1
|
Saputra HA, Chung JH, Sahin MAZ, Park DS, Shim YB. Dual-signal output biosensor for the detection of program death-ligand 1 and therapy progress monitoring of cancer. Biosens Bioelectron 2024; 262:116565. [PMID: 39003918 DOI: 10.1016/j.bios.2024.116565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
A disposable dual-output biosensor to detect program death-ligand 1 (PD-L1) was developed for immunotherapy progress monitoring and early cancer detection in a single experimental setup. The aptamer probe was assembled on rGO composited with carboxylated terthiophene polymer (rGO-pTBA) to specifically capture PD-L1 protein labeled with a new redox mediator, ortho-amino phenol para sulphonic acid, for amperometric detection. Each sensing layer was characterized through electrochemical and surface analysis experiments, then confirmed the sensing performance. The calibration plots for the standard PD-L1 protein detection revealed two dynamic ranges of 0.5-100.0 pM and 100.0-500.0 pM, where the detection limit was 0.20 ± 0.001 pM (RSD ≤5.2%) by amperometry. The sensor reliability was evaluated by detecting A549 lung cancer cell-secreted PD-L1 and clinically relevant serum levels of soluble PD-L1 (sPD-L1) using both detection methods. In addition, therapeutic trials were studied through the quantification of sPD-L1 levels for a small cohort of lung cancer patients. A significantly higher level of sPD-L1 was observed for patients (221.6-240.4 pM) compared to healthy individuals (16.2-19.6 pM). After immunotherapy, the patients' PD-L1 level decreased to the range of 126.7-141.2 pM. The results indicated that therapy monitoring was successfully done using both the proposed methods. Additionally, based on a comparative study on immune checkpoint-related proteins, PD-L1 is a more effective biomarker than granzyme B and interferon-gamma.
Collapse
Affiliation(s)
- Heru Agung Saputra
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan, 46241, South Korea
| | - Jae Heun Chung
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, 626-770, South Korea
| | - Md Ali Zaber Sahin
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan, 46241, South Korea
| | - Deog-Su Park
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan, 46241, South Korea
| | - Yoon-Bo Shim
- Department of Chemistry and Chemistry Institute for Functional Materials, Pusan National University, Busan, 46241, South Korea.
| |
Collapse
|
2
|
Haveman NJ, Schuerger AC, Yu PL, Brown M, Doebler R, Paul AL, Ferl RJ. Advancing the automation of plant nucleic acid extraction for rapid diagnosis of plant diseases in space. FRONTIERS IN PLANT SCIENCE 2023; 14:1194753. [PMID: 37389293 PMCID: PMC10304293 DOI: 10.3389/fpls.2023.1194753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023]
Abstract
Human space exploration missions will continue the development of sustainable plant cultivation in what are obviously novel habitat settings. Effective pathology mitigation strategies are needed to cope with plant disease outbreaks in any space-based plant growth system. However, few technologies currently exist for space-based diagnosis of plant pathogens. Therefore, we developed a method of extracting plant nucleic acid that will facilitate the rapid diagnosis of plant diseases for future spaceflight applications. The microHomogenizer™ from Claremont BioSolutions, originally designed for bacterial and animal tissue samples, was evaluated for plant-microbial nucleic acid extractions. The microHomogenizer™ is an appealing device in that it provides automation and containment capabilities that would be required in spaceflight applications. Three different plant pathosystems were used to assess the versatility of the extraction process. Tomato, lettuce, and pepper plants were respectively inoculated with a fungal plant pathogen, an oomycete pathogen, and a plant viral pathogen. The microHomogenizer™, along with the developed protocols, proved to be an effective mechanism for producing DNA from all three pathosystems, in that PCR and sequencing of the resulting samples demonstrated clear DNA-based diagnoses. Thus, this investigation advances the efforts to automate nucleic acid extraction for future plant disease diagnosis in space.
Collapse
Affiliation(s)
- Natasha J. Haveman
- NASA Utilization & Life Sciences Office (UB-A), Kennedy Space Center, Merritt Island, FL, United States
| | - Andrew C. Schuerger
- Department of Plant Pathology, University of Florida, Space Life Science Lab, Merritt Island, FL, United States
| | - Pei-Ling Yu
- Department of Plant Pathology, University of Florida, Gainesville, FL, United States
| | - Mark Brown
- Claremont BioSolutions Limited Liability Company (LLC), Upland, CA, United States
| | - Robert Doebler
- Claremont BioSolutions Limited Liability Company (LLC), Upland, CA, United States
| | - Anna-Lisa Paul
- Department of Horticultural Sciences, University of Florida, Gainesville, FL, United States
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, United States
| | - Robert J. Ferl
- Department of Horticultural Sciences, University of Florida, Gainesville, FL, United States
- University of Florida Office of Research, University of Florida, Gainesville, FL, United States
| |
Collapse
|
3
|
Mahajan S, Nambirajan A, Gupta I, Gupta N, Gupta P, Jain D. Malignant pleural effusion cell blocks are reliable resources for PD-L1 analysis in advanced lung adenocarcinomas: a concordance study with matched histologic samples. J Am Soc Cytopathol 2022; 11:253-263. [PMID: 35589508 DOI: 10.1016/j.jasc.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION In lung cancer patients presenting with malignant pleural effusion (MPE), cytology might represent the only source of tumor tissue for diagnosis and predictive biomarker testing. Programmed death ligand 1 (PD-L1) expression in tumor cells is a predictive biomarker for immunotherapy in non-small cell lung carcinomas and is tested using immunohistochemistry. However, knowledge of the validity of PD-L1 testing on MPE samples is limited. We evaluated the feasibility of immunocytochemistry (ICC) for PD-L1 in MPE cell blocks (CBs) and assessed the concordance in expression with patient-matched histologic samples. MATERIALS AND METHODS ICC for PD-L1 was performed on formalin-fixed paraffin-embedded CBs of MPE and patient-matched histologic samples, if available, using the automated Ventana PD-L1 SP263 assay. The tumor proportion score (TPS), based on partial or complete membranous tumor cell staining, was categorized as negative (<1%), low (≥1% to <50%), and high (≥50%). In CBs with any degree of PD-L1 expression, ICC for CD163 highlighting macrophages was performed to exclude nonspecific PD-L1 expression in macrophages. The CB PD-L1 TPS was compared with the TPS obtained from the patient-matched histologic samples. RESULTS Of 43 MPE CBs available, 25 were positive for PD-L1 (25 of 42; 59%), and 1 sample was inadequate. Of the 11 patient-matched histologic samples tested, the PD-L1 TPS categories were concordant for 10 of the 11 (91% concordance) cases. CONCLUSIONS PD-L1 expression in MPE CBs showed good concordance with expression in histologic samples and is feasible as a source for PD-L1 testing. The concurrent use of CD163 immunostains will aid in the manual assessment of PD-L1 TPS.
Collapse
Affiliation(s)
- Swati Mahajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Ishan Gupta
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Nalini Gupta
- Department of Cytology and Gynecologic Pathology, Post Graduate Institute of Medical Sciences and Research, Chandigarh, India
| | - Parikshaa Gupta
- Department of Cytology and Gynecologic Pathology, Post Graduate Institute of Medical Sciences and Research, Chandigarh, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
4
|
Tamura K, Miyato H, Kanamaru R, Sadatomo A, Takahashi K, Ohzawa H, Koyanagi T, Saga Y, Takei Y, Fujiwara H, Lefor AK, Sata N, Kitayama J. Neutrophil extracellular traps (NETs) reduce the diffusion of doxorubicin which may attenuate its ability to induce apoptosis of ovarian cancer cells. Heliyon 2022; 8:e09730. [PMID: 35756123 PMCID: PMC9218137 DOI: 10.1016/j.heliyon.2022.e09730] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/08/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose Although neutrophil extracellular traps (NETs) are present in various tumors, their roles in tumor biology have not been clarified yet. In this study, we examined how NETs affect the pharmacokinetics and effects of doxorubicin (DOX). Methods NETs were generated by neutrophils stimulated with phorbol 12-myristate 13-acetate (PMA) or lipopolysaccharide (LPS). DOX was added to NETs and their distribution was observed under fluorescein microscopy, and the diffusion of DOX through 3 μM pores from lower to upper chambers was evaluated with a fluorescence-based assay. Ovarian cancer cells, KOC-2S and SKOV3, were embedded in collagen gel droplets and cultured in 3D way and their apoptosis was examined with flow cytometry. Results DOX was mostly co-localized with NETs. The transfer of DOX to upper chambers increased over time, which was significantly decreased by the presence of neutrophils stimulated with PMA or LPS in the lower chamber. DOX outside of the gel increased the rates of annexin V (+) apoptotic cells, which were significantly reduced by the addition of LPS-stimulated neutrophils in media both in KOC-2S and SKOV3. The reduced diffusion and apoptosis were mostly restored by the destruction of the NETs structure with 1000 u/ml DNAse I. Conclusion NETs efficiently trap and inhibit the diffusion of DOX which may attenuate its ability to induce apoptosis of ovarian cancer cells. Degradation of NETs with DNAse I may augment the response of ovarian cancer to DOX. Doxorubicin is efficiently trapped by neutrophil extracellular traps (NETs). NETs suppress diffusion of doxorubicin through micro-pores and infiltration into resected tumor. NETs suppress doxorubicin-induced apoptosis of tumor cells in 3-D culture. DNAse may augment the effect of anti-cancer drugs by modulating pharmacokinetics.
Collapse
Affiliation(s)
- Kohei Tamura
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | - Hideyo Miyato
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Rihito Kanamaru
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Ai Sadatomo
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Kazuya Takahashi
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Hideyuki Ohzawa
- Department of Clinical Oncology, Jichi Medical University, Shimotsuke, Japan
| | - Takahiro Koyanagi
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | - Yasushi Saga
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | - Yuji Takei
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | | | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Joji Kitayama
- Department of Surgery, Jichi Medical University, Shimotsuke, Japan
- Center for Clinical Research, Jichi Medical University Hospital, Shimotsuke, Japan
- Corresponding author.
| |
Collapse
|
5
|
Saito A, Tojo M, Kumagai Y, Ohzawa H, Yamaguchi H, Miyato H, Sadatomo A, Naoi D, Ota G, Koinuma K, Horie H, Lefor AK, Sata N, Kitayama J. Flow cytometry detection of cell type-specific expression of programmed death receptor ligand-1 (PD-L1) in colorectal cancer specimens. Heliyon 2020; 7:e05880. [PMID: 33458446 PMCID: PMC7797507 DOI: 10.1016/j.heliyon.2020.e05880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/28/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
Aim PD-1/PD-L1 blockade therapy is now widely used for the treatment of advanced malignancies. Although PD-L1 is known to be expressed by various host cells as well as tumor cells, the role of PD-L1 on non-malignant cells and its clinical significance is unknown. We evaluated cell type-specific expression of PD-L1 in colorectal cancer (CRC) specimens using multicolor flow cytometry. Methods Single cell suspensions were made from 21 surgically resected CRC specimens, and immunostained with various mAbs conjugated with different fluorescent dyes. Tumor cells, stromal cells, and immune cells were identified as CD326(+), CD90(+) and CD45(+) phenotype, respectively. CD11b(+) myeloid cells, CD19(+) B cells and CD4(+) or CD8(+) T cells were also stained in different samples, and their frequencies in the total cell population and the ratio of PD-L1(+) cells to each phenotype were determined. Results PD-L1 was expressed by all the cell types. The ratio of PD-L1(+) cells to CD326(+) tumor cells was 19.1% ± 14.0%, lower than those for CD90(+) stromal cells (39.6% ± 16.0%) and CD11b(+) myeloid cells (31.9% ± 14.3%). The ratio of PD-L1(+) cells in tumor cells correlated strongly with the ratio in stromal cells, while only weakly with that in myeloid cells. Tumor cells were divided into two populations by CD326 expression levels, and the PD-L1 positive ratios were inversely correlated with the rate of CD326 highly expressing cells as well as mean fluorescein intensity of CD326 in tumor cells, while positively correlated with the frequencies of stromal cells or myeloid cells in CRC. Conclusion PD-L1 is differentially expressed on various cell types in CRC. PD-L1 on tumor cells may be upregulated together with CD326 downregulation in the process of epithelial mesenchymal transition. Quantification of cell type-specific expression of PD-L1 using multicolor flow cytometry may provide useful information for the immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Akira Saito
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Mineyuki Tojo
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Yuko Kumagai
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Hideyuki Ohzawa
- Department of Clinical Oncology, Jichi Medical University, Japan
| | | | - Hideyo Miyato
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Ai Sadatomo
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Daishi Naoi
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Gaku Ota
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Koji Koinuma
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Hisanaga Horie
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | | | - Naohiro Sata
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
| | - Joji Kitayama
- Department of Gastrointestinal Surgery, Jichi Medical University, Japan
- Corresponding author.
| |
Collapse
|
6
|
He H, Liu D, Feng Z, Guo A, Liu L, Chen X. Antifade Carbon Dots on a Plasmonic Substrate for Enhanced Protein Detection in Immunotherapy. ACS Sens 2020; 5:4027-4034. [PMID: 33253549 DOI: 10.1021/acssensors.0c01983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fluorescence microscopic analysis of checkpoint protein expression is capable of predicting clinical outcomes for checkpoint blockade immunotherapy. However, accurate detection of their expression levels is hindered by fluorophore photobleaching and cell autofluorescence. We now develop a sensitive and robust fluorescence microscopy method that uses antifade graphite-structured carbon dots (GCDs) on a plasmonic Ag substrate (named ACPAS) for the accurate detection of checkpoint proteins in immunotherapy. In ACPAS, a Ag substrate is used to enhance the fluorescence of GCDs while a continuous illumination is implemented to quench cell autofluorescence, thus enabling a dramatic improvement in the signal-to-background ratio by up to 33-fold. We use ACPAS to monitor programmed death ligand-1 (PD-L1) expression levels on various tumor cells and finely differentiate their microscopic changes in combination with chemokine receptor CXCR4-targeted treatments. ACPAS analysis reveals for the first time that CXCR4 agonist (SDF-1α) and antagonist (AMD3100) can potentiate PD-L1 expression by down-regulating CXCR4 expression on tumor cells, which provides valuable information on the development of anti-PD-L1 and anti-CXCR4 combination therapy. We envision that ACPAS will become a broadly useful tool for protein expression studies in biomedicine and life sciences.
Collapse
Affiliation(s)
- Hua He
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Di Liu
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Zhenzhen Feng
- Technical Center of Qingdao Customs District, Qingdao 266500, China
| | - Aijun Guo
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Lihua Liu
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiaoliang Chen
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
7
|
Wang X, Jaimes M, Gu H, Shults K, Putta S, Sharma V, Chow W, Gogoi P, Handique K, Patterson BK. Cell by cell immuno- and cancer marker profiling of non-small cell lung cancer tissue: Checkpoint marker expression on CD103+, CD4+ T-cells predicts circulating tumor cells. Transl Oncol 2020; 14:100953. [PMID: 33217647 PMCID: PMC7683336 DOI: 10.1016/j.tranon.2020.100953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has a poor prognosis. Targeted therapy and immunotherapy in recent years has significantly improved NSCLC patient outcome. In this study, we employed cell-by-cell immune and cancer marker profiling of the primary tumor cells to investigate possible signatures that might predict the presence or absence of circulating tumor cells (CTCs). We performed a comprehensive study on 10 NSCLC patient tissue samples with paired blood samples. The solid tissue biopsy samples were dissociated into single cells by non-enzymatic tissue homogenization and stained with a total 25 immune, cancer markers and DNA content dye and analyzed with high-parameter flow cytometry. CTCs were isolated and analyzed from the paired peripheral blood. We investigated a total of 74 biomarkers for their correlation with CTC number. Strong correlations were observed between CTC number and the frequency of immune checkpoint marker expressing lymphocytes (CTLA-4, LAG3, TIM3, PD-1), within the CD103+CD4+ T lymphocyte subset. CTC number is also correlated with the frequency of PD-L1 expressing cancer cells and cancer cell DNA content. In contrast, CTC number inversely correlated to the frequency of CD44+E-cadherin- cancer cells. Unsupervised clustering analysis based on the biomarker analysis separated the CTC negative patients from the CTC positive patients. Profiling multiple immune and cancer markers on cancer samples with multi-parametric flow cytometry allowed us to obtain protein expression information at the single cell level. Clustering analysis of the proteomic data revealed a signature driven by checkpoint marker expression on CD103+CD4+ T cells that could potentially be predictive of CTCs and targets of therapy.
Collapse
Affiliation(s)
- Xiaoyang Wang
- IncellDx Inc, 1541 Industrial Road, San Carlos, CA, United States
| | - Maria Jaimes
- Cytek Biosciences Inc, Fremont, CA, United States
| | - Huimin Gu
- Cytek Biosciences Inc, Fremont, CA, United States
| | - Keith Shults
- IncellDx Inc, 1541 Industrial Road, San Carlos, CA, United States
| | | | | | - Will Chow
- Celsee Inc, Plymouth MI, United States
| | | | | | | |
Collapse
|
8
|
Zhu Y, Zalaznick J, Sleczka B, Parrish K, Yang Z, Olah T, Shipkova P. Immunoaffinity microflow liquid chromatography/tandem mass spectrometry for the quantitation of PD1 and PD-L1 in human tumor tissues. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34:e8896. [PMID: 32666620 DOI: 10.1002/rcm.8896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 06/11/2023]
Abstract
RATIONALE High tumor expression of programmed cell death protein (PD1) and programmed death-ligand 1 (PD-L1) is thought to be associated with positive clinical outcomes after treatment with anti-PD1 or anti-PD-L1 agents. Several sensitive methods based on immunohistochemistry, ligand binding assay (LBA), and liquid chromatography/mass spectrometry involving the measurement of PD1 and PD-L1 expression have been reported. Here, we expand on the characterization of different tumor types using a highly specific, sensitive, and robust immunoaffinity liquid chromatography/tandem mass spectrometry (IA-LC/MS/MS)-based method for the simultaneous quantitation of PD1 and PD-L1 in tumor tissues. METHODS Human tumor tissue samples were homogenized using a Precellys Evolution homogenizer. The samples were incubated with anti-PD1 and anti-PD-L1 capture polyclonal antibodies, which were bound to magnetic beads. Following enrichment, samples were digested with trypsin. A Waters iKEY HSS T3 1.8 um (150 μm × 100 mm) column with a gradient flow rate of 3 μL/min was used for chromatographic separation, and a Waters TQ-S triple quadrupole mass spectrometer was used for detection. Selected reaction monitoring (SRM) transitions with unit resolution for precursor/product ion masses were optimized for PD1 and PD-L1 surrogate peptides. RESULTS The surrogate peptides LAAFPEDR for PD1 and FTVTVPK for PD-L1 yielded the most intense SRM transitions at m/z 459.7 > 516.2 and m/z 396.2 > 543.3, respectively, and thus were selected for the quantitation of PD1 and PD-L1. The lower limit of quantitation for PD1 and PD-L1 was 0.062 ng/mL with an assay range up to 10 ng/mL. Using this method, human PD1 and PD-L1 were detected and quantified from four different types of tumor tissues. The data show that PD1 expression level was highly correlated with that of PD-L1 in all tumor tissues analyzed here. CONCLUSIONS A highly specific and sensitive immunoaffinity microflow LC/MS/MS method for the simultaneous quantification of PD1 and PD-L1 in tumor tissues was developed and implemented. This method combines the advantage of immuno-capture for analyte enrichment with the high specificity of detection of multiple surrogate peptides by LC/MS/MS. The quantification of PD1 and PD-L1 co-expression in tumor could help evaluate their role in assessing tumor type selection and patient stratification.
Collapse
Affiliation(s)
- Yongxin Zhu
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Jacob Zalaznick
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Bogdan Sleczka
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Karen Parrish
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Zheng Yang
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Timothy Olah
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Petia Shipkova
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| |
Collapse
|
9
|
PD-L1 Detection-Pearls and Pitfalls Associated With Current Methodologies Focusing on Entities Relevant to Dermatopathology. Am J Dermatopathol 2020; 41:539-565. [PMID: 31335407 DOI: 10.1097/dad.0000000000001287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PD-L1 is a transmembrane glycoprotein with an extracellular as well as an intracellular cytoplasmic domain. Physiologically, it plays a pivotal role in regulating T-cell activation and tolerance. Many tumor cells have exploited this regulatory mechanism by overexpressing PD-L1 in an effort to escape immunologic surveillance. In this review, we parse the literature regarding the prognostic value of tumoral PD-L1 expression before discussing the various methodologies as well as the pearls and pitfalls associated with each for predicting response to anti-PD-1/PD-L1 therapies. Special attention is given to cutaneous entities in which PD-L1 expression has been documented with an emphasis on cutaneous malignancies that have seen the broadest applications of anti-PD-L1/PD-1 therapies. Currently, immunohistochemistry is the method that is most commonly used for detection of PD-L1. However, with the wide array of immunohistochemistry protocols and staining platforms available in the market, there seems to be different cutoffs not just for different entities but also for the same entity. This review is an attempt to address the need for standardization and validation of existing protocols for PD-L1 detection.
Collapse
|
10
|
Alanee S, Deebajah M, Chen PI, Mora R, Guevara J, Francisco B, Patterson BK. Using adaptive genetic algorithms combined with high sensitivity single cell-based technology to detect bladder cancer in urine and provide a potential noninvasive marker for response to anti-PD1 immunotherapy. Urol Oncol 2019; 38:77.e9-77.e15. [PMID: 31570249 DOI: 10.1016/j.urolonc.2019.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/23/2019] [Accepted: 08/30/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To use adaptive genetic algorithms (AGA) in combination with single-cell flow cytometry technology to develop a noninvasive test to detect bladder cancer. MATERIALS AND METHODS Fifty high grade, cystoscopy confirmed, superficial bladder cancer patients, and 15 healthy donor early morning urine samples were collected in an optimized urine collection media. These samples were then used to develop an assay to distinguish healthy from cancer patients' urine using AGA in combination with single-cell flow cytometry technology. Cell recovery and test performance were verified based on cystoscopy and histology for both bladder cancer determination and PD-L1 status. RESULTS Bladder cancer patients had a significantly higher percentage of white blood cells with substantial PD-L1 expression (P< 0.0001), significantly increased post-G1 epithelial cells (P < 0.005) and a significantly higher DNA index above 1.05 (P < 0.05). AGA allowed parameter optimization to differentiate normal from malignant cells with high accuracy. The resulting prediction model showed 98% sensitivity and 87% specificity with a high area under the ROC value (90%). CONCLUSIONS Using single-cell technology and machine learning; we developed a new assay to distinguish bladder cancer from healthy patients. Future studies are planned to validate this assay.
Collapse
Affiliation(s)
- Shaheen Alanee
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI.
| | - Mustafa Deebajah
- Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI
| | | | - Rodrigo Mora
- DCX XLab, Lab of Cancer and Surgery, The University of Costa Rica, San José, Costa Rica
| | - Jose Guevara
- DCX XLab, Lab of Cancer and Surgery, The University of Costa Rica, San José, Costa Rica
| | | | | |
Collapse
|
11
|
Feng E, Zheng T, He X, Chen J, Tian Y. A novel ternary heterostructure with dramatic SERS activity for evaluation of PD-L1 expression at the single-cell level. SCIENCE ADVANCES 2018; 4:eaau3494. [PMID: 30406203 PMCID: PMC6214639 DOI: 10.1126/sciadv.aau3494] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/02/2018] [Indexed: 05/03/2023]
Abstract
Surface-enhanced Raman scattering (SERS) probes based on a charge transfer (CT) process with high stability and reproducibility are powerful tools under open-air conditions. However, the key problem ahead of practical usage of CT-based SERS technology is how to effectively improve sensitivity. Here, a novel ternary heterostructure SERS substrate, Fe3O4@GO@TiO2, with a significant enhancement factor of 8.08 × 106 was first synthesized. We found the remarkable enhanced effect of SERS signal to be attributed to the resonance effect of CuPc, CT between GO and TiO2, and enrichment from a porous TiO2 shell. In addition, we developed a robust SERS probe with good recyclability under visible light illumination on Fe3O4@GO@TiO2 nanocomposites toward ultrasensitive detection of cancer cells down to three cells. We have now successfully applied this probe for in situ quantification and imaging of programmed cell death receptor ligand 1 (PD-L1) on triple-negative breast cancer cell surface at the single-cell level and for monitoring the expression variation of PD-L1 during drug treatment.
Collapse
Affiliation(s)
- Enduo Feng
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai 200241, P. R. China
| | - Tingting Zheng
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai 200241, P. R. China
- Corresponding author. (T.Z.); (Y.T.)
| | - Xiaoxiao He
- State Key Laboratory of Precision Spectroscopy, East China Normal University, North Zhongshan Road 3663, Shanghai 200062, P. R. China
| | - Jinquan Chen
- State Key Laboratory of Precision Spectroscopy, East China Normal University, North Zhongshan Road 3663, Shanghai 200062, P. R. China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai 200241, P. R. China
- Corresponding author. (T.Z.); (Y.T.)
| |
Collapse
|
12
|
Young S, Griego-Fullbright C, Wagner A, Chargin A, Patterson BK, Chabot-Richards D. Concordance of PD-L1 Expression Detection in Non-Small Cell Lung Cancer (NSCLC) Tissue Biopsy Specimens Between OncoTect iO Lung Assay and Immunohistochemistry (IHC). Am J Clin Pathol 2018; 150:346-352. [PMID: 30052717 DOI: 10.1093/ajcp/aqy063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES We report on the validity of a fully quantitative technology to determine tumor cells' PD-L1 expression compared with a standard immunohistochemical (IHC) assay in non-small cell lung cancer. METHODS Nineteen fresh tissue specimens were processed into single-cell suspensions using the IncellPREP Kit. Cells were treated with the OncoTect iO Lung Assay, which quantitatively assessed tumor-infiltrating lymphocytes (TILs), DNA content, and PD-L1 expression on diploid and aneuploid tumor populations. RESULTS Comparison of the OncoTect iO Lung Assay with IHC revealed a concordance of 95% overall (negative percent agreement, 97%; positive percent agreement, 89%). PD-L1 expression varied depending on the DNA content. The number of TILs and antigen-presenting cells (APCs) was significantly decreased in tumor compared with normal lung tissue. CONCLUSIONS The nonsubjective OncoTect iO Lung Assay has been shown to be at least as accurate and sensitive as IHC for the detection of PD-L1 expression while providing additional information to guide treatment.
Collapse
|
13
|
Tallón de Lara P, Cecconi V, Hiltbrunner S, Yagita H, Friess M, Bode B, Opitz I, Vrugt B, Weder W, Stolzmann P, Felley-Bosco E, Stahel RA, Tischler V, Britschgi C, Soldini D, van den Broek M, Curioni-Fontecedro A. Gemcitabine Synergizes with Immune Checkpoint Inhibitors and Overcomes Resistance in a Preclinical Model and Mesothelioma Patients. Clin Cancer Res 2018; 24:6345-6354. [PMID: 30154226 DOI: 10.1158/1078-0432.ccr-18-1231] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/24/2018] [Accepted: 08/21/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Combination of immune checkpoint inhibitors with chemotherapy is under investigation for cancer treatment. EXPERIMENTAL DESIGN We studied the rationale of such a combination for treating mesothelioma, a disease with limited treatment options. RESULTS The combination of gemcitabine and immune checkpoint inhibitors outperformed immunotherapy alone with regard to tumor control and survival in a preclinical mesothelioma model; however, the addition of dexamethasone to gemcitabine and immune checkpoint inhibitors nullified the synergistic clinical response. Furthermore, treatment with gemcitabine plus anti-PD-1 resulted in an objective clinical response in two patients with mesothelioma, who were resistant to gemcitabine or anti-PD-1 as monotherapy. CONCLUSIONS Thus, treatment of mesothelioma with a combination of gemcitabine with immune checkpoint inhibitors is feasible and results in synergistic clinical response compared with single treatment in the absence of steroids.
Collapse
Affiliation(s)
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Martina Friess
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Beata Bode
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Bart Vrugt
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Paul Stolzmann
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | | | - Rolf A Stahel
- Department of Hematology and Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Verena Tischler
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Christian Britschgi
- Department of Hematology and Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Davide Soldini
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| | | |
Collapse
|
14
|
Ilie M, Hofman P. Atezolizumab in advanced non-small cell lung cancer. J Thorac Dis 2017; 9:3603-3606. [PMID: 29268352 PMCID: PMC5723868 DOI: 10.21037/jtd.2017.09.73] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 09/13/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Marius Ilie
- Université Côte d'Azur, University Hospital Federation OncoAge, Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Nice, France
- Université Côte d'Azur, Institute for Research on Cancer and Ageing, Nice (IR-CAN), Inserm U1081 and UMR CNRS 7284, Nice, France
- Université Côte d'Azur, University Hospital Federation OncoAge, Hospital-Related Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France
| | - Paul Hofman
- Université Côte d'Azur, University Hospital Federation OncoAge, Laboratory of Clinical and Experimental Pathology, Hôpital Pasteur, Nice, France
- Université Côte d'Azur, Institute for Research on Cancer and Ageing, Nice (IR-CAN), Inserm U1081 and UMR CNRS 7284, Nice, France
- Université Côte d'Azur, University Hospital Federation OncoAge, Hospital-Related Biobank (BB-0033-00025), Hôpital Pasteur, Nice, France
| |
Collapse
|
15
|
Chow JCH, Cheung KM, Cho WCS. Atezolizumab in non-small cell lung cancer: the era of precision immuno-oncology. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:265. [PMID: 28706933 DOI: 10.21037/atm.2017.03.89] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
| | - Ka Man Cheung
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong 999077, China
| | | |
Collapse
|
16
|
Takahashi Y, Matsutani N, Nakayama T, Dejima H, Uehara H, Kawamura M. Immunological effect of local ablation combined with immunotherapy on solid malignancies. CHINESE JOURNAL OF CANCER 2017; 36:49. [PMID: 28592286 PMCID: PMC5463413 DOI: 10.1186/s40880-017-0216-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 05/30/2017] [Indexed: 02/07/2023]
Abstract
Recent comprehensive investigations clarified that immune microenvironment surrounding tumor cells are deeply involved in tumor progression, metastasis, and response to treatment. Furthermore, several immunotherapeutic trials have achieved successful results, and the immunotherapeutic agents are available in clinical practice. To enhance their demonstrated efficacy, combination of immunotherapy and ablation has begun to emerge. Local ablations have considerable advantages as an alternative therapeutic option, especially its minimal invasiveness. In addition, local ablations have shown immune-regulatory effect in preclinical and clinical studies. Although the corresponding mechanisms are still unclear, the local ablations combined with immunotherapy have been suggested in the treatment of several solid malignancies. This article aims to review the published data on the immune-regulatory effects of local ablations including stereotactic body radiotherapy, cryoablation, radiofrequency ablation, and high-intensity-focused ultrasound. We also discuss the value of local ablations combined with immunotherapy. Local ablations have the potential to improve future patient outcomes; however, the effectiveness and safety of local ablations combined with immunotherapy should be further investigated.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan.
| | - Noriyuki Matsutani
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Takashi Nakayama
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Hitoshi Dejima
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Hirofumi Uehara
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Masafumi Kawamura
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| |
Collapse
|