterogeneity and the available choices among many established and novel therapeutic approaches. The purpose of this single-center prospective study was to evaluate the comparative prognostic utility of PERCIST 1.0 in predicting overall survival (OS) in patients with mCRPC compared to RECIST 1.1 and prostate-specific antigen (PSA)-based treatment response assessments. Methods: Patients with mCRPC were prospectively enrolled if they were beginning systemic medical therapy or transitioning to new systemic therapy after not responding to a prior treatment. All patients underwent a baseline 18F-fluorodeoxyglucose (FDG) positron emission tomography/ computed tomography (PET/CT) prior to the initiation of treatment and again 4 months after the start of therapy. Patients' responses to treatment at 4 months compared to baseline were evaluated with RECIST 1.1, PERCIST 1.0 and PSA response criteria. The associations between patients' response categories and OS were evaluated. OS was defined as the duration in time between the date of baseline PET/CT to death from any cause. Patients with different response status were compared with logrank tests. Survival probabilities were calculated using the Kaplan-Meier method. Results: Patients with progressive disease by PSA response criteria at 4 months demonstrated significantly shorter OS (24-month OS probability: 18% ± 11%) compared to patients with stable disease, SD, (44% ± 19%, p=0.03) and complete response, CR, or partial response, PR, (53% ± 11%, p=0.03). RECIST 1.1 response criteria demonstrated a similar trend in OS, however no statistically significant differences were noted between patients with PD (25% ± 15%) compared to SD/non-CR, non-PD (54% ± 13%) and CR/PR (54% ± 14%) (p=0.13). PERCIST 1.0 criteria demonstrated significant differences in OS between responders, CMR/PMR (56% ± 12%), compared to SMD (38% ± 17%, p=0.03) and PMD (21% ± 10%, p=0.01). Patients with progressive disease by both PERICST 1.0 and PSA response criteria demonstrated significantly worse OS (24-month OS: 0%, 12-month OS: 31% ± 14%) compared to patients with progressive disease by either response criteria. Conclusion: PERCIST 1.0 may provide significant prognostic information for patients with mCRPC undergoing systemic chemotherapy, particularly when incorporated with PSA treatment response criteria.
Collapse
|
18
|
Dong J, Chen JF, Smalley M, Zhao M, Ke Z, Zhu Y, Tseng HR. Nanostructured Substrates for Detection and Characterization of Circulating Rare Cells: From Materials Research to Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903663. [PMID: 31566837 PMCID: PMC6946854 DOI: 10.1002/adma.201903663] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/02/2019] [Indexed: 05/03/2023]
Abstract
Circulating rare cells in the blood are of great significance for both materials research and clinical applications. For example, circulating tumor cells (CTCs) have been demonstrated as useful biomarkers for "liquid biopsy" of the tumor. Circulating fetal nucleated cells (CFNCs) have shown potential in noninvasive prenatal diagnostics. However, it is technically challenging to detect and isolate circulating rare cells due to their extremely low abundance compared to hematologic cells. Nanostructured substrates offer a unique solution to address these challenges by providing local topographic interactions to strengthen cell adhesion and large surface areas for grafting capture agents, resulting in improved cell capture efficiency, purity, sensitivity, and reproducibility. In addition, rare-cell retrieval strategies, including stimulus-responsiveness and additive reagent-triggered release on different nanostructured substrates, allow for on-demand retrieval of the captured CTCs/CFNCs with high cell viability and molecular integrity. Several nanostructured substrate-enabled CTC/CFNC assays are observed maturing from enumeration and subclassification to molecular analyses. These can one day become powerful tools in disease diagnosis, prognostic prediction, and dynamic monitoring of therapeutic response-paving the way for personalized medical care.
Collapse
Affiliation(s)
- Jiantong Dong
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jie-Fu Chen
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew Smalley
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Meiping Zhao
- Beijing National Laboratory for Molecular Sciences, MOE Key Laboratory of Bioorganic Chemistry and Molecular Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|