1
|
Hendriks J, Schasfoort RBM, Huskens J, Saris DF, Karperien M. Kinetic characterization of SPR-based biomarker assays enables quality control, calibration free measurements and robust optimization for clinical application. Anal Biochem 2022; 658:114918. [PMID: 36170905 DOI: 10.1016/j.ab.2022.114918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/27/2022]
Abstract
Biomarker measurements are essential for the early diagnosis of complex diseases. However, many current biomarker assays lack sensitivity and multiplexing capacity, work in a narrow detection range and importantly lack real time quality control opportunities, which hampers clinical translation. In this paper, we demonstrate a toolbox to kinetically characterize a biomarker measurement assay using Surface Plasmon Resonance imaging (SPRi) with ample opportunities for real time quality control by exploiting quantitative descriptions of the various biomolecular interactions. We show an accurate prediction of SPRi measurements at both low and high concentrations of various analytes with deviations <5% between actual measurements and predicted measurement. The biphasic binding sites model was accurate for fitting the experimental curves and enables optimal detection of heterophilic antibodies, cross-reactivity, spotting irregularities and/or other confounders. The toolbox can also be used to create a (simulated) calibration curve, enabling calibration-free measurements with good recovery, it allows for easy assay optimizations, and could help bridge the gap to bring new biomarker assays to the clinic.
Collapse
Affiliation(s)
- Jan Hendriks
- Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, the Netherlands
| | - Richard B M Schasfoort
- Medical Cell Biophysics, Technical Medical Centre, University of Twente, the Netherlands
| | - Jurriaan Huskens
- Molecular Nanofabrication, MESA+ Institute for Nanotechnology, University of Twente, the Netherlands
| | - DaniëlB F Saris
- Department of Orthopedics, Mayo Clinic, Rochester, MN, USA; Department of Orthopedics, UMC Utrecht, the Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, the Netherlands.
| |
Collapse
|
2
|
Zhou C, O'Connor J, Backen A, Valle JW, Bridgewater J, Dive C, Jayson GC. Plasma Tie2 trajectories identify vascular response criteria for VEGF inhibitors across advanced biliary tract, colorectal and ovarian cancers. ESMO Open 2022; 7:100417. [PMID: 35279528 PMCID: PMC9058891 DOI: 10.1016/j.esmoop.2022.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor inhibitors (VEGFi) are compromised by a lack of validated biomarkers. Previously we showed that changes in the concentration of plasma Tie2 (pTie2) was a response biomarker for bevacizumab. Here, we investigated whether pTie2 can predict response and progression cross-tumour for generic VEGFi treatment. PATIENTS AND METHODS Patients (n = 124) with advanced biliary tract cancer (ABC) received cisplatin/gemcitabine with cediranib or placebo (ABC-03 trial). Concentrations of pTie2 were measured longitudinally from before treatment until disease progression. Data from patients with ovarian cancer (n = 92, ICON7 trial) and patients with colorectal cancer (CRC) (n = 70, Travastin trial) were also included. RESULTS Cediranib-treated ABC patients were deconvoluted into distinct groups where in one group pTie2 trajectories resembled those seen in placebo-treated patients and in another pTie2 significantly reduced (t-test P = 2.7 × 10-14). Using the 95% confidence interval for these two groups, we defined a vascular complete response (vCR) as a 24% reduction in pTie2 within 9 weeks; vascular no response (vNR) as a 7% increase in pTie2, and a vascular partial response (between these limits). vCR cediranib-treated patients had significantly improved progression-free survival (8.8 versus 7.5 months, restricted mean ratio 0.73, P = 0.012) and overall survival (18.8 versus 12.1 months, hazard ratio 0.49, P = 0.02). By integrating data across ovarian cancer, CRC and ABC, we show that (i) patients with vNR do not benefit from VEGFi and (ii) Tie2-defined vascular progression occurs sufficiently in advance of radiological progressive disease that changes in treatment could be offered to prevent clinical deterioration. CONCLUSION pTie2 is the first cross-tumour, generic VEGFi, vascular response biomarker to guide optimum use of VEGFi in clinical practice.
Collapse
Affiliation(s)
- C Zhou
- CRUK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - J O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - A Backen
- Division of Cancer Sciences, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK
| | - J W Valle
- The Christie NHS Foundation Trust, Manchester, UK
| | - J Bridgewater
- University College Hospital Macmillan Cancer Centre, Huntley Street, London, UK
| | - C Dive
- CRUK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, UK
| | - G C Jayson
- Division of Cancer Sciences, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
3
|
Tobos CI, Kim S, Rissin DM, Johnson JM, Douglas S, Yan S, Nie S, Rice B, Sung KJ, Sikes HD, Duffy DC. Sensitivity and binding kinetics of an ultra-sensitive chemiluminescent enzyme-linked immunosorbent assay at arrays of antibodies. J Immunol Methods 2019; 474:112643. [PMID: 31401067 DOI: 10.1016/j.jim.2019.112643] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/07/2019] [Indexed: 12/29/2022]
Abstract
We have characterized the sensitivity and kinetics of a multiplex immunoassay system based on detection of chemiluminescence (CL) at arrays of antibodies. This enzyme-linked immunosorbent assay (ELISA) was based on the spotting of different antibodies in a circular pattern at the bottom of a well of a microtiter plate. Sandwich immunocomplexes within each spot were labeled with horse radish peroxidase, and CL was generated locally to each spot in the array from turnover of luminol substrate. CL from the arrays across the plate was collected in single images; long exposure times were used to maximize sensitivity, and short exposure times were used to extend the dynamic range at higher signals. Image analysis was used to determine the intensity of light from each spot in the array, and intensity was converted to concentration of protein via comparison to a calibration curve. To determine the intrinsic sensitivity of the CL ELISA array, streptavidin horseradish peroxidase (SA-HRP) was captured on an array spotted with biotinylated detection antibodies. The limit of detection (LOD) of SA-HRP was 105 aM, or 3200 enzymes per 50 μL. A single-plex assay for prostate specific antigen (PSA) was developed that had an LOD of 79 aM when the microtiter plate was shaken orbitally, comparable to the most sensitive immunoassays reported to date. Normalization of CL signals in the PSA assay to signal per molecule of SA-HRP showed that the efficiency of the shaken assay was ~40%. When the plates were not shaken, the efficiency was ~4.5%, i.e., ~9-fold lower than when shaken. To better understand the theoretical basis of the sensitivity of these assays, we developed COMSOL numerical models of the binding kinetics at the array for plates that were shaken orbitally and those not shaken. Experimental data from the orbitally shaken PSA assay were best modeled by inertial mixing in a three-layer system that included a 8-μm-thick concentration boundary layer. Experimental data from the unshaken PSA assay were well modeled by diffusion-limited kinetics. A single-plex assay for IL-10 was developed with an LOD of 69 aM or 1.5 fg/mL, and used to measure this cytokine in plasma and serum of 10 healthy individuals. A 5-plex assay for IL-5, IL-6, IL-10, IL-22, and TNF-α was developed with LODs of 56 aM, 237 aM, 69 aM, 88 aM, and 373 aM, respectively. The assay was used to measure these 5 cytokines in the plasma and serum of the same individuals. The correlation in concentration of IL-10 measured in single-plex and multiplex assays was good (r2 = 0.89; bias = 14.5%). The factors that result in the high sensitivity of CL ELISA arrays-mostly high signal to noise ratio of extended chemiluminescent imaging-are discussed. This multiplex CL ELISA could be used for sensitive profiling of multiple proteins for in vitro diagnostics and biomarker detection in the development of therapeutics.
Collapse
Affiliation(s)
- Carmen I Tobos
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Seunghyeon Kim
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - David M Rissin
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Joseph M Johnson
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Scott Douglas
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Susan Yan
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Shuai Nie
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Bradley Rice
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Ki-Joo Sung
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Hadley D Sikes
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - David C Duffy
- Quanterix Corporation, 900 Middlesex Turnpike, Billerica, MA 01821, USA.
| |
Collapse
|
4
|
Zhou C, Taylor S, Tugwood J, Simpson K, Jayson GC, Symonds P, Paul J, Davidson S, Carty K, McCartney E, Rai D, Dive C, West C. Dynamics of circulating vascular endothelial growth factor-A predict benefit from antiangiogenic cediranib in metastatic or recurrent cervical cancer patients. Br J Clin Pharmacol 2019; 85:1781-1789. [PMID: 30980733 PMCID: PMC6624436 DOI: 10.1111/bcp.13965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 03/04/2019] [Accepted: 03/27/2019] [Indexed: 02/04/2023] Open
Abstract
AIMS There is a need for predictive and surrogate response biomarkers to support treatment with antiangiogenic vascular endothelial growth factor (VEGF) inhibitors. We aimed to identify a minimally-invasive biomarker predicting benefit from cediranib pretreatment or early during treatment in patients with recurrent or metastatic cervical cancer. METHODS Blood samples were collected before treatment, during treatment and upon disease progression where appropriate from patients enrolled in CIRCCa, a randomised phase II trial of carboplatin and paclitaxel with or without cediranib. Plasma concentrations of VEGF-A, VEGF-receptor 2, Ang1 and Tie2 were measured using multiplex enzyme-linked immunosorbent assay. Pretreatment and temporal changes of the biomarkers were investigated using proportional hazard regression and unsupervised clustering analysis. RESULTS Samples (n = 556) from 52 patients were analysed. VEGF-receptor 2 (P = .0006) and Tie2 (P = .04) were downregulated following cediranib, while VEGF-A (P = .0025) was upregulated. High Eastern Cooperative Oncology Group performance status (P = .02, hazard ratio [HR] = 2.15, 95% confidence interval [CI] 1.13-4.09) and low pretreatment Tie2 concentrations (P = .003, HR = 0.57, 95%CI 0.39-0.83) were independent prognostic factors associated with reduced progression-free survival. Two patterns of changes in VEGF-A following cediranib were identified. Patients with elevated VEGF-A in the first 3 treatment cycles, regardless of magnitude, had reduced progression-free survival in the placebo arm but improved survival with the addition of cediranib (P = .019, HR = 0.13, 95% CI 0.02-0.71). CONCLUSION Patterns of early elevation in plasma VEGF-A should be studied further as a potential biomarker to predict treatment benefit from cediranib.
Collapse
Affiliation(s)
- Cong Zhou
- Division of Cancer SciencesUniversity of Manchester, Manchester Academic Health Science CentreManchesterUK
- Clinical & Experimental Pharmacology Group, Cancer Research UK Manchester Institute and Manchester Centre for Cancer Biomarker SciencesUniversity of ManchesterUK
| | - Sarah Taylor
- Clinical & Experimental Pharmacology Group, Cancer Research UK Manchester Institute and Manchester Centre for Cancer Biomarker SciencesUniversity of ManchesterUK
| | - Jonathan Tugwood
- Clinical & Experimental Pharmacology Group, Cancer Research UK Manchester Institute and Manchester Centre for Cancer Biomarker SciencesUniversity of ManchesterUK
| | - Kathryn Simpson
- Clinical & Experimental Pharmacology Group, Cancer Research UK Manchester Institute and Manchester Centre for Cancer Biomarker SciencesUniversity of ManchesterUK
| | - Gordon C. Jayson
- Division of Cancer SciencesUniversity of Manchester, Christie Hospital NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
| | - Paul Symonds
- Department of Cancer StudiesUniversity of LeicesterLeicesterUK
| | - James Paul
- Cancer Research UK Clinical Trials Unit, Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | | | - Karen Carty
- Cancer Research UK Clinical Trials Unit, Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Elaine McCartney
- Cancer Research UK Clinical Trials Unit, Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Debbie Rai
- Cancer Research UK Clinical Trials Unit, Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Caroline Dive
- Clinical & Experimental Pharmacology Group, Cancer Research UK Manchester Institute and Manchester Centre for Cancer Biomarker SciencesUniversity of ManchesterUK
| | - Catharine West
- Division of Cancer SciencesUniversity of Manchester, Christie Hospital NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
| |
Collapse
|
5
|
Marti FEM, Jayson GC, Manoharan P, O'Connor J, Renehan AG, Backen AC, Mistry H, Ortega F, Li K, Simpson KL, Allen J, Connell J, Underhill S, Misra V, Williams KJ, Stratford I, Jackson A, Dive C, Saunders MP. Novel phase I trial design to evaluate the addition of cediranib or selumetinib to preoperative chemoradiotherapy for locally advanced rectal cancer: the DREAMtherapy trial. Eur J Cancer 2019; 117:48-59. [PMID: 31229949 DOI: 10.1016/j.ejca.2019.04.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/21/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND The DREAMtherapy (Dual REctal Angiogenesis MEK inhibition radiotherapy) trial is a novel intertwined design whereby two tyrosine kinase inhibitors (cediranib and selumetinib) were independently evaluated with rectal chemoradiotherapy (CRT) in an efficient manner to limit the extended follow-up period often required for radiotherapy studies. PATIENTS AND METHODS Cediranib or selumetinib was commenced 10 days before and then continued with RT (45 Gy/25#/5 wks) and capecitabine (825 mg/m2 twice a day (BID)). When three patients in the cediranib 15-mg once daily (OD) cohort were in the surveillance period, recruitment to the selumetinib cohort commenced. This alternating schedule was followed throughout. Three cediranib (15, 20 and 30 mg OD) and two selumetinib cohorts (50 and 75 mg BID) were planned. Circulating and imaging biomarkers of inflammation/angiogenesis were evaluated. RESULTS In case of cediranib, dose-limiting diarrhoea, fatigue and skin reactions were seen in the 30-mg OD cohort, and therefore, 20 mg OD was defined as the maximum tolerated dose. Forty-one percent patients achieved a clinical or pathological complete response (7/17), and 53% (9/17) had an excellent clinical or pathological response (ECPR). Significantly lower level of pre-treatment plasma tumour necrosis factor alpha (TNFα) was found in patients who had an ECPR. In case of selumetinib, the 50-mg BID cohort was poorly tolerated (fatigue and diarrhoea); a reduced dose cohort of 75-mg OD was opened which was also poorly tolerated, and further recruitment was abandoned. Of the 12 patients treated, two attained an ECPR (17%). CONCLUSIONS This novel intertwined trial design is an effective way to independently investigate multiple agents with radiotherapy. The combination of cediranib with CRT was well tolerated with encouraging efficacy. TNFα emerged as a potential predictive biomarker of response and warrants further evaluation.
Collapse
Affiliation(s)
| | - G C Jayson
- The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - P Manoharan
- The Christie NHS Foundation Trust, Manchester, UK; Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - J O'Connor
- The Christie NHS Foundation Trust, Manchester, UK; Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - A G Renehan
- The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A C Backen
- The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - H Mistry
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - F Ortega
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - K Li
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - K L Simpson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - J Allen
- The Christie NHS Foundation Trust, Manchester, UK
| | - J Connell
- The Christie NHS Foundation Trust, Manchester, UK
| | - S Underhill
- The Christie NHS Foundation Trust, Manchester, UK
| | - V Misra
- The Christie NHS Foundation Trust, Manchester, UK
| | - K J Williams
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK; Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - I Stratford
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, M13 9PT, UK
| | - A Jackson
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - C Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - M P Saunders
- The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
6
|
Jayson GC, Zhou C, Backen A, Horsley L, Marti-Marti K, Shaw D, Mescallado N, Clamp A, Saunders MP, Valle JW, Mullamitha S, Braun M, Hasan J, McEntee D, Simpson K, Little RA, Watson Y, Cheung S, Roberts C, Ashcroft L, Manoharan P, Scherer SJ, Del Puerto O, Jackson A, O'Connor JPB, Parker GJM, Dive C. Plasma Tie2 is a tumor vascular response biomarker for VEGF inhibitors in metastatic colorectal cancer. Nat Commun 2018; 9:4672. [PMID: 30405103 PMCID: PMC6220185 DOI: 10.1038/s41467-018-07174-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 10/04/2018] [Indexed: 12/22/2022] Open
Abstract
Oncological use of anti-angiogenic VEGF inhibitors has been limited by the lack of informative biomarkers. Previously we reported circulating Tie2 as a vascular response biomarker for bevacizumab-treated ovarian cancer patients. Using advanced MRI and circulating biomarkers we have extended these findings in metastatic colorectal cancer (n = 70). Bevacizumab (10 mg/kg) was administered to elicit a biomarker response, followed by FOLFOX6-bevacizumab until disease progression. Bevacizumab induced a correlation between Tie2 and the tumor vascular imaging biomarker, Ktrans (R:-0.21 to 0.47) implying that Tie2 originated from the tumor vasculature. Tie2 trajectories were independently associated with pre-treatment tumor vascular characteristics, tumor response, progression free survival (HR for progression = 3.01, p = 0.00014; median PFS 248 vs. 348 days p = 0.0008) and the modeling of progressive disease (p < 0.0001), suggesting that Tie2 should be monitored clinically to optimize VEGF inhibitor use. A vascular response is defined as a 30% reduction in Tie2; vascular progression as a 40% increase in Tie2 above the nadir. Tie2 is the first, validated, tumor vascular response biomarker for VEGFi.
Collapse
Affiliation(s)
- Gordon C Jayson
- The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, M20 4BX, UK.
| | - Cong Zhou
- Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, M20 4GJ, UK
| | - Alison Backen
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute & Manchester Centre for Cancer Biomarker Sciences, Manchester, M20 4BX, UK
| | - Laura Horsley
- The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, M20 4BX, UK
| | - Kalena Marti-Marti
- The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, M20 4BX, UK
| | - Danielle Shaw
- Clatterbridge Cancer Centre, Liverpool, CH63 4JY, UK
| | - Nerissa Mescallado
- The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, M20 4BX, UK
| | - Andrew Clamp
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Mark P Saunders
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Juan W Valle
- The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, M20 4BX, UK
| | - Saifee Mullamitha
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Mike Braun
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Jurjees Hasan
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Delyth McEntee
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Kathryn Simpson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute & Manchester Centre for Cancer Biomarker Sciences, Manchester, M20 4BX, UK
| | - Ross A Little
- Imaging Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Yvonne Watson
- Imaging Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Susan Cheung
- Imaging Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Caleb Roberts
- Imaging Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Linda Ashcroft
- Manchester Academic Health Science Centre, Trials Co-ordination Unit, The Christie NHS Foundation Trust, Withington Hall Block C, Wilmslow Road, Manchester, M20 4BX, UK
| | - Prakash Manoharan
- The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, M20 4BX, UK
| | - Stefan J Scherer
- Novartis Pharmaceuticals Corporation, One Health Plaza, 337, East Hanover, NJ, 07936-1080, USA
| | - Olivia Del Puerto
- Del Puerto Limited, 23 Porters Wood; Saint Albans, Hertfordshire, AL3 6PQ, UK
| | - Alan Jackson
- Imaging Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - James P B O'Connor
- Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, M20 4GJ, UK
| | - Geoff J M Parker
- Imaging Sciences, University of Manchester, Manchester, M13 9PT, UK
- Bioxydyn Ltd, Manchester, M15 6SZ, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute & Manchester Centre for Cancer Biomarker Sciences, Manchester, M20 4BX, UK
| |
Collapse
|
7
|
Backen AC, Lopes A, Wasan H, Palmer DH, Duggan M, Cunningham D, Anthoney A, Corrie PG, Madhusudan S, Maraveyas A, Ross PJ, Waters JS, Steward WP, Rees C, McNamara MG, Beare S, Bridgewater JA, Dive C, Valle JW. Circulating biomarkers during treatment in patients with advanced biliary tract cancer receiving cediranib in the UK ABC-03 trial. Br J Cancer 2018; 119:27-35. [PMID: 29925934 PMCID: PMC6035166 DOI: 10.1038/s41416-018-0132-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/28/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Advanced biliary tract cancer (ABC) has a poor prognosis. Cediranib, in addition to cisplatin/gemcitabine [CisGem], improved the response rate, but did not improve the progression-free survival (PFS) in the ABC-03 study. Minimally invasive biomarkers predictive of cediranib benefit may improve patient outcomes. METHODS Changes in 15 circulating plasma angiogenesis or inflammatory-related proteins and cytokeratin-18 (CK18), measured at baseline and during therapy until disease progression, were correlated with overall survival (OS) using time-varying covariate Cox models (TVC). RESULTS Samples were available from n = 117/124 (94%) patients. Circulating Ang1&2, FGFb, PDGFbb, VEGFC, VEGFR1 and CK18 decreased as a result of the therapy, independent of treatment with cediranib. Circulating VEGFR2 and Tie2 were preferentially reduced by cediranib. Patients with increasing levels of VEGFA at any time had a worse PFS and OS; this detrimental effect was attenuated in patients receiving cediranib. TVC analysis revealed CK18 and VEGFR2 increases correlated with poorer OS in all patients (P < 0.001 and P = 0.02, respectively). CONCLUSIONS Rising circulating VEGFA levels in patients with ABC, treated with CisGem, are associated with worse PFS and OS, not seen in patients receiving cediranib. Rising levels of markers of tumour burden (CK18) and potential resistance (VEGFR2) are associated with worse outcomes and warrant validation.
Collapse
Affiliation(s)
- Alison C Backen
- Centre for Cancer Biomarker Sciences, Cancer Research UK Manchester Institute, Manchester, M20 4BX, UK
| | - Andre Lopes
- Cancer Research UK & University College London Cancer Trials Center, London, W1T 4TJ, UK
| | - Harpreet Wasan
- Hammersmith Hospital, Imperial College Healthcare Trust, London, W12 0HS, UK
| | - Daniel H Palmer
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool Cancer Research UK Center, Liverpool, L69 3GL, UK
| | - Marian Duggan
- Cancer Research UK & University College London Cancer Trials Center, London, W1T 4TJ, UK
| | | | - Alan Anthoney
- Leeds Cancer Research UK Clinical Center, Leeds, LS2 9JT, UK
| | - Pippa G Corrie
- Cambridge Cancer Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Srinivasan Madhusudan
- Division of Cancer & Stem Cells, University of Nottingham, Nottingham University Hospitals, Nottingham, NG7 2UH, UK
| | | | - Paul J Ross
- Department of Oncology, King's College Hospital, London, SE5 9RS, UK
| | | | | | - Charlotte Rees
- University Hospital Southampton NHS Foundation Trust, Southampton, SO16 6YD, UK
- Hampshire Hospitals NHS Foundation Trust, Basingstoke, RG24 9NA, UK
| | - Mairéad G McNamara
- Division of Cancer Sciences, University of Manchester, Manchester, M13 9PL, UK
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Sandy Beare
- Cancer Research UK & University College London Cancer Trials Center, London, W1T 4TJ, UK
| | | | - Caroline Dive
- Centre for Cancer Biomarker Sciences, Cancer Research UK Manchester Institute, Manchester, M20 4BX, UK
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Manchester, M13 9PL, UK.
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK.
| |
Collapse
|
8
|
Hendriks J, Stojanovic I, Schasfoort RBM, Saris DBF, Karperien M. Nanoparticle Enhancement Cascade for Sensitive Multiplex Measurements of Biomarkers in Complex Fluids with Surface Plasmon Resonance Imaging. Anal Chem 2018; 90:6563-6571. [PMID: 29732889 PMCID: PMC5990928 DOI: 10.1021/acs.analchem.8b00260] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
![]()
There is a large
unmet need for reliable biomarker measurement
systems for clinical application. Such systems should meet challenging
requirements for large scale use, including a large dynamic detection
range, multiplexing capacity, and both high specificity and sensitivity.
More importantly, these requirements need to apply to complex biological
samples, which require extensive quality control. In this paper, we
present the development of an enhancement detection cascade for surface
plasmon resonance imaging (SPRi). The cascade applies an antibody
sandwich assay, followed by neutravidin and a gold nanoparticle enhancement
for quantitative biomarker measurements in small volumes of complex
fluids. We present a feasibility study both in simple buffers and
in spiked equine synovial fluid with four cytokines, IL-1β,
IL-6, IFN-γ, and TNF-α. Our enhancement cascade leads
to an antibody dependent improvement in sensitivity up to 40 000
times, resulting in a limit of detection as low as 50 fg/mL and a
dynamic detection range of more than 7 logs. Additionally, measurements
at these low concentrations are highly reliable with intra- and interassay
CVs between 2% and 20%. We subsequently showed this assay is suitable
for multiplex measurements with good specificity and limited cross-reactivity.
Moreover, we demonstrated robust detection of IL-6 and IL-1β
in spiked undiluted equine synovial fluid with small variation compared
to buffer controls. In addition, the availability of real time measurements
provides extensive quality control opportunities, essential for clinical
applications. Therefore, we consider this method is suitable for broad
application in SPRi for multiplex biomarker detection in both research
and clinical settings.
Collapse
Affiliation(s)
- Jan Hendriks
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine , University of Twente , Enschede , 7522 NB , The Netherlands
| | - Ivan Stojanovic
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine , University of Twente , Enschede , 7522 NB , The Netherlands
| | - Richard B M Schasfoort
- Medical Cell Biophysics, MIRA Institute for Biomedical Technology and Technical Medicine , University of Twente , Enschede , 7522 NB , The Netherlands
| | - Daniël B F Saris
- Department of Orthopedics , UMC Utrecht , Utrecht , 3584 CX , The Netherlands.,Department of Reconstructive Medicine, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology , University of Twente , Enschede , 7522 NB , The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine , University of Twente , Enschede , 7522 NB , The Netherlands
| |
Collapse
|
9
|
Salem A, Mistry H, Backen A, Hodgson C, Koh P, Dean E, Priest L, Haslett K, Trigonis I, Jackson A, Asselin MC, Dive C, Renehan A, Faivre-Finn C, Blackhall F. Cell Death, Inflammation, Tumor Burden, and Proliferation Blood Biomarkers Predict Lung Cancer Radiotherapy Response and Correlate With Tumor Volume and Proliferation Imaging. Clin Lung Cancer 2018; 19:239-248.e7. [PMID: 29398577 PMCID: PMC5927801 DOI: 10.1016/j.cllc.2017.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/21/2017] [Accepted: 12/01/2017] [Indexed: 12/25/2022]
Abstract
INTRODUCTION There is an unmet need to develop noninvasive biomarkers to stratify patients in drug-radiotherapy trials. In this pilot study we investigated lung cancer radiotherapy response and toxicity blood biomarkers and correlated findings with tumor volume and proliferation imaging. PATIENTS AND METHODS Blood samples were collected before and during (day 21) radiotherapy. Twenty-six cell-death, hypoxia, angiogenesis, inflammation, proliferation, invasion, and tumor-burden biomarkers were evaluated. Clinical and laboratory data were collected. Univariate analysis was performed on small-cell and non-small-cell lung cancer (NSCLC) whereas multivariate analysis focused on NSCLC. RESULTS Blood samples from 78 patients were analyzed. Sixty-one (78.2%) harbored NSCLC, 48 (61.5%) received sequential chemoradiotherapy. Of tested baseline biomarkers, undetectable interleukin (IL)-1b (hazard ratio [HR], 4.02; 95% confidence interval [CI], 2.04-7.93; P < .001) was the only significant survival covariate. Of routinely collected laboratory tests, high baseline neutrophil count was a significant survival covariate (HR, 1.07; 95% CI, 1.02-1.11; P = .017). Baseline IL-1b and neutrophil count were prognostic for survival in a multivariate model. The addition of day-21 cytokeratin-19 antigen modestly improved this model's survival prediction (concordance probability, 0.75-0.78). Chemotherapy (P < .001) and baseline keratinocyte growth factor (P = .019) predicted acute esophagitis, but only chemotherapy remained significant after Bonferroni correction. Baseline angioprotein-1 and hepatocyte growth factor showed a direct correlation with tumor volume whereas changes in vascular cell adhesion molecule 1 showed significant correlations with 18F-fluorothymidine (FLT) positron emission tomography (PET). CONCLUSION Select biomarkers are prognostic after radiotherapy in this lung cancer series. The correlation between circulating biomarkers and 18F-FLT PET is shown, to our knowledge for the first time, highlighting their potential role as imaging surrogates.
Collapse
Affiliation(s)
- Ahmed Salem
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom.
| | - Hitesh Mistry
- Division of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Alison Backen
- Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Clare Hodgson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Pek Koh
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Emma Dean
- Early Phase Oncology, AstraZeneca, Cambridge, United Kingdom
| | - Lynsey Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Kate Haslett
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Ioannis Trigonis
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Alan Jackson
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Marie-Claude Asselin
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Andrew Renehan
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Fiona Blackhall
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Fit-for-purpose biomarker immunoassay qualification and validation: three case studies. Bioanalysis 2016; 8:2329-2340. [PMID: 27712082 DOI: 10.4155/bio-2016-0184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIM To improve on the efficiency of biomarker assay readiness, and for reliable biomarker data to support three drug programs, we implemented a fit-for-purpose approach, qualifying two biomarker assays and validating a third. Results/methodology: The qualification strategy and selection of experiments for two exploratory biomarkers (CXCL1, CCL19) was determined by the intended use of the biomarker data. The third biomarker, IL-6, was validated as the data would be used in monitoring patient safety during dose-escalation studies in a Phase I trial. All three assays passed a priori acceptance criteria. CONCLUSION These assays highlight strategies and methodologies for a fit-for-purpose approach. Minimum qualification, full qualification and validation were chosen and supported programs at different stages of drug development.
Collapse
|
11
|
Evaluation of Solid Supports for Slide- and Well-Based Recombinant Antibody Microarrays. MICROARRAYS 2016; 5:microarrays5020016. [PMID: 27600082 PMCID: PMC5003492 DOI: 10.3390/microarrays5020016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 11/17/2022]
Abstract
Antibody microarrays have emerged as an important tool within proteomics, enabling multiplexed protein expression profiling in both health and disease. The design and performance of antibody microarrays and how they are processed are dependent on several factors, of which the interplay between the antibodies and the solid surfaces plays a central role. In this study, we have taken on the first comprehensive view and evaluated the overall impact of solid surfaces on the recombinant antibody microarray design. The results clearly demonstrated the importance of the surface-antibody interaction and showed the effect of the solid supports on the printing process, the array format of planar arrays (slide- and well-based), the assay performance (spot features, reproducibility, specificity and sensitivity) and assay processing (degree of automation). In the end, two high-end recombinant antibody microarray technology platforms were designed, based on slide-based (black polymer) and well-based (clear polymer) arrays, paving the way for future large-scale protein expression profiling efforts.
Collapse
|
12
|
Jani D, Allinson J, Berisha F, Cowan KJ, Devanarayan V, Gleason C, Jeromin A, Keller S, Khan MU, Nowatzke B, Rhyne P, Stephen L. Recommendations for Use and Fit-for-Purpose Validation of Biomarker Multiplex Ligand Binding Assays in Drug Development. AAPS JOURNAL 2015; 18:1-14. [PMID: 26377333 DOI: 10.1208/s12248-015-9820-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/12/2015] [Indexed: 11/30/2022]
Abstract
Multiplex ligand binding assays (LBAs) are increasingly being used to support many stages of drug development. The complexity of multiplex assays creates many unique challenges in comparison to single-plexed assays leading to various adjustments for validation and potentially during sample analysis to accommodate all of the analytes being measured. This often requires a compromise in decision making with respect to choosing final assay conditions and acceptance criteria of some key assay parameters, depending on the intended use of the assay. The critical parameters that are impacted due to the added challenges associated with multiplexing include the minimum required dilution (MRD), quality control samples that span the range of all analytes being measured, quantitative ranges which can be compromised for certain targets, achieving parallelism for all analytes of interest, cross-talk across assays, freeze-thaw stability across analytes, among many others. Thus, these challenges also increase the complexity of validating the performance of the assay for its intended use. This paper describes the challenges encountered with multiplex LBAs, discusses the underlying causes, and provides solutions to help overcome these challenges. Finally, we provide recommendations on how to perform a fit-for-purpose-based validation, emphasizing issues that are unique to multiplex kit assays.
Collapse
Affiliation(s)
- Darshana Jani
- Pfizer Inc., One Burtt Road, Andover, Massachusetts, 01810, USA.
| | - John Allinson
- LGC Ltd, Newmarket Road, Fordham, Cambridgeshire, CB7 5WW, UK
| | - Flora Berisha
- Kyowa-Kirin Pharmaceuticals, 212 Carnegie Center #101, Princeton, New Jersey, 08540, USA
| | - Kyra J Cowan
- Genentech, 1 DNA Way, South San Francisco, California, 94080, USA
| | | | - Carol Gleason
- Bristol-Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey, 08540, USA
| | - Andreas Jeromin
- Quanterix Corporation, 113 Hartwell Avenue, Lexington, Massachusetts, 02421, USA
| | - Steve Keller
- Abbvie Inc., 1500 Seaport Blvd, Redwood City, California, 94063, USA
| | - Masood U Khan
- KCAS Bioanalytical and Biomarker Services, 12400 Shawnee Mission Parkway, Shawnee, Kansas, 66216, USA
| | - Bill Nowatzke
- Radix Biosolutions, 111 Cooperative Way #120, Georgetown, Texas, 78626, USA
| | - Paul Rhyne
- Quintiles Corporation, 1600 Terrell Mill Road Suite 100, Marietta, Georgia, 30067, USA
| | - Laurie Stephen
- Ampersand Biosciences, LLC, 3 Main St., Saranac Lake, New York, 12983, USA
| |
Collapse
|
13
|
Valle JW, Wasan H, Lopes A, Backen AC, Palmer DH, Morris K, Duggan M, Cunningham D, Anthoney DA, Corrie P, Madhusudan S, Maraveyas A, Ross PJ, Waters JS, Steward WP, Rees C, Beare S, Dive C, Bridgewater JA. Cediranib or placebo in combination with cisplatin and gemcitabine chemotherapy for patients with advanced biliary tract cancer (ABC-03): a randomised phase 2 trial. Lancet Oncol 2015; 16:967-78. [PMID: 26179201 PMCID: PMC4648082 DOI: 10.1016/s1470-2045(15)00139-4] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cisplatin and gemcitabine is the standard first-line chemotherapy regimen for patients with advanced biliary tract cancer; expression of VEGF and its receptors is associated with adverse outcomes. We aimed to assess the effect of the addition of cediranib (an oral inhibitor of VEGF receptor 1, 2, and 3) to cisplatin and gemcitabine on progression-free survival. METHODS In this multicentre, placebo-controlled, randomised phase 2 study, we recruited patients aged 18 years or older with histologically confirmed or cytologically confirmed advanced biliary tract cancer from hepatobiliary oncology referral centres in the UK. Patients were eligible if they had an ECOG performance status of 0-1 and an estimated life expectancy of longer than 3 months. Patients were given first-line cisplatin and gemcitabine chemotherapy (25 mg/m(2) cisplatin and 1000 mg/m(2) gemcitabine [on days 1 and 8 every 21 days, for up to eight cycles]) with either 20 mg oral cediranib or placebo once a day until disease progression. We randomly assigned patients (1:1) with a minimisation algorithm, incorporating the stratification factors: extent of disease, primary disease site, previous treatment, ECOG performance status, and centre. The primary endpoint was progression-free survival in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT00939848, and was closed on Sept 30, 2014; results of the final analysis for the primary endpoint are presented. FINDINGS Between April 5, 2011, and Sept 28, 2012, we enrolled 124 patients (62 in each group). With a median follow-up of 12·2 months (IQR 7·3-18·5), median progression-free survival was 8·0 months (95% CI 6·5-9·3) in the cediranib group and 7·4 months (5·7-8·5) in the placebo group (HR 0·93, 80% CI 0·74-1·19, 95% CI 0·65-1·35; p=0·72). Patients who received cediranib had more grade 3-4 toxic effects than did patients who received placebo: hypertension (23 [37%] vs 13 [21%]; p=0·05), diarrhoea (eight [13%] vs two [3%]; p=0·05); platelet count decreased (ten [16%] vs four [6%]; p=0·09), white blood cell decreased (15 [24%] vs seven [11%]; p=0·06) and fatigue (16 [24%] vs seven [11%]; p=0·04). INTERPRETATION Cediranib did not improve the progression-free survival of patients with advanced biliary tract cancer in combination with cisplatin and gemcitabine, which remains the standard of care. Although patients in the cediranib group had more adverse events, we recorded no unexpected toxic effects. The role of VEGF inhibition in addition to chemotherapy for patients with advanced biliary tract cancer remains investigational. FUNDING Cancer Research UK and AstraZeneca Pharmaceuticals.
Collapse
Affiliation(s)
- Juan W Valle
- Institute of Cancer Studies, University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Manchester, UK.
| | | | - Andre Lopes
- Cancer Research UK and UCL Cancer Clinical Trials Centre, London, UK
| | - Alison C Backen
- Institute of Cancer Studies, University of Manchester, Manchester, UK
| | - Daniel H Palmer
- University of Liverpool and Clatterbridge Cancer Centre, Liverpool, UK
| | - Karen Morris
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Marian Duggan
- Cancer Research UK and UCL Cancer Clinical Trials Centre, London, UK
| | | | - D Alan Anthoney
- St James University Hospital, The Leeds Teaching Hospital Trust, Beckett Street, Leeds, UK
| | - Pippa Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | - Paul J Ross
- Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Will P Steward
- Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Charlotte Rees
- Southampton University Hospitals NHS Foundation Trust, Southampton, UK
| | - Sandy Beare
- Cancer Research UK and UCL Cancer Clinical Trials Centre, London, UK
| | - Caroline Dive
- Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | | |
Collapse
|
14
|
Walker MJ, Zhou C, Backen A, Pernemalm M, Williamson AJ, Priest LJ, Koh P, Faivre-Finn C, Blackhall FH, Dive C, Whetton AD. Discovery and Validation of Predictive Biomarkers of Survival for Non-small Cell Lung Cancer Patients Undergoing Radical Radiotherapy: Two Proteins With Predictive Value. EBioMedicine 2015; 2:841-50. [PMID: 26425690 PMCID: PMC4563120 DOI: 10.1016/j.ebiom.2015.06.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/09/2015] [Accepted: 06/17/2015] [Indexed: 02/01/2023] Open
Abstract
Lung cancer is the most frequent cause of cancer-related death world-wide. Radiotherapy alone or in conjunction with chemotherapy is the standard treatment for locally advanced non-small cell lung cancer (NSCLC). Currently there is no predictive marker with clinical utility to guide treatment decisions in NSCLC patients undergoing radiotherapy. Identification of such markers would allow treatment options to be considered for more effective therapy. To enable the identification of appropriate protein biomarkers, plasma samples were collected from patients with non-small cell lung cancer before and during radiotherapy for longitudinal comparison following a protocol that carries sufficient power for effective discovery proteomics. Plasma samples from patients pre- and during radiotherapy who had survived > 18 mo were compared to the same time points from patients who survived < 14 mo using an 8 channel isobaric tagging tandem mass spectrometry discovery proteomics platform. Over 650 proteins were detected and relatively quantified. Proteins which showed a change during radiotherapy were selected for validation using an orthogonal antibody-based approach. Two of these proteins were verified in a separate patient cohort: values of CRP and LRG1 combined gave a highly significant indication of extended survival post one week of radiotherapy treatment.
Collapse
Key Words
- AC, adenocarcinoma
- Biomarker
- CEA, carcinoembryonic antigen
- CRP, C-reactive protein
- EGFR, epidermal growth factor receptor
- FDR, false discovery rate
- IL-6, Interleukin 6
- LBP, lipopolysaccharide binding protein
- LRG1, leucine-rich alpha-2-glycoprotein
- Lung cancer
- MS/MS, tandem mass spectrometry
- NSCLC, non-small cell lung cancer
- PCA, principal component analysis
- Proteomics
- Radiotherapy
- SCLC, small cell lung cancer
- SqCC, squamous cell carcinoma
- TEAB, triethyl ammonium bicarbonate
- VEGF, vascular endothelial growth factor
- iTRAQ, isobaric tagging for relative and absolute quantification
- mo, months
- v/v, volume/volume
Collapse
Affiliation(s)
- Michael J. Walker
- Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
| | - Cong Zhou
- Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester Academic Health Science Centre, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
| | - Alison Backen
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester Academic Health Science Centre, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
| | - Maria Pernemalm
- Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
- Karolinska Institutet, Scilifelab, Department of Oncology and Pathology, Tomtebodavägen 23, 171 65 Stockholm, Sweden
| | - Andrew J.K. Williamson
- Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
| | - Lynsey J.C. Priest
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester Academic Health Science Centre, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
- Faculty Institute of Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, M20 4BX, UK
| | - Pek Koh
- Faculty Institute of Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, M20 4BX, UK
| | - Corinne Faivre-Finn
- Faculty Institute of Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, M20 4BX, UK
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Fiona H. Blackhall
- Faculty Institute of Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, M20 4BX, UK
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester Academic Health Science Centre, Christie Hospital, University of Manchester, Manchester M20 4BX, UK
| | - Anthony D. Whetton
- Stoller Biomarker Discovery Centre, Manchester Academic Health Science Centre, The University of Manchester, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
| |
Collapse
|
15
|
Tighe PJ, Ryder RR, Todd I, Fairclough LC. ELISA in the multiplex era: potentials and pitfalls. Proteomics Clin Appl 2015; 9:406-22. [PMID: 25644123 PMCID: PMC6680274 DOI: 10.1002/prca.201400130] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/08/2014] [Accepted: 01/19/2015] [Indexed: 12/20/2022]
Abstract
Multiplex immunoassays confer several advantages over widely adopted singleplex immunoassays including increased efficiency at a reduced expense, greater output per sample volume ratios and higher throughput predicating more resolute, detailed diagnostics and facilitating personalised medicine. Nonetheless, to date, relatively few protein multiplex immunoassays have been validated for in vitro diagnostics in clinical/point-of-care settings. This review article will outline the challenges, which must be ameliorated prior to the widespread integration of multiplex immunoassays in clinical settings: (i) biomarker validation; (ii) standardisation of immunoassay design and quality control (calibration and quantification); (iii) availability, stability, specificity and cross-reactivity of reagents; (iv) assay automation and the use of validated algorithms for transformation of raw data into diagnostic results. A compendium of multiplex immunoassays applicable to in vitro diagnostics and a summary of the diagnostic products currently available commercially are included, along with an analysis of the relative states of development for each format (namely planar slide based, suspension and planar/microtitre plate based) with respect to the aforementioned issues.
Collapse
Affiliation(s)
- Patrick J Tighe
- School of Life Sciences, The University of Nottingham, Nottingham, UK
| | | | | | | |
Collapse
|
16
|
Backen A, Renehan AG, Clamp AR, Berzuini C, Zhou C, Oza A, Bannoo S, Scherer SJ, Banks RE, Dive C, Jayson GC. The combination of circulating Ang1 and Tie2 levels predicts progression-free survival advantage in bevacizumab-treated patients with ovarian cancer. Clin Cancer Res 2014; 20:4549-4558. [PMID: 24947924 PMCID: PMC4154862 DOI: 10.1158/1078-0432.ccr-13-3248] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Randomized ovarian cancer trials, including ICON7, have reported improved progression-free survival (PFS) when bevacizumab was added to conventional cytotoxic therapy. The improvement was modest prompting the search for predictive biomarkers for bevacizumab. EXPERIMENTAL DESIGN Pretreatment training (n=91) and validation (n=114) blood samples were provided by ICON7 patients. Plasma concentrations of 15 angio-associated factors were determined using validated multiplex ELISAs. Our statistical approach adopted PFS as the primary outcome measure and involved (i) searching for biomarkers with prognostic relevance or which related to between-individual variation in bevacizumab effect; (ii) unbiased determination of cutoffs for putative biomarker values; (iii) investigation of biologically meaningfully predictive combinations of putative biomarkers; and (iv) replicating the analysis on candidate biomarkers in the validation dataset. RESULTS The combined values of circulating Ang1 (angiopoietin 1) and Tie2 (Tunica internal endothelial cell kinase 2) concentrations predicted improved PFS in bevacizumab-treated patients in the training set. Using median concentrations as cutoffs, high Ang1/low Tie2 values were associated with significantly improved PFS for bevacizumab-treated patients in both datasets (median, 23.0 months vs. 16.2; P=0.003) for the interaction of Ang1-Tie2 treatment in Cox regression analysis. The prognostic indices derived from the training set also distinguished high and low probability for progression in the validation set (P=0.008), generating similar values for HR (0.21 vs. 0.27) between treatment and control arms for patients with high Ang1 and low Tie2 values. CONCLUSIONS The combined values of Ang1 and Tie2 are predictive biomarkers for improved PFS in bevacizumab-treated patients with ovarian cancer. These findings need to be validated in larger trials due to the limitation of sample size in this study.
Collapse
Affiliation(s)
- Alison Backen
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Andrew G Renehan
- Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Center, The Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Andrew R Clamp
- Dept Medical Oncology, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - Carlo Berzuini
- Center for Biostatistics, Institute of Population Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Cong Zhou
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Amit Oza
- Bras Family Drug Development Program, Princess Margaret Hospital, 610 University Avenue, Toronto M5G 2M9
| | - Selina Bannoo
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London WC2B 6NH
| | - Stefan J Scherer
- Biocartis SA, EPFL-Quartier de l'Innovation, Bâtimont G, CH-1015 Lausanne
| | - Rosamonde E Banks
- Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds LS9 7TF, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Gordon C Jayson
- Translational Angiogenesis Group, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| |
Collapse
|
17
|
Backen A, Renehan AG, Clamp AR, Berzuini C, Zhou C, Oza A, Bannoo S, Scherer SJ, Banks RE, Dive C, Jayson GC. The combination of circulating Ang1 and Tie2 levels predicts progression-free survival advantage in bevacizumab-treated patients with ovarian cancer. Clin Cancer Res 2014. [PMID: 24947924 DOI: 10.1158/1078-0432.ccr-13-3248] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Randomized ovarian cancer trials, including ICON7, have reported improved progression-free survival (PFS) when bevacizumab was added to conventional cytotoxic therapy. The improvement was modest prompting the search for predictive biomarkers for bevacizumab. EXPERIMENTAL DESIGN Pretreatment training (n=91) and validation (n=114) blood samples were provided by ICON7 patients. Plasma concentrations of 15 angio-associated factors were determined using validated multiplex ELISAs. Our statistical approach adopted PFS as the primary outcome measure and involved (i) searching for biomarkers with prognostic relevance or which related to between-individual variation in bevacizumab effect; (ii) unbiased determination of cutoffs for putative biomarker values; (iii) investigation of biologically meaningfully predictive combinations of putative biomarkers; and (iv) replicating the analysis on candidate biomarkers in the validation dataset. RESULTS The combined values of circulating Ang1 (angiopoietin 1) and Tie2 (Tunica internal endothelial cell kinase 2) concentrations predicted improved PFS in bevacizumab-treated patients in the training set. Using median concentrations as cutoffs, high Ang1/low Tie2 values were associated with significantly improved PFS for bevacizumab-treated patients in both datasets (median, 23.0 months vs. 16.2; P=0.003) for the interaction of Ang1-Tie2 treatment in Cox regression analysis. The prognostic indices derived from the training set also distinguished high and low probability for progression in the validation set (P=0.008), generating similar values for HR (0.21 vs. 0.27) between treatment and control arms for patients with high Ang1 and low Tie2 values. CONCLUSIONS The combined values of Ang1 and Tie2 are predictive biomarkers for improved PFS in bevacizumab-treated patients with ovarian cancer. These findings need to be validated in larger trials due to the limitation of sample size in this study.
Collapse
Affiliation(s)
- Alison Backen
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Andrew G Renehan
- Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Center, The Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Andrew R Clamp
- Dept Medical Oncology, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - Carlo Berzuini
- Center for Biostatistics, Institute of Population Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Cong Zhou
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Amit Oza
- Bras Family Drug Development Program, Princess Margaret Hospital, 610 University Avenue, Toronto M5G 2M9
| | - Selina Bannoo
- MRC Clinical Trials Unit, Aviation House, 125 Kingsway, London WC2B 6NH
| | - Stefan J Scherer
- Biocartis SA, EPFL-Quartier de l'Innovation, Bâtimont G, CH-1015 Lausanne
| | - Rosamonde E Banks
- Leeds Institute of Cancer and Pathology, St James University Hospital, Leeds LS9 7TF, UK
| | - Caroline Dive
- Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, United Kingdom
| | - Gordon C Jayson
- Translational Angiogenesis Group, University of Manchester and Christie Hospital, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| |
Collapse
|
18
|
Investigation of novel circulating proteins, germ line single-nucleotide polymorphisms, and molecular tumor markers as potential efficacy biomarkers of first-line sunitinib therapy for advanced renal cell carcinoma. Cancer Chemother Pharmacol 2014; 74:739-50. [PMID: 25100134 PMCID: PMC4175044 DOI: 10.1007/s00280-014-2539-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 07/11/2014] [Indexed: 11/22/2022]
Abstract
Purpose
Sunitinib is a first-line advanced renal cell carcinoma (RCC) standard of care. In a randomized phase II trial comparing sunitinib treatment schedules, separate exploratory biomarker analyses investigated the correlations of efficacy with selected serum, germ line single-nucleotide polymorphism (SNP), or tumor markers. Methods Advanced RCC patients received first-line sunitinib 50 mg/day on the approved 4-week-on-2-week-off schedule (n = 146) or 37.5 mg/day continuous dosing (n = 146). The following correlation analyses were performed: (1) response evaluation criteria in solid tumors-defined tumor response with serum soluble protein levels via two distinct multiplex (n < 1,000) platforms; (2) response and time-to-event outcomes with germ line SNPs in vascular endothelial growth factor (VEGF)-A and VEGF receptor (VEGFR)3 genes; and (3) response and time-to-event outcomes with tumor immunohistochemistry status for hypoxia-inducible factor 1-alpha (HIF-1α) and carbonic anhydrase-IX or tumor Von Hippel–Lindau (VHL) gene inactivation status. Results Lower baseline angiopoietin-2 (Ang-2) and higher baseline matrix metalloproteinase-2 (MMP-2) levels were identified by both platforms as statistically significantly associated with tumor response. There were no significant correlations between VEGF-A or VEGFR3 SNPs and outcomes. Progression-free survival was longer for HIF-1α percent of tumor expression groups 0–2 (HIF-1α low) versus 3–4 (HIF-1α high; p = 0.034). There were no significant correlations between outcomes and each VHL inactivation mechanism [mutation (86 % of VHL-inactive patients), methylation (14 %), and large deletion (7 %)] or mechanisms combined. Conclusions Serum Ang-2 and MMP-2 and tumor HIF-1α were identified as relevant baseline biomarkers of sunitinib activity in advanced RCC, warranting further research into their prognostic versus predictive value. Electronic supplementary material The online version of this article (doi:10.1007/s00280-014-2539-0) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
A multiplex immunoassay gives different results than singleplex immunoassays which may bias epidemiologic associations. Clin Biochem 2012; 45:848-51. [DOI: 10.1016/j.clinbiochem.2012.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 03/28/2012] [Accepted: 04/08/2012] [Indexed: 11/19/2022]
|
20
|
Serum biomarker modulation following molecular targeting of epidermal growth factor and cyclooxygenase pathways: a pilot randomized trial in head and neck cancer. Oral Oncol 2012; 48:1136-45. [PMID: 22732263 DOI: 10.1016/j.oraloncology.2012.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/16/2012] [Accepted: 05/21/2012] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Targeting the epidermal growth factor receptor (EGFR) using the tyrosine kinase inhibitor (TKI) erlotinib has demonstrated activity in aerodigestive tract malignancies. Co-targeting of the G-protein-coupled receptor cyclooxygenase (COX) with EGFR inhibitors has shown promise in preclinical models and early phase clinical studies. MATERIALS AND METHODS We studied the modulation of serum proteins after neoadjuvant treatment with erlotinib with or without sulindac in head and neck cancer patients. In a prospective, randomized, double-blind clinical trial, paired serum samples were obtained before and after neoadjuvant treatment in three groups of patients (n = 23 total), who were randomized to receive 7-14 consecutive days of erlotinib alone, erlotinib plus sulindac, or placebo. Two separate multiplexed ELISA systems (SearchLight™ or Luminex™) were used to measure serum biomarkers. HGF and IL-6 levels were tested on both systems, and validated using single analyte ELISAs. RESULTS Several analytes were significantly altered (generally decreased) post-treatment, in patients who received erlotinib (with or without sulindac) as well as in the placebo groups. No single analyte was differentially altered across the three treatment groups using either multiplex platform. Single HGF ELISA suggested a nonspecific decrease in all patients. CONCLUSION These results demonstrate the importance of a placebo group when assessing changes in expression of serum biomarkers. While multiplex platforms can provide quantitative information on a large number of serum analytes, results should be cautiously compared across platforms due to their intrinsic features. Furthermore, the dynamic range of expression of a single analyte is constrained in multiplex versus standard ELISA.
Collapse
|
21
|
Trune DR, Larrain BE, Hausman FA, Kempton JB, MacArthur CJ. Simultaneous measurement of multiple ear proteins with multiplex ELISA assays. Hear Res 2011; 275:1-7. [PMID: 21144888 PMCID: PMC3087854 DOI: 10.1016/j.heares.2010.11.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/09/2010] [Accepted: 11/29/2010] [Indexed: 10/18/2022]
Abstract
A recent advancement in enzyme-linked immunosorbent assay (ELISA) technology is the multiplex antibody array that measures multiple proteins simultaneously within a single sample. This allows reduction in sample volume, time, labor, and material costs, while increasing sensitivity over single ELISA. Current multiplex platforms include planar-based systems using microplates or slides, or bead-based suspension assay with microspheres. To determine the applicability of this technology for ear research, we used 3 different multiplex ELISA-based immunoassay arrays from 4 different companies to measure cytokine levels in mouse middle and inner ear tissue lysate extracts 24 h following transtympanic Haemophilus influenzae inoculation. Middle and inner ear tissue lysates were analyzed using testing services from Quansys Biosciences, Aushon Biosystems SearchLight (both microplate-based), MILLIPLEX MAP Sample (bead-based), and a RayBiotech, Inc (slide-based) kit. Samples were assayed in duplicate or triplicate. Results were compared to determine their relative sensitivity and reliability for measures of cytokines related to inflammation. The cytokine pg/ml amounts varied among the multiplex assays, so a comparison also was made of the mean fold increase in cytokines from untreated controls. Several cytokines and chemokines were elevated, the extent dependent upon the assay sensitivity. Those most significantly elevated were IL-1α, IL-1β, IL-6, TNFα, VEGF, and IL-8/MIP-2. The results of the multiplex systems were compared with single ELISA kits (IL-1β, IL-6) to assess sensitivity over the traditional method. Overall, the Quansys Biosciences and SearchLight arrays showed the greatest sensitivity, both employing the same multiplex methodology of a spotted array within a microplate well with chemiluminescent detection. They also were more sensitive than the traditional single ELISA performed with commercial kits and matched gene expression changes determined by quantitative RT-PCR. The Quansys array showed a limit of detection for ear IL-6 down to 2-4 pg/ml, indicating it is sufficiently sensitive to detect ear proteins present in low concentrations. Thus, the multiplex ELISA procedures appear suitable and reliable for the study of hearing related proteins, providing accurate, quantitative, reproducible results with considerable improvement in sensitivity and economy.
Collapse
Affiliation(s)
- Dennis R Trune
- Oregon Hearing Research Center, Department of Otolaryngology-Head & Neck Surgery, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code NRC04, Portland, OR 97239-3098, USA.
| | | | | | | | | |
Collapse
|
22
|
Lancashire LJ, Roberts DL, Dive C, Renehan AG. The development of composite circulating biomarker models for use in anticancer drug clinical development. Int J Cancer 2011; 128:1843-51. [PMID: 20549702 DOI: 10.1002/ijc.25513] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The development of informative composite circulating biomarkers predicting cancer presence or therapy response is clinically attractive but optimal approaches to modeling are as yet unclear. This study investigated multidimensional relationships within an example panel of serum insulin-like growth factor (IGF) peptides using logistic regression (LR), fractional polynomial (FP), regression, artificial neural networks (ANNs) and support vector machines (SVMs) to derive predictive models for colorectal cancer (CRC). Two phase 2 biomarker validation analyses were performed: controls were ambulant adults (n = 722); cases were: (i) CRC patients (n = 100) and (ii) patients with acromegaly (n = 52), the latter as "positive" discriminators. Serum IGF-I, IGF-II, IGF binding protein (IGFBP)-2 and -3 were measured. Discriminatory characteristics were compared within and between models. For the LR, FP and ANN models, and to a lesser extent SVMs, the addition of covariates at several steps improved discrimination characteristics. The optimum biomarker combination discriminating CRC vs. controls was achieved using ANN models [sensitivity, 94%; specificity, 90%; accuracy, 0.975 (95% CIs: 0.948 1.000)]. ANN modeling significantly outperformed LR, FP and SVM in terms of discrimination (p < 0.0001) and calibration. The acromegaly analysis demonstrated expected high performance characteristics in the ANN model [accuracy, 0.993 (95% CIs: 0.977, 1.000)]. Curved decision surfaces generated from the ANNs revealed the potential clinical utility. This example demonstrated improved discriminatory characteristics within the composite biomarker ANN model and a final model that outperformed the three other models. This modeling approach forms the basis to evaluate composite biomarkers as pharmacological and predictive biomarkers in future clinical trials.
Collapse
Affiliation(s)
- Lee J Lancashire
- Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, Manchester, UK
| | | | | | | |
Collapse
|
23
|
Bastarache JA, Koyama T, Wickersham NE, Mitchell DB, Mernaugh RL, Ware LB. Accuracy and reproducibility of a multiplex immunoassay platform: a validation study. J Immunol Methods 2011; 367:33-9. [PMID: 21277854 PMCID: PMC3108329 DOI: 10.1016/j.jim.2011.01.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 01/17/2011] [Accepted: 01/17/2011] [Indexed: 01/17/2023]
Abstract
BACKGROUND Multiplex immunoassays offer many advantages over singleplex assays for the analysis of multiple analytes in a single sample. We sought to validate a specific multiplex cytokine immunoassay (Human 9-plex cytokine array on the Searchlight® platform by Thermoscientific) prior to use in a large clinical study. METHODS We compared spike and recovery of recombinant proteins on the Searchlight® platform to singleplex immunoassays purchased from R&D Systems, measured identical patient samples on the two different platforms, and measured identical patient samples on different days to measure intra- and inter-assay variability. RESULTS Assays using the Searchlight® platform had inefficient recovery of spiked recombinant proteins compared to R&D Systems singleplex assays. Assaying identical patients samples on different days on the Searchlight platform had acceptable intra-assay variability (intra-assay coefficient of variation (CV%) range for all analytes of 9.1-13.7) but unacceptably high inter-assay variability (CV% range for all analytes 16.7-119.3) suggesting plate-to plate variability. Similar assays for individual cytokines on the R&D platform had an intra-assay CV% range of 1.6-6.4 and an inter-assay CV% range of 3.8-7.1. Some deficiencies in Searchlight® assay performance may be due to irregularities in spotting of capture antibodies during manufacturing. CONCLUSIONS We conclude that the Searchlight® multiplex immunoassay platform would require extensive additional assay optimization prior to widespread clinical research use.
Collapse
Affiliation(s)
- Julie A Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, United States.
| | | | | | | | | | | |
Collapse
|
24
|
Parazzi V, Lazzari L, Rebulla P. Platelet gel from cord blood: a novel tool for tissue engineering. Platelets 2011; 21:549-54. [PMID: 20873963 DOI: 10.3109/09537104.2010.514626] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent findings show that growth factors (GF) play a relevant role in regenerative medicine. Platelets (PLT) may be used as “drug-stores” of GF that can be released upon activation by PLT granules. In this context, PLT gel (PG) from peripheral blood is currently used to improve tissue healing in orthopedic, oral maxillofacial and dermatologic surgery. Recent findings on multiple biological properties of human umbilical cord blood (CB) and its high level of viral safety prompted us to investigate the characteristics of its PLTs and the possibility to produce PLT gel from cord blood. Our study shows that CB PG releases high levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor-BB (PDGF-BB), substantial amounts of fibroblast growth factor (FGF), hepatocyte growth factor (HGF) and transforming growth factor-beta 1 (TGFbeta1), and minimal amounts of PDGF-AB. These findings suggest that CB PG can be a preferable tool for tissue engineering applications where high levels of VEGF and PDGF may be desirable.
Collapse
Affiliation(s)
- Valentina Parazzi
- Cell Factory, Center of Transfusion Medicine, Cellular Therapy and Cryobiology, Department of Regenerative Medicine, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | |
Collapse
|
25
|
Sun M, Manolopoulou J, Spyroglou A, Beuschlein F, Hantel C, Wu Z, Bielohuby M, Hoeflich A, Liu C, Bidlingmaier M. A microsphere-based duplex competitive immunoassay for the simultaneous measurements of aldosterone and testosterone in small sample volumes: validation in human and mouse plasma. Steroids 2010; 75:1089-96. [PMID: 20654638 DOI: 10.1016/j.steroids.2010.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 07/07/2010] [Accepted: 07/09/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND The small blood volumes available in rodent studies often limit adequate quantification of all hormones of interest. We report here the development of two new assays combining an extraction step with multiplex immunoassay (MIA) technology for the simultaneous determination of aldosterone and testosterone in 50 μl sample volume. METHODS Following solvent extraction, aldosterone and testosterone competitive immunoassays are performed incorporating biotinylated tracers and antibody-coated beads each having a unique fluorescence. Quantification is via addition of streptavidin-R-phycoerythrin (SA-PE). The assays were validated and compared to established methods. Baseline hormone levels in mice from four different strains, and changes after ACTH and HCG stimulation in CD-1 mice are shown. RESULTS The assays are sensitive (aldosterone 15 pg/ml, testosterone 12 pg/ml), reproducible (intra-/inter-assay imprecision aldosterone 5.1-15.6%/9.9-15.8% and testosterone 9.7-10.9%/7.7-11.4%) and correlate significantly to established assays (r=0.94-0.95). Baseline aldosterone levels varied between strains, but not between the genders. Testosterone was significantly higher in male of all strains except in C57BL/6 × NMRI mice. After ACTH injection, aldosterone (median, interquartile range) rose from 354 (261-396) pg/ml to 2008 (875-2467) in male and from 260 (210-576) to 1120 (734-1528) in female CD-1 mice. HCG injection in the same strain increased testosterone in male mice only (3.5 (0.4-8.3) ng/ml to 31.8 (30.4-33.9) ng/ml, P<0.01). CONCLUSIONS We describe a MIA for the simultaneous measurement of aldosterone and testosterone in small volumes after extraction. In addition to presenting a new tool for steroid research in rodent models, our data show strain-dependent differences in steroid hormone metabolism in rodents.
Collapse
Affiliation(s)
- Min Sun
- Department of Endocrinology, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Boja E, Rivers R, Kinsinger C, Mesri M, Hiltke T, Rahbar A, Rodriguez H. Restructuring proteomics through verification. Biomark Med 2010; 4:799-803. [PMID: 21133699 PMCID: PMC3041639 DOI: 10.2217/bmm.10.92] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Proteomics technologies have revolutionized cell biology and biochemistry by providing powerful new tools to characterize complex proteomes, multiprotein complexes and post-translational modifications. Although proteomics technologies could address important problems in clinical and translational cancer research, attempts to use proteomics approaches to discover cancer biomarkers in biofluids and tissues have been largely unsuccessful and have given rise to considerable skepticism. The National Cancer Institute has taken a leading role in facilitating the translation of proteomics from research to clinical application, through its Clinical Proteomic Technologies for Cancer. This article highlights the building of a more reliable and efficient protein biomarker development pipeline that incorporates three steps: discovery, verification and qualification. In addition, we discuss the merits of multiple reaction monitoring mass spectrometry, a multiplex targeted proteomics platform, which has emerged as a potentially promising, high-throughput protein biomarker measurements technology for preclinical 'verification'.
Collapse
Affiliation(s)
- Emily Boja
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| | - Robert Rivers
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| | - Christopher Kinsinger
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| | - Tara Hiltke
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| | - Amir Rahbar
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, Center for Strategic Scientific Initiative, National Cancer Institute, NIH, 31 Center Drive, MS 2590, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Cummings J, Ward TH, Dive C. Fit-for-purpose biomarker method validation in anticancer drug development. Drug Discov Today 2010; 15:816-25. [DOI: 10.1016/j.drudis.2010.07.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 06/21/2010] [Accepted: 07/29/2010] [Indexed: 12/31/2022]
|
28
|
Mitchell CL, O'Connor JPB, Jackson A, Parker GJM, Roberts C, Watson Y, Cheung S, Davies K, Buonaccorsi GA, Clamp AR, Hasan J, Byrd L, Backen A, Dive C, Jayson GC. Identification of early predictive imaging biomarkers and their relationship to serological angiogenic markers in patients with ovarian cancer with residual disease following cytotoxic therapy. Ann Oncol 2010; 21:1982-1989. [PMID: 20351070 DOI: 10.1093/annonc/mdq079] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025] Open
Abstract
BACKGROUND Patients with recurrent ovarian cancer often achieve partial response following chemotherapy, resulting in persistent small volume disease. After completion of treatment, the dilemma of when to initiate subsequent chemotherapy arises. Identification of biomarkers that could be used to predict when subsequent treatment is needed would be of significant benefit. DESIGN Twenty-three patients with advanced ovarian cancer and residual asymptomatic disease following chemotherapy underwent dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) at study entry, 4, 8, 12, 18 and 26 weeks or disease progression. A subgroup of patients provided plasma samples within which a panel of angiogenic biomarkers was quantified. RESULTS By 4 weeks, significant differences in whole tumour volume, enhancing fraction and Ca125 were observed between patients whose disease progressed by 26 weeks and those who remained stable. Significant correlations between plasma soluble vascular endothelial growth factor receptor-1 (sVEGFR-1) and sVEGFR-2 concentrations, and blood volume and tumour endothelial permeability surface area product measured by DCE-MRI were observed. CONCLUSIONS Imaging markers have a potential role in early prediction of disease progression in patients with residual ovarian cancer and may supplement current measures of progression. The correlation of DCE-MRI and serological biomarkers suggests that tumour angiogenesis affects these markers through common biological means and warrants further investigation.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/blood
- CA-125 Antigen/blood
- Contrast Media
- Cystadenocarcinoma, Serous/blood
- Cystadenocarcinoma, Serous/diagnosis
- Cystadenocarcinoma, Serous/drug therapy
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Magnetic Resonance Imaging
- Membrane Proteins/blood
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Staging
- Neoplasm, Residual/blood
- Neoplasm, Residual/diagnosis
- Neoplasm, Residual/drug therapy
- Neovascularization, Pathologic
- Ovarian Neoplasms/blood
- Ovarian Neoplasms/diagnosis
- Ovarian Neoplasms/drug therapy
- Peritoneal Neoplasms/blood
- Peritoneal Neoplasms/diagnosis
- Peritoneal Neoplasms/drug therapy
- Prognosis
- Survival Rate
- Vascular Endothelial Growth Factor Receptor-1/blood
- Vascular Endothelial Growth Factor Receptor-2/blood
Collapse
Affiliation(s)
- C L Mitchell
- Cancer Research UK, Department of Medical Oncology, Christie Hospital and University of Manchester.
| | - J P B O'Connor
- Cancer Research UK, Department of Medical Oncology, Christie Hospital and University of Manchester; Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - A Jackson
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - G J M Parker
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - C Roberts
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - Y Watson
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - S Cheung
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - K Davies
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - G A Buonaccorsi
- Imaging Science and Biomedical Engineering Department, School of Cancer and Imaging Sciences, University of Manchester
| | - A R Clamp
- Cancer Research UK, Department of Medical Oncology, Christie Hospital and University of Manchester
| | - J Hasan
- Cancer Research UK, Department of Medical Oncology, Christie Hospital and University of Manchester
| | - L Byrd
- Cancer Research UK, Department of Medical Oncology, Christie Hospital and University of Manchester
| | - A Backen
- Department of Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - C Dive
- Department of Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - G C Jayson
- Cancer Research UK, Department of Medical Oncology, Christie Hospital and University of Manchester; Department of Clinical and Experimental Pharmacology, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| |
Collapse
|
29
|
Brookes K, Cummings J, Backen A, Greystoke A, Ward T, Jayson GC, Dive C. Issues on fit-for-purpose validation of a panel of ELISAs for application as biomarkers in clinical trials of anti-Angiogenic drugs. Br J Cancer 2010; 102:1524-32. [PMID: 20407440 PMCID: PMC2869162 DOI: 10.1038/sj.bjc.6605661] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/16/2010] [Accepted: 03/22/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Successful introduction of new anticancer agents into the clinic is often hampered by a lack of qualified biomarkers. Studies have been conducted of 17 ELISAs representing a potential panel of pharmacodynamic/predictive biomarkers for drugs targeted to tumour vasculature. METHODS The fit-for-purpose approach to method validation was used. Stability studies were performed using recombinant proteins in surrogate matrices, endogenous analytes in healthy volunteer and cancer patient plasma. The impact of platelet depletion was investigated. RESULTS Method validation focused on measuring precision and showed that 15 of the 17 assays were within acceptable limits. Stability at -80 degrees C was shown for 3 months with all recombinant proteins in surrogate matrices, whereas under the same conditions instability was observed with KGF in platelet-rich and platelet-depleted plasma, and with PDGF-BB in platelet-depleted plasma from cancer patients. For measurement of extracellular circulating analytes, platelet depletion should be conducted before freezing of plasma to prevent release of PDGF-BB, FGFb and VEGF-A. A protocol was developed to remove >90% platelets from plasma requiring centrifugation at 2000 g for 25 min. CONCLUSIONS These studies highlight the need for assay validation and crucial assessment of sample handling issues before commencement of biomarker analysis in clinical trials.
Collapse
Affiliation(s)
- K Brookes
- Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
- Translational Angiogenesis Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - J Cummings
- Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - A Backen
- Translational Angiogenesis Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - A Greystoke
- Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - T Ward
- Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - G C Jayson
- Translational Angiogenesis Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - C Dive
- Clinical and Experimental Pharmacology Group, Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
30
|
Randlev B, Huang LC, Watatsu M, Marcus M, Lin A, Shih SJ. Validation of a quantitative flow cytometer assay for monitoring HER-2/neu expression level in cell-based cancer immunotherapy products. Biologicals 2010; 38:249-59. [PMID: 20080049 DOI: 10.1016/j.biologicals.2009.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 10/08/2009] [Accepted: 12/02/2009] [Indexed: 11/26/2022] Open
Abstract
GVAX immunotherapy for prostate cancer is comprised of two genetically modified prostate cancer cell lines, CG1940 and CG8711, engineered to secrete granulocyte macrophage-colony-stimulating factor. As part of the matrix of potency assays, CG1940 and CG8711 are tested for the expression level of cell surface HER-2/neu using a quantitative flow cytometer assay. This assay reports the antibody binding capacity value of the cells as a measure of HER-2/neu expression using cells immediately after thawing from cryogenic storage. With optimized cell handling and staining procedure and appropriate system suitability controls, the assay was validated as a quantitative assay. The validation results showed that assay accuracy, specificity, precision, linearity, and range were suitable for the intended use of ensuring lot-to-lot consistency of HER-2/neu expression. Assay robustness was demonstrated using design of experiments that evaluated critical assay parameters. Finally, the assay was successfully transferred to a current good manufacturing practice Quality Control laboratory in a separate facility. Since the overall precision of this assay is better than that of ELISA methods and it can be performed with ease and high throughput, quantitative flow cytometer-based assays may be an appropriate immunological assay platform for Quality Control laboratories for characterization and release of cell-based therapies.
Collapse
Affiliation(s)
- Britta Randlev
- Assay Development, Cell Genesys, Inc., 500 Forbes Boulevard, South San Francisco, CA 94404, USA
| | | | | | | | | | | |
Collapse
|
31
|
Ligand-binding assays: risk of using a platform supported by a single vendor. Bioanalysis 2009; 1:629-36. [DOI: 10.4155/bio.09.46] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The use of biological reagents in ligand-binding assays (LBAs) presents inherent challenges when measuring the concentration of large molecules in complex matrices. As a result, there are relatively few platforms that provide the accuracy, precision and robustness needed to determine the concentration of macromolecular therapies and biomarkers, and demonstrate the presence or absence of an immune response. Some bioanalytical laboratories use only one LBA platform to reduce costs, increase efficiency and maintain optimal assay performance. However, the business and regulatory risks of using a single platform supported by only one vendor should be considered. This article summarizes the immunological methods used to support bioanalysis for large molecules that are supported by a single vendor, the benefits of being dedicated to a single platform for bioanalysis used for regulatory filings, the costs associated with restructuring if an immunoassay platform is discontinued and recommendations to mitigate risk when using LBAs in drug development. The experience with the recent discontinuation of the BioVeris™ electrochemiluminescent-based platform is discussed.
Collapse
|
32
|
Zimmermann R, Ringwald J, Eckstein R. EDTA plasma is unsuitable for in vivo determinations of platelet-derived angiogenic cytokines. J Immunol Methods 2009; 347:91-2. [PMID: 19394338 DOI: 10.1016/j.jim.2009.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 04/20/2009] [Accepted: 04/21/2009] [Indexed: 11/29/2022]
|