1
|
Behera P, Baidya S, Sahoo J, Jaiswal K, Singh DP, Pradhan S, Saini DK, Agasti SS, De M. Multistep Array-Based Sensing of Bioanalytes Using Modified MoS 2, Fluorescence Proteins, and Cucurbituril. ACS APPLIED BIO MATERIALS 2024; 7:6371-6381. [PMID: 39321472 DOI: 10.1021/acsabm.4c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
One pot sensor by multiplexing in the array is an attractive system for rapid discrimination of multiple analytes. Multiplexing can be achieved in two ways, i.e., using multiple signal transducers or adding sequential agents to the sensor media. Herein, we have used a combination of both multichannel and sequential ON-OFF strategies for the discrimination of different bioanalytes. The sensor array was constructed by implementing positively charged MoS2 as a receptor and different fluorescent proteins possessing distinguishable emission profiles as signal transducers. The sensing setup was constructed with the interaction between oppositely charged MoS2 and the host-guest combination between a cationic headgroup of MoS2 and Cucurbit [7] uril (CB7) to alter the fluorescence of signal transducers in situ noncovalently. Electrodynamic analysis and optical assays suggest that the electrostatic interaction played a major role in the modulation of the fluorescence outcomes in the array. Both cationic and anionic proteins were discriminated at a 50 nM concentration. The detection limit of the sensor array by using β-gal protein was found to be 1 nM. The sensor array was further implemented for the discrimination of normal and diseased cell lines and lysates, which indicates the versatile detection ability of this reported sensor array.
Collapse
Affiliation(s)
- Pradipta Behera
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Sourav Baidya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Jagabandhu Sahoo
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Komal Jaiswal
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Devendra Pratap Singh
- Department of Developmental Biology & Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Soumen Pradhan
- New Chemistry Unit, Chemistry & Physics of Materials Unit, and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Deepak Kumar Saini
- Department of Developmental Biology & Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Sarit S Agasti
- New Chemistry Unit, Chemistry & Physics of Materials Unit, and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Mrinmoy De
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
2
|
Alnajeebi AM, Alharbi HFH, Alelwani W, Babteen NA, Alansari WS, Shamlan G, Eskandrani AA. COVID-19 Candidate Genes and Pathways Potentially Share the Association with Lung Cancer. Comb Chem High Throughput Screen 2022; 25:2463-2472. [PMID: 34254909 DOI: 10.2174/1386207324666210712092649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 01/27/2023]
Abstract
COVID-19 is considered as the most challenging in the current situation but lung cancer is also the leading cause of death in the global population. These two malignancies are among the leading human diseases and are highly complex in terms of diagnostic and therapeutic approaches as well as the most frequent and highly complex and heterogeneous in nature. Based on the latest update, it is known that the patients suffering from lung cancer, are considered to be significantly at higher risk of COVID-19 infection in terms of survival and there are a number of evidences which support the hypothesis that these diseases may share the same functions and functional components. Multi-level unwanted alterations such as (epi-)genetic alterations, changes at the transcriptional level, and altered signaling pathways (receptor, cytoplasmic, and nuclear level) are the major sources which promote a number of complex diseases and such heterogeneous level of complexities are considered as the major barrier in the development of therapeutics. With so many challenges, it is critical to understand the relationships and the common shared aberrations between them which is difficult to unravel and understand. A simple approach has been applied for this study where differential gene expression analysis, pathway enrichment, and network level understanding are carried out. Since, gene expression changes and genomic alterations are related to the COVID-19 and lung cancer but their pattern varies significantly. Based on the recent studies, it appears that the patients suffering from lung cancer and and simultaneously infected with COVID-19, then survival chance is lessened. So, we have designed our goal to understand the genes commonly overexpressed and commonly enriched pathways in case of COVID-19 and lung cancer. For this purpose, we have presented the summarized review of the previous works where the pathogenesis of lung cancer and COVID-19 infection have been focused and we have also presented the new finding of our analysis. So, this work not only presents the review work but also the research work. This review and research study leads to the conclusion that growth promoting pathways (EGFR, Ras, and PI3K), growth inhibitory pathways (p53 and STK11), apoptotic pathways (Bcl- 2/Bax/Fas), and DDR pathways and genes are commonly and dominantly altered in both the cases COVID-19 and lung cancer.
Collapse
Affiliation(s)
- Afnan M Alnajeebi
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Hend F H Alharbi
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, KSA
| | - Walla Alelwani
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Nouf A Babteen
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Wafa S Alansari
- College of Science, Department of Biochemistry, University of Jeddah, Jeddah, Saudi Arabia
| | - Ghalia Shamlan
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Areej A Eskandrani
- Chemistry Department, Faculty of Science, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
3
|
Rahmanian M, Sartipzadeh Hematabad O, Askari E, Shokati F, Bakhshi A, Moghadam S, Olfatbakhsh A, Al Sadat Hashemi E, Khorsand Ahmadi M, Morteza Naghib S, Sinha N, Tel J, Eslami Amirabadi H, den Toonder JMJ, Majidzadeh-A K. A micropillar array-based microfluidic chip for label-free separation of circulating tumor cells: The best micropillar geometry? J Adv Res 2022; 47:105-121. [PMID: 35964874 PMCID: PMC10173300 DOI: 10.1016/j.jare.2022.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/22/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022] Open
Abstract
INTRODUCTION The information derived from the number and characteristics of circulating tumor cells (CTCs), is crucial to ensure appropriate cancer treatment monitoring. Currently, diverse microfluidic platforms have been developed for isolating CTCs from blood, but it remains a challenge to develop a low-cost, practical, and efficient strategy. OBJECTIVES This study aimed to isolate CTCs from the blood of cancer patients via introducing a new and efficient micropillar array-based microfluidic chip (MPA-Chip), as well as providing prognostic information and monitoring the treatment efficacy in cancer patients. METHODS We fabricated a microfluidic chip (MPA-Chip) containing arrays of micropillars with different geometries (lozenge, rectangle, circle, and triangle). We conducted numerical simulations to compare velocity and pressure profiles inside the micropillar arrays. Also, we experimentally evaluated the capture efficiency and purity of the geometries using breast and prostate cancer cell lines as well as a blood sample. Moreover, the device's performance was validated on 12 patients with breast cancer (BC) in different states. RESULTS The lozenge geometry was selected as the most effective and optimized micropillar design for CTCs isolation, providing high capture efficiency (>85 %), purity (>90 %), and viability (97 %). Furthermore, the lozenge MPA-chip was successfully validated by the detection of CTCs from 12 breast cancer (BC) patients, with non-metastatic (median number of 6 CTCs) and metastatic (median number of 25 CTCs) diseases, showing different prognoses. Also, increasing the chemotherapy period resulted in a decrease in the number of captured CTCs from 23 to 7 for the metastatic patient. The MPA-Chip size was only 0.25 cm2 and the throughput of a single chip was 0.5 ml/h, which can be increased by multiple MPA-Chips in parallel. CONCLUSION The lozenge MPA-Chip presented a novel micropillar geometry for on-chip CTC isolation, detection, and staining, and in the future, the possibilities can be extended to the culture of the CTCs.
Collapse
Affiliation(s)
- Mehdi Rahmanian
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Omid Sartipzadeh Hematabad
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Esfandyar Askari
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Farhad Shokati
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Atin Bakhshi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shiva Moghadam
- Breast Diseases Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Asiie Olfatbakhsh
- Breast Diseases Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Esmat Al Sadat Hashemi
- Breast Diseases Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Khorsand Ahmadi
- Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Nidhi Sinha
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jurjen Tel
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Hossein Eslami Amirabadi
- Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; AZAR Innovations, Utrecht, the Netherlands
| | - Jaap M J den Toonder
- Microsystems Research Section, Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
4
|
van de Stolpe A, Verhaegh W, Blay JY, Ma CX, Pauwels P, Pegram M, Prenen H, De Ruysscher D, Saba NF, Slovin SF, Willard-Gallo K, Husain H. RNA Based Approaches to Profile Oncogenic Pathways From Low Quantity Samples to Drive Precision Oncology Strategies. Front Genet 2021; 11:598118. [PMID: 33613616 PMCID: PMC7893109 DOI: 10.3389/fgene.2020.598118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Precision treatment of cancer requires knowledge on active tumor driving signal transduction pathways to select the optimal effective targeted treatment. Currently only a subset of patients derive clinical benefit from mutation based targeted treatment, due to intrinsic and acquired drug resistance mechanisms. Phenotypic assays to identify the tumor driving pathway based on protein analysis are difficult to multiplex on routine pathology samples. In contrast, the transcriptome contains information on signaling pathway activity and can complement genomic analyses. Here we present the validation and clinical application of a new knowledge-based mRNA-based diagnostic assay platform (OncoSignal) for measuring activity of relevant signaling pathways simultaneously and quantitatively with high resolution in tissue samples and circulating tumor cells, specifically with very small specimen quantities. The approach uses mRNA levels of a pathway's direct target genes, selected based on literature for multiple proof points, and used as evidence that a pathway is functionally activated. Using these validated target genes, a Bayesian network model has been built and calibrated on mRNA measurements of samples with known pathway status, which is used next to calculate a pathway activity score on individual test samples. Translation to RT-qPCR assays enables broad clinical diagnostic applications, including small analytes. A large number of cancer samples have been analyzed across a variety of cancer histologies and benchmarked across normal controls. Assays have been used to characterize cell types in the cancer cell microenvironment, including immune cells in which activated and immunotolerant states can be distinguished. Results support the expectation that the assays provide information on cancer driving signaling pathways which is difficult to derive from next generation DNA sequencing analysis. Current clinical oncology applications have been complementary to genomic mutation analysis to improve precision medicine: (1) prediction of response and resistance to various therapies, especially targeted therapy and immunotherapy; (2) assessment and monitoring of therapy efficacy; (3) prediction of invasive cancer cell behavior and prognosis; (4) measurement of circulating tumor cells. Preclinical oncology applications lie in a better understanding of cancer behavior across cancer types, and in development of a pathophysiology-based cancer classification for development of novel therapies and precision medicine.
Collapse
Affiliation(s)
| | | | - Jean-Yves Blay
- Medical Oncology, Université Claude Bernard Lyon 1, Lyon, France
- Centre Léon Bérard, Lyon, France
| | - Cynthia X. Ma
- Medicine, Division of Oncology, Section of Medical Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Patrick Pauwels
- Molecular Pathology, Centre for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Mark Pegram
- Stanford University School of Medicine, Clinical Research, Stanford Cancer Institute, Stanford, CA, United States
| | - Hans Prenen
- Oncology Department, Head of Phase I – Early Clinical Trials Unit, Clinical Trial Management Program, Oncology Department, Antwerp University Hospital, Antwerp, Belgium
| | - Dirk De Ruysscher
- Oncology-Radiotherapy, Maastro/Maastricht University Medical Center, Maastricht, Netherlands
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, GA, United States
- Head and Neck Medical Oncology Program, Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | | | | | - Hatim Husain
- University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
5
|
Papaccio F. Circulating cancer stem cells: an interesting niche to explore. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:253-258. [PMID: 36046778 PMCID: PMC9400729 DOI: 10.37349/etat.2020.00016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Federica Papaccio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy
| |
Collapse
|
6
|
Ye X, Li G, Han C, Han Q, Shang L, Su H, Han B, Gong Y, Lu G, Peng T. Circulating tumor cells as a potential biomarker for postoperative clinical outcome in HBV-related hepatocellular carcinoma. Cancer Manag Res 2018; 10:5639-5647. [PMID: 30532586 PMCID: PMC6245351 DOI: 10.2147/cmar.s175489] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND This study aimed to determine if the number of circulating tumor cells (CTCs) and changes in their numbers affected tumor recurrence and metastasis after surgical resection in patients with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). METHODS The primary endpoints were overall survival (OS) and progression-free survival (PFS). A total of 42 patients with HCC were selected from the First Affiliated Hospital of Guangxi Medical College from 2014 to 2017. CTCs were counted 1 day prior to and 30 days after surgical excision of HCC using the CanPatrol™ system. RESULTS Numbers of CTCs (> 2 CTCs and > 5 CTCs per 5 ml peripheral blood) were significantly associated with Edmondson stage in HBV-related HCC prior to surgery (P = 0.004 and 0.014, respectively). However there were no significant associations between other tested clinicopathological factors and CTC counts. Postoperative CTC counts (> 2 and > 5) and pre/postoperative change in CTC counts were significantly associated with PFS (P = 0.02, 0.009, and 0.001, respectively), but not with OS. Receiver operating characteristic curve analysis showed that pre/postoperative changes in the CTC count were a better predictor of performance than absolute count. The postoperative CTC count was also significantly associated with positive TP53 expression (P < 0.05). CONCLUSION These results demonstrate that postoperative CTC counts (> 2 and > 5) and changes in CTC counts may be independent prognostic indicators for PFS in patients with HBV-related HCC, with the change in number of CTCs showing better predictive performance.
Collapse
Affiliation(s)
- Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Guanghui Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Quanfa Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Liming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Bowen Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Yizhen Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China,
| | - Guodong Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi Province, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China, ,Correspondence: Tao Peng, Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, Guangxi Province, China, Tel +86 771 535 6528, Fax +86 771 535 0031, Email
| |
Collapse
|
7
|
Kolostova K, Rzechonek A, Schützner J, Grill R, Lischke R, Hladik P, Simonek J, Bobek V. Circulating Tumor Cells as an Auxiliary Diagnostic Tool in Surgery. ACTA ACUST UNITED AC 2018; 31:1197-1202. [PMID: 29102946 DOI: 10.21873/invivo.11190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/21/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND In general, the presence of circulating tumor cells (CTCs) in peripheral blood (PB) is associated with a relative shorter overall survival in cancer patients. The clinical utility of CTC diagnostics is changing: from prognostic test to an assay predicting therapy response, enabling the right choice of therapy and monitoring the effect of administered therapy. We present two case reports of patients with suspicion of lung and pancreatic cancer, without obtainable preoperative biopsy for histological verification. The focus of the presented study was not to deliver a complete tumor tissue classification to the surgeon, but to answer the question if there is malignant disease or not. The results are based on CTC presence and characterization. MATERIALS AND METHODS A size-based separation method for viable CTC enrichment from anticoagulated PB was used. The separated cells were cytomorphologically examined using vital fluorescent microscopy. Additionally, to confirm the epithelial origin of the cells on the separation membrane, CTC gene expression analysis was performed. RESULTS CTCs were successfully enriched and cultured in vitro in both tested samples. The epithelial character of the captured cells was confirmed by quantitative-polymerase chain reaction (qPCR) analysis for a set of tumor-associated genes. CONCLUSION Detection of cancer cells in PB (liquid biopsy) and their molecular characterization could significantly help complete the tumor diagnostic process in a time-efficient manner.
Collapse
Affiliation(s)
- Katarina Kolostova
- Department of Laboratory Genetics, Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Adam Rzechonek
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Jan Schützner
- Third Department of Surgery, First Faculty of Medicine Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Robert Grill
- Department of Laboratory Genetics, Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Robert Lischke
- Third Department of Surgery, First Faculty of Medicine Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Pavel Hladik
- Third Department of Surgery, First Faculty of Medicine Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Jan Simonek
- Third Department of Surgery, First Faculty of Medicine Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Vladimir Bobek
- Department of Laboratory Genetics, Kralovske Vinohrady University Hospital, Prague, Czech Republic .,Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland.,Third Department of Surgery, First Faculty of Medicine Charles University in Prague and Motol University Hospital, Prague, Czech Republic.,Department of Thoracic Surgery, Masaryk's Hospital in Ustinad Labem, Labem, Czech Republic
| |
Collapse
|
8
|
Mahato K, Kumar A, Maurya PK, Chandra P. Shifting paradigm of cancer diagnoses in clinically relevant samples based on miniaturized electrochemical nanobiosensors and microfluidic devices. Biosens Bioelectron 2018; 100:411-428. [DOI: 10.1016/j.bios.2017.09.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/03/2017] [Accepted: 09/03/2017] [Indexed: 02/08/2023]
|
9
|
Le NDB, Tonga GY, Mout R, Kim ST, Wille ME, Rana S, Dunphy KA, Jerry DJ, Yazdani M, Ramanathan R, Rotello CM, Rotello VM. Cancer Cell Discrimination Using Host-Guest "Doubled" Arrays. J Am Chem Soc 2017; 139:8008-8012. [PMID: 28535040 PMCID: PMC5848078 DOI: 10.1021/jacs.7b03657] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We report a nanosensor that uses cell lysates to rapidly profile the tumorigenicity of cancer cells. This sensing platform uses host-guest interactions between cucurbit[7]uril and the cationic headgroup of a gold nanoparticle to non-covalently modify the binding of three fluorescent proteins of a multi-channel sensor in situ. This approach doubles the number of output channels to six, providing single-well identification of cell lysates with 100% accuracy. Significantly, this classification could be extended beyond the training set, determining the invasiveness of novel cell lines. The unique fingerprint of these cell lysates required minimal sample quantity (200 ng, ∼1000 cells), making the methodology compatible with microbiopsy technology.
Collapse
Affiliation(s)
- Ngoc D. B. Le
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gulen Yesilbag Tonga
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Rubul Mout
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Sung-Tae Kim
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Department of Pharmaceutical Engineering, Inje University, 197, Inje-ro, Gimhae-si, Gyeongsangnam-do, Republic of Korea
| | - Marcos E. Wille
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Subinoy Rana
- Department of Materials, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Karen A. Dunphy
- Department of Veterinary and Animal Science, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - D. Joseph Jerry
- Department of Veterinary and Animal Science, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Mahdieh Yazdani
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Rajesh Ramanathan
- Ian Potter NanoBioSensing Facility, NanoBiotechnology Research Laboratory, School of Sciences, RMIT University GPO Box 2476 V, Melbourne, Victoria 3001, Australia
| | - Caren M. Rotello
- Department of Psychology and Brain Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
10
|
Mirza S, Jain N, Rawal R. Evidence for circulating cancer stem-like cells and epithelial-mesenchymal transition phenotype in the pleurospheres derived from lung adenocarcinoma using liquid biopsy. Tumour Biol 2017; 39:1010428317695915. [PMID: 28347243 DOI: 10.1177/1010428317695915] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Lung cancer stem cells are supposed to be the main drivers of tumor initiation, maintenance, drug resistance, and relapse of the disease. Hence, identification of the cellular and molecular aspects of these cells is a prerequisite for targeted therapy of lung cancer. Currently, analysis of circulating tumor cells has the potential to become the main diagnostic technique to monitor disease progression or therapeutic response as it is non-invasive. However, accurate detection of circulating tumor cells has remained a challenge, as epithelial cell markers used so far are not always trustworthy for detecting circulating tumor cells, especially during epithelial-mesenchymal transition. As cancer stem cells are the only culprit to initiate metastatic tumors, our aim was to isolate and characterize circulating tumor stem cells rather than circulating tumor cells from the peripheral blood of NSCLC adenocarcinoma as limited data are available addressing the gene expression profiling of lung cancer stem cells. Here, we reveal that CD44(+)/CD24(-) population in circulation not only exhibit stem cell-related genes but also possess epithelial-mesenchymal transition characteristics. In conclusion, the use of one or more cancer stem cell markers along with epithelial, mesenchymal and epithelial mesenchymal transition markers will prospectively provide the most precise assessment of the threat for recurrence and metastatic disease and has a great potential for forthcoming applications in harvesting circulating tumor stem cells and their downstream applications. Our results will aid in developing diagnostic and prognostic modalities and personalized treatment regimens like dendritic cell-based immunotherapy that can be utilized for targeting and eliminating circulating tumor stem cells, to significantly reduce the possibility of relapse and improve clinical outcomes.
Collapse
Affiliation(s)
- Sheefa Mirza
- 1 Department of Life Sciences, University School of Sciences, Gujarat University, Ahmedabad, India.,2 Division of Medicinal Chemistry and Pharmacogenomics, Department of Cancer Biology, The Gujarat Cancer & Research Institute, Ahmedabad, India
| | - Nayan Jain
- 1 Department of Life Sciences, University School of Sciences, Gujarat University, Ahmedabad, India
| | - Rakesh Rawal
- 2 Division of Medicinal Chemistry and Pharmacogenomics, Department of Cancer Biology, The Gujarat Cancer & Research Institute, Ahmedabad, India
| |
Collapse
|
11
|
Qasaimeh MA, Wu YC, Bose S, Menachery A, Talluri S, Gonzalez G, Fulciniti M, Karp JM, Prabhala RH, Karnik R. Isolation of Circulating Plasma Cells in Multiple Myeloma Using CD138 Antibody-Based Capture in a Microfluidic Device. Sci Rep 2017; 7:45681. [PMID: 28374831 PMCID: PMC5379479 DOI: 10.1038/srep45681] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/02/2017] [Indexed: 12/15/2022] Open
Abstract
The necessity for bone marrow aspiration and the lack of highly sensitive assays to detect residual disease present challenges for effective management of multiple myeloma (MM), a plasma cell cancer. We show that a microfluidic cell capture based on CD138 antigen, which is highly expressed on plasma cells, permits quantitation of rare circulating plasma cells (CPCs) in blood and subsequent fluorescence-based assays. The microfluidic device is based on a herringbone channel design, and exhibits an estimated cell capture efficiency of ~40–70%, permitting detection of <10 CPCs/mL using 1-mL sample volumes, which is difficult using existing techniques. In bone marrow samples, the microfluidic-based plasma cell counts exhibited excellent correlation with flow cytometry analysis. In peripheral blood samples, the device detected a baseline of 2–5 CD138+ cells/mL in healthy donor blood, with significantly higher numbers in blood samples of MM patients in remission (20–24 CD138+ cells/mL), and yet higher numbers in MM patients exhibiting disease (45–184 CD138+ cells/mL). Analysis of CPCs isolated using the device was consistent with serum immunoglobulin assays that are commonly used in MM diagnostics. These results indicate the potential of CD138-based microfluidic CPC capture as a useful ‘liquid biopsy’ that may complement or partially replace bone marrow aspiration.
Collapse
Affiliation(s)
- Mohammad A Qasaimeh
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE.,Mechanical and Aerospace Engineering Department, New York University, Brooklyn, NY 11201, USA.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yichao C Wu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Suman Bose
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anoop Menachery
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Srikanth Talluri
- VA Boston Healthcare System, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Jeffrey M Karp
- Division of BioEngineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard Stem Cell Institute, Harvard-MIT Division of Health Sciences and Technology, 65 Landsdowne St., Cambridge, MA 02139, USA
| | - Rao H Prabhala
- VA Boston Healthcare System, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA.,Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Rohit Karnik
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
12
|
Caceres G, Puskas JA, Magliocco AM. Circulating Tumor Cells: A Window Into Tumor Development and Therapeutic Effectiveness. Cancer Control 2016; 22:167-76. [PMID: 26068761 DOI: 10.1177/107327481502200207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are an important diagnostic tool for understanding the metastatic process and the development of cancer. METHODS This review covers the background, relevance, and potential limitations of CTCs as a measurement of cancer progression and how information derived from CTCs may affect treatment efficacy. It also highlights the difficulties of characterizing these rare cells due to the limited cell surface molecules unique to CTCs and each particular type of cancer. RESULTS The analysis of cancer in real time, through the measure of the number of CTCs in a " liquid" biopsy specimen, gives us the ability to monitor the therapeutic efficacy of treatments and possibly the metastatic potential of a tumor. CONCLUSIONS Through novel and innovative techniques yielding encouraging results, including microfluidic techniques, isolating and molecularly analyzing CTCs are becoming a reality. CTCs hold promise for understanding how tumors work and potentially aiding in their demise.
Collapse
Affiliation(s)
- Gisela Caceres
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | | | |
Collapse
|
13
|
Stadler M, Walter S, Walzl A, Kramer N, Unger C, Scherzer M, Unterleuthner D, Hengstschläger M, Krupitza G, Dolznig H. Increased complexity in carcinomas: Analyzing and modeling the interaction of human cancer cells with their microenvironment. Semin Cancer Biol 2015; 35:107-24. [DOI: 10.1016/j.semcancer.2015.08.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 02/08/2023]
|
14
|
Das DK, Naidoo MK, Ilboudo A, DuBois P, Durojaiye V, Liu C, Ogunwobi OO. Isolation and Propagation of Circulating Tumor Cells from a Mouse Cancer Model. J Vis Exp 2015. [PMID: 26484891 DOI: 10.3791/52861] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cancer metastasis is the foremost cause of cancer-associated deaths. Recent studies have shown that circulating tumor cells (CTCs) are important in cancer metastasis. Indeed, the number of CTCs correlates with tumor size. Here, a detailed description is provided of a methodology for isolation and propagation of CTCs from a syngeneic mouse model of hepatocellular carcinoma (HCC) which allows for downstream analysis of potentially important molecular mechanisms of solid organ tumor metastasis. This method is efficient and reproducible. It is a non-invasive technique and, therefore, has potential to replace the invasive biopsy of tissues from humans which may be associated with complications. Therefore, the method discussed here allows for the isolation and propagation of CTCs from whole blood samples such that they can be examined and characterized. This has potential for future adaptation for clinical applications such as diagnosis, and personalized targeted therapy.
Collapse
Affiliation(s)
- Dibash K Das
- Department of Biological Sciences, Hunter College of The City University of New York; Departments of Biology and Biochemistry, The Graduate Center of The City University of New York
| | - Michelle K Naidoo
- Department of Biological Sciences, Hunter College of The City University of New York
| | - Adeodat Ilboudo
- Department of Biological Sciences, Hunter College of The City University of New York
| | - Pascal DuBois
- Department of Biological Sciences, Hunter College of The City University of New York
| | - Victoria Durojaiye
- Department of Biological Sciences, Hunter College of The City University of New York
| | - Chen Liu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida
| | - Olorunseun O Ogunwobi
- Department of Biological Sciences, Hunter College of The City University of New York; Departments of Biology and Biochemistry, The Graduate Center of The City University of New York;
| |
Collapse
|
15
|
Dokmanovic M, Wu WJ. Monitoring Trastuzumab Resistance and Cardiotoxicity: A Tale of Personalized Medicine. Adv Clin Chem 2015; 70:95-130. [PMID: 26231486 DOI: 10.1016/bs.acc.2015.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
While approval of trastuzumab, a recombinant monoclonal antibody directed against HER2, along with a diagnostic kit to detect breast cancers which are positive for HER2 overexpression, has advanced a new era of stratified and personalized medicine, it also created several challenges to our scientific and clinical practice. These problems include trastuzumab resistance and trastuzumab-induced cardiotoxicity. In this review, we will summarize data from the literature regarding mechanisms of trastuzumab resistance and trastuzumab-induced cardiotoxicity and present some promising model systems that may advance our understanding of these mechanisms. Our discussion will include development of circulating tumor cells and circulating tumor DNA for monitoring tumor burden, of patient-derived xenograft models for preclinical testing of novel therapies, and of novel therapeutic strategies for trastuzumab-resistance and possible integration of these strategies in the design of co-clinical studies for testing in relevant patient subpopulations.
Collapse
|
16
|
Schwarzenbach H. The potential of circulating nucleic acids as components of companion diagnostics for predicting and monitoring chemotherapy response. Expert Rev Mol Diagn 2014; 15:267-75. [PMID: 25382372 DOI: 10.1586/14737159.2015.980817] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
An effective personalized medicine is associated with the ability of identifying cancer patients who respond to anticancer targeted therapies. Therefore, new companion biomarkers that facilitate drug development are urgently needed. Since clinically relevant genetic and epigenetic alterations can be detected in cell-free nucleic acids in the blood circulation of cancer patients, these molecules may be a new promising class of potential liquid biomarkers. They can be obtained in real-time from blood, and their analyses could, consequently, facilitate treatment decisions. Screening of these liquid biopsies may provide information on the aberrant signaling pathway that should be blocked by the chosen targeted therapy. This article will discuss the potential of circulating nucleic acids as therapeutics for overcoming chemotherapeutic resistance in anticancer strategies.
Collapse
Affiliation(s)
- Heidi Schwarzenbach
- Department of Tumour Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|