1
|
Sanavandi M, Aalikhani K, Shafiee M, Rabbani H, Fazli G, Sadeghi N, Shokri B. A One-Step Plasma Assisted Synthesis of Gold Nanoparticles and Simultaneous Linker-Free Conjugation with Nestin: An In Vitro Study of Cellular Toxicity. NANO LETTERS 2025; 25:1974-1983. [PMID: 39868719 DOI: 10.1021/acs.nanolett.4c05641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
We present a method for conjugating antigens to gold nanoparticles (GNPs) during their synthesis via gas plasma, eliminating the need for chemical linkers and significantly speeding up the process (taking only 15 min). This fast, linker-free method produces biocompatible and stable GNPs, with potential for immunotherapy applications, such as antigen and antibody conjugation and drug delivery. We demonstrate the conjugation of the antigen Nestin (NES), a tumor marker, to GNPs using two approaches. The first method involves synthesizing GNPs with citrate, followed by NES conjugation via plasma. The second method synthesizes and conjugates GNPs to NES simultaneously using plasma treatment. Conjugation was confirmed by Enzyme-Linked Immunosorbent Assay, Zeta-sizer, UV-vis spectroscopy, and Transmission Electron Microscopy. In addition, the toxicity of the prepared samples was investigated in vitro using peripheral blood mononuclear cells (PBMCs) and flow cytometry, which proved the nontoxicity of the samples.
Collapse
Affiliation(s)
- Melika Sanavandi
- Department of Physics, Shahid Beheshti University, Tehran 1635649771, Iran
| | - Kimia Aalikhani
- Department of Physics, Shahid Beheshti University, Tehran 1635649771, Iran
| | - Mojtaba Shafiee
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran 1658645578, Iran
| | - Hodjatallah Rabbani
- Institute of Medical Biotechnology, National Institute of Genetic Engineering & Biotechnology (NIGEB), Tehran 1983969412, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Shahid Beheshti University, Tehran 1985743413, Iran
| | - Ghazaleh Fazli
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Shahid Beheshti University, Tehran 1985743413, Iran
| | - Niloufar Sadeghi
- Institute of Medical Biotechnology, National Institute of Genetic Engineering & Biotechnology (NIGEB), Tehran 1983969412, Iran
| | - Babak Shokri
- Department of Physics, Shahid Beheshti University, Tehran 1635649771, Iran
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran 1658645578, Iran
| |
Collapse
|
2
|
Patel RK, Parappilly M, Farley HC, Latour EJ, Wang LG, Nair AM, Lu ES, Sims Z, Park B, Nelson K, Mayo SC, Mills GB, Sheppard BC, Chang YH, Gibbs SL, Kardosh A, Lopez CD, Wong MH. Circulating Neoplastic-Immune Hybrid Cells Are Biomarkers of Occult Metastasis and Treatment Response in Pancreatic Cancer. Cancers (Basel) 2024; 16:3650. [PMID: 39518088 PMCID: PMC11545756 DOI: 10.3390/cancers16213650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) presents significant diagnostic and prognostic challenges, as current biomarkers frequently fail to accurately stage disease, predict rapid metastatic recurrence (rPDAC), or assess response to neoadjuvant therapy (NAT). We investigated the potential for circulating neoplastic-immune hybrid cells (CHCs) as a non-invasive, multifunctional biomarker for PDAC. METHODS Peripheral blood specimens were obtained from patients diagnosed with PDAC. CHCs were detected by co-expression of pan-cytokeratin and CD45, normalized to 50,000 peripheral blood mononuclear cells. rPDAC was defined as metastatic recurrence within six months of margin-negative pancreatectomy. Cyclic immunofluorescence (CyCIF) analyses compared hybrid phenotypes in blood and tumors. RESULTS Blood samples were collected from 42 patients with PDAC prior to resection. Those with radiographically occult metastatic disease and rPDAC had higher preoperative CHC numbers compared to patients who did not (65.0 and 74.4, vs. 11.52 CHCs; p < 0.001). Patients with complete or near-complete pathologic responses to NAT had lower preoperative CHC numbers than partial and/or non-responders (1.7 vs. 13.1 CHCs; p = 0.008). When assessed longitudinally, those with partial pathologic response saw CHC levels become undetectable while on treatment but increase in the interval between NAT completion and resection. In contrast, patients with poor responses or development of metastatic disease experienced persistent CHC detection during therapy or rising levels prior to radiographic evidence of metastases. Further, in metastatic PDAC patients, treatment-induced phenotypic changes in hybrid cells mirrored those in paired metastatic tumor samples. CONCLUSIONS CHC enumeration and phenotyping display promise as a real-time indicator of disease burden, recurrence risk, and treatment response in PDAC. CHCs have great potential as tumor-derived biomarkers to optimize therapeutic strategies and improve survival in patients with PDAC.
Collapse
Affiliation(s)
- Ranish K. Patel
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
| | - Michael Parappilly
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR 97201, USA
| | - Hannah C. Farley
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR 97201, USA
| | - Emile J. Latour
- Biostatistics Shared Resource, Knight Cancer Institute, OHSU, Portland, OR 97239, USA
| | - Lei G. Wang
- Department of Biomedical Engineering, OHSU, Portland, OR 97201, USA
| | - Ashvin M. Nair
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR 97201, USA
| | - Ethan S. Lu
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR 97201, USA
| | - Zachary Sims
- Department of Biomedical Engineering, OHSU, Portland, OR 97201, USA
| | - Byung Park
- Biostatistics Shared Resource, Knight Cancer Institute, OHSU, Portland, OR 97239, USA
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
| | - Katherine Nelson
- Gastrointestinal Clinical Trials, Knight Cancer Institute, OHSU, Portland, OR 97239, USA
| | - Skye C. Mayo
- Department of Surgery, Division of Surgical Oncology, Oregon Health & Science University (OHSU), Portland, OR 97239, USA; (R.K.P.)
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
| | - Gordon B. Mills
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
- Division of Oncological Sciences, Knight Cancer Institute, OHSU, Portland, OR 97239, USA
| | - Brett C. Sheppard
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
- Department of Surgery, Division of General Surgery, OHSU, Portland, OR 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, OHSU, Portland, OR 97201, USA
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
| | - Summer L. Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR 97201, USA
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
| | - Adel Kardosh
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, OHSU, Portland, OR 97239, USA
| | - Charles D. Lopez
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
- Department of Medicine, Division of Medical Oncology, OHSU, Portland, OR 97239, USA
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR 97201, USA
- Knight Cancer Institute, OHSU, Portland, OR 97201, USA
| |
Collapse
|
3
|
Gustafson KT, Sayar Z, Modestino A, Le HH, Gower A, Civitci F, Esener SC, Heller MJ, Eksi SE. Oligo cyc-DEP: On-chip cyclic immunofluorescence profiling of cell-derived nanoparticles. Electrophoresis 2024; 45:1715-1720. [PMID: 39049673 DOI: 10.1002/elps.202400088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/29/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
We present a follow-on technique for the cyclic-immunofluorescence profiling of suspension particles isolated using dielectrophoresis. The original lab-on-chip technique ("cyc-DEP" [cyclic immunofluorescent imaging on dielectrophoretic chip]) was designed for the multiplex surveillance of circulating biomarkers. Nanoparticles were collected from low-volume liquid biopsies using microfluidic dielectrophoretic chip technology. Subsequent rounds of cyclic immunofluorescent labeling and quenching were imaged and quantified with a custom algorithm to detect multiple proteins. While cyc-DEP improved assay multiplicity, long runtimes threatened its clinical adoption. Here, we modify the original cyc-DEP platform to reduce assay runtimes. Nanoparticles were formulated from human prostate adenocarcinoma cells and collected using dielectrophoresis. Three proteins were labeled on-chip with a mixture of short oligonucleotide-conjugated antibodies. The sample was then incubated with complementary fluorophore-conjugated oligonucleotides, which were dehybridized using an ethylene carbonate buffer after each round of imaging. Oligonucleotide removal exhibited an average quenching efficiency of 98 ± 3% (n = 12 quenching events), matching the original cyc-DEP platform. The presented "oligo cyc-DEP" platform achieved clinically relevant sample-to-answer times, reducing the duration for three rounds of cyclic immunolabeling from approximately 20 to 6.5 h-a 67% decrease attributed to rapid fluorophore removal and the consolidated co-incubation of antibodies.
Collapse
Affiliation(s)
- Kyle T Gustafson
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Zeynep Sayar
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Augusta Modestino
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Hillary H Le
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Austin Gower
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Fehmi Civitci
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Sadik C Esener
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Michael J Heller
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Sebnem Ece Eksi
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
4
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Pang A, Farley HC, Gillingham AR, Dawson AD, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Detection of neoplastic-immune hybrid cells with metastatic properties in uveal melanoma. Biomark Res 2024; 12:67. [PMID: 39030653 PMCID: PMC11264923 DOI: 10.1186/s40364-024-00609-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/18/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Uveal melanoma is the most common non-cutaneous melanoma and is an intraocular malignancy affecting nearly 7,000 individuals per year worldwide. Of these, approximately 50% will progress to metastatic disease for which there are currently no effective curative therapies. Despite advances in molecular profiling and metastatic stratification of uveal melanoma tumors, little is known regarding their underlying biology of metastasis. Our group has identified a disseminated neoplastic cell population characterized by co-expression of immune and melanoma proteins, circulating hybrid cells (hybrids), in patients with uveal melanoma. Compared to circulating tumor cells, which lack expression of immune proteins, hybrids are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. METHODS To ascertain mechanisms underlying enhanced hybrid cell dissemination we identified hybrid cells within primary uveal melanoma tumors using single cell RNA sequencing (n = 8) and evaluated their gene expression and predicted ligand-receptor interactions in relation to other melanoma and immune cells within the primary tumor. We then verified expression of upregulated hybrid pathways within patient-matched tumor and peripheral blood hybrids (n = 4) using cyclic immunofluorescence and quantified their protein expression relative to other non-hybrid tumor and disseminated tumor cells. RESULTS Among the top upregulated genes and pathways in hybrid cells were those involved in enhanced cell motility and cytoskeletal rearrangement, immune evasion, and altered cellular metabolism. In patient-matched tumor and peripheral blood, we verified gene expression by examining concordant protein expression for each pathway category: TMSB10 (cell motility), CD74 (immune evasion) and GPX1 (metabolism). Both TMSB10 and GPX1 were expressed on significantly higher numbers of disseminated hybrid cells compared to circulating tumor cells, and CD74 and GPX1 were expressed on more disseminated hybrids than tumor-resident hybrids. Lastly, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting metastasis including GAS6-AXL, CXCL12-CXCR4, LGALS9-P4HB and IGF1-IGFR1. CONCLUSION These findings highlight the importance of TMSB10, GPX1 and CD74 for successful hybrid cell dissemination and survival in circulation. Our results contribute to the understanding of uveal melanoma tumor progression and interactions between tumor cells and immune cells in the tumor microenvironment that may promote metastasis.
Collapse
Affiliation(s)
- Ashley N Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Patrick Conley
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Christopher D Klocke
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
| | - Sidharth K Sengupta
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Amara Pang
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Hannah C Farley
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Abigail R Gillingham
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Aubrey D Dawson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Yichen Fan
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Alison H Skalet
- Casey Eye Institute, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Guanming Wu
- Department of Medical Informatics and Clinical Epidemiology, OHSU, Portland, OR, USA
- Knight Cancer Institute, OHSU, Portland, OR, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University (OHSU), Portland, OR, USA.
- Knight Cancer Institute, OHSU, Portland, OR, USA.
| |
Collapse
|
5
|
Reiber T, Dose C, Yushchenko DA. A novel dual-release scaffold for fluorescent labels improves cyclic immunofluorescence. RSC Chem Biol 2024; 5:684-690. [PMID: 38966675 PMCID: PMC11221530 DOI: 10.1039/d4cb00007b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024] Open
Abstract
Cyclic immunofluorescence is a powerful method to generate high-content imaging datasets for investigating cell biology and developing therapies. This method relies on fluorescent labels that determine the quality of immunofluorescence and the maximum number of staining cycles that can be performed. Here we present a novel fluorescent labelling strategy, based on antibodies conjugated to a scaffold containing two distinct sites for enzymatic cleavage of fluorophores. The scaffold is composed of a dextran decorated with short ssDNA that upon hybridization with complementary dye-modified oligos result in fluorescent molecules. The developed fluorescent labels exhibit specific staining and remarkable brightness in flow cytometry and fluorescence microscopy. We showed that the combination of DNase-mediated degradation of DNA and dextranse-mediated degradation of the dextran as two complementary enzymatic release mechanisms in one molecule, improves signal erasure from labelled epitopes. We envision that such dual-release labels with high brightness and efficient and specific erasure will advance multiplexed cyclic immunofluorescence approaches and thereby will contribute to gaining new insights in cell biology.
Collapse
Affiliation(s)
- Thorge Reiber
- Department of Chemical Biology, Miltenyi Biotec GmbH Friedrich-Ebert Straße 68 Bergisch Gladbach 51429 Germany
- Department of Chemistry, Humboldt-Universität zu Berlin Brook-Taylor-Str. 2 Berlin 12489 Germany
| | - Christian Dose
- Department of Chemical Biology, Miltenyi Biotec GmbH Friedrich-Ebert Straße 68 Bergisch Gladbach 51429 Germany
| | - Dmytro A Yushchenko
- Department of Chemical Biology, Miltenyi Biotec GmbH Friedrich-Ebert Straße 68 Bergisch Gladbach 51429 Germany
| |
Collapse
|
6
|
McMahon NP, Solanki A, Wang LG, Montaño AR, Jones JA, Samkoe KS, Tichauer KM, Gibbs SL. In situ single-cell therapeutic response imaging facilitated by the TRIPODD fluorescence imaging platform. Theranostics 2024; 14:2816-2834. [PMID: 38773974 PMCID: PMC11103495 DOI: 10.7150/thno.93256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/21/2024] [Indexed: 05/24/2024] Open
Abstract
Purpose: Small molecule drugs such as tyrosine kinase inhibitors (TKIs) targeting tumoral molecular dependencies have become standard of care for numerous cancer types. Notably, epidermal growth factor receptor (EGFR) TKIs (e.g., erlotinib, afatinib, osimertinib) are the current first-line treatment for non-small cell lung cancer (NSCLC) due to their improved therapeutic outcomes for EGFR mutated and overexpressing disease over traditional platinum-based chemotherapy. However, many NSCLC tumors develop resistance to EGFR TKI therapy causing disease progression. Currently, the relationship between in situ drug target availability (DTA), local protein expression and therapeutic response cannot be accurately assessed using existing analytical tools despite being crucial to understanding the mechanism of therapeutic efficacy. Procedure: We have previously reported development of our fluorescence imaging platform termed TRIPODD (Therapeutic Response Imaging through Proteomic and Optical Drug Distribution) that is capable of simultaneous quantification of single-cell DTA and protein expression with preserved spatial context within a tumor. TRIPODD combines two complementary fluorescence imaging techniques: intracellular paired agent imaging (iPAI) to measure DTA and cyclic immunofluorescence (cyCIF), which utilizes oligonucleotide conjugated antibodies (Ab-oligos) for spatial proteomic expression profiling on tissue samples. Herein, TRIPODD was modified and optimized to provide a downstream analysis of therapeutic response through single-cell DTA and proteomic response imaging. Results: We successfully performed sequential imaging of iPAI and cyCIF resulting in high dimensional imaging and biomarker assessment to quantify single-cell DTA and local protein expression on erlotinib treated NSCLC models. Pharmacodynamic and pharmacokinetic studies of the erlotinib iPAI probes revealed that administration of 2.5 mg/kg each of the targeted and untargeted probe 4 h prior to tumor collection enabled calculation of DTA values with high Pearson correlation to EGFR, the erlotinib molecular target, expression in the tumors. Analysis of single-cell biomarker expression revealed that a single erlotinib dose was insufficient to enact a measurable decrease in the EGFR signaling cascade protein expression, where only the DTA metric detected the presence of bound erlotinib. Conclusion: We demonstrated the capability of TRIPODD to evaluate therapeutic response imaging to erlotinib treatment as it relates to signaling inhibition, DTA, proliferation, and apoptosis with preserved spatial context.
Collapse
Affiliation(s)
- Nathan P. McMahon
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Allison Solanki
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Lei G. Wang
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Antonio R. Montaño
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Jocelyn A. Jones
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Kimberley S. Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Kenneth M. Tichauer
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Summer L. Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
7
|
Whalen RM, Anderson AN, Jones JA, Sims Z, Chang YH, Nederlof MA, Wong MH, Gibbs SL. Ultra high content analyses of circulating and tumor associated hybrid cells reveal phenotypic heterogeneity. Sci Rep 2024; 14:7350. [PMID: 38538742 PMCID: PMC10973471 DOI: 10.1038/s41598-024-57381-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/18/2024] [Indexed: 07/03/2024] Open
Abstract
Persistently high, worldwide mortality from cancer highlights the unresolved challenges of disease surveillance and detection that impact survival. Development of a non-invasive, blood-based biomarker would transform survival from cancer. We demonstrate the functionality of ultra-high content analyses of a newly identified population of tumor cells that are hybrids between neoplastic and immune cells in patient matched tumor and peripheral blood specimens. Using oligonucleotide conjugated antibodies (Ab-oligo) permitting cyclic immunofluorescence (cyCIF), we present analyses of phenotypes among tumor and peripheral blood hybrid cells. Interestingly, the majority of circulating hybrid cell (CHC) subpopulations were not identified in tumor-associated hybrids. These results highlight the efficacy of ultra-high content phenotypic analyses using Ab-oligo based cyCIF applied to both tumor and peripheral blood specimens. The combination of a multiplex phenotypic profiling platform that is gentle enough to analyze blood to detect and evaluate disseminated tumor cells represents a novel approach to exploring novel tumor biology and potential utility for developing the population as a blood-based biomarker in cancer.
Collapse
Affiliation(s)
- Riley M Whalen
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA
| | - Ashley N Anderson
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA
| | - Jocelyn A Jones
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
| | - Zachary Sims
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | | | - Melissa H Wong
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University (OHSU), Portland, OR, 97201, USA.
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.
| | - Summer L Gibbs
- Department of Biomedical Engineering, OHSU, Portland, OR, 97201, USA.
- Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.
| |
Collapse
|
8
|
Anderson AN, Conley P, Klocke CD, Sengupta SK, Pang A, Farley HC, Gillingham AR, Dawson AD, Fan Y, Jones JA, Gibbs SL, Skalet AH, Wu G, Wong MH. Detection of neoplastic-immune hybrid cells with metastatic properties in uveal melanoma. RESEARCH SQUARE 2023:rs.3.rs-3694879. [PMID: 38106024 PMCID: PMC10723549 DOI: 10.21203/rs.3.rs-3694879/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Uveal melanoma is the most common non-cutaneous melanoma and is an intraocular malignancy affecting nearly 7,000 individuals per year worldwide. Of these, approximately 50% will progress to metastatic disease for which there are currently no effective therapies. Despite advances in molecular profiling and metastatic stratification of uveal melanoma tumors, little is known regarding their underlying biology of metastasis. Our group has identified a disseminated neoplastic cell population characterized by co-expression of immune and melanoma proteins, circulating hybrid cells (hybrids), in patients with uveal melanoma. Compared to circulating tumor cells, which lack expression of immune proteins, hybrids are detected at an increased prevalence in peripheral blood and can be used as a non-invasive biomarker to predict metastatic progression. Methods To ascertain mechanisms underlying enhanced hybrid cell dissemination we identified hybrid cells within primary uveal melanoma tumors using single cell RNA sequencing and evaluated their gene expression and predicted ligand-receptor interactions in relation to other melanoma and immune cells within the primary tumor. We then verified expression of upregulated hybrid pathways within patient-matched tumor and peripheral blood hybrids using cyclic immunofluorescence and quantified their protein expression relative to other non-hybrid tumor and disseminated tumor cells. Results Among the top upregulated genes and pathways in hybrid cells were those involved in enhanced cell motility and cytoskeletal rearrangement, immune evasion, and altered cellular metabolism. In patient-matched tumor and peripheral blood, we verified gene expression by examining concordant protein expression for each pathway category: TMSB10 (cell motility), CD74 (immune evasion) and GPX1 (metabolism). Both TMSB10 and GPX1 were expressed on significantly higher numbers of disseminated hybrid cells compared to circulating tumor cells, and CD74 and GPX1 were expressed on more disseminated hybrids than tumor-resident hybrids. Lastly, we identified that hybrid cells express ligand-receptor signaling pathways implicated in promoting metastasis including GAS6-AXL, CXCL12-CXCR4, LGALS9-P4HB and IGF1-IGFR1. Conclusion These findings highlight the importance of TMSB10, GPX1 and CD74 for successful hybrid cell dissemination and survival in circulation. Our results contribute to the understanding of uveal melanoma tumor progression and interactions between tumor cells and immune cells in the tumor microenvironment that may promote metastasis.
Collapse
|
9
|
Anderson AN, Gibbs SL. Shooting for multiplexed pathology with Orion. NATURE CANCER 2023; 4:930-932. [PMID: 37495737 PMCID: PMC11262064 DOI: 10.1038/s43018-023-00590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Pathological diagnosis relies on morphological assessment of tissue using histological staining and molecular phenotyping through immunostaining that must be performed on separate tissue sections. Orion is a newly reported methodology that facilitates multiplexed immunostaining with histological staining on the same slide.
Collapse
Affiliation(s)
- Ashley N Anderson
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Summer L Gibbs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Ban Ž, Barišić A, Crnolatac I, Kazazić S, Škulj S, Savini F, Bertoša B, Barišić I, Piantanida I. Highly selective preparation of N-terminus Horseradish peroxidase-DNA conjugate with fully retained enzymatic activity: HRP-DNA structure - activity relation. Enzyme Microb Technol 2023; 168:110257. [PMID: 37209508 DOI: 10.1016/j.enzmictec.2023.110257] [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: 01/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
Within the last decade, the field of bio-nanoengineering has achieved significant advances allowing us to generate, e.g., nanoscaled molecular machineries with arbitrary shapes. To unleash the full potential of novel methods such as DNA origami technology, it is important to functionalise complex molecules and nanostructures precisely. Thus, considerable attention has been given to site-selective modifications of proteins allowing further incorporation of various functionalities. Here, we describe a method for the covalent attachment of oligonucleotides to the glycosylated horseradish peroxidase protein (HRP) with high N-terminus selectivity and significant yield while conserving the enzymatic activity. This two-step process includes a pH-controlled metal-free diazotransfer reaction using imidazole-1-sulfonyl azide hydrogen sulfate, which at pH 8.5 results in an N-terminal azide-functionalized protein, followed by the Cu-free click SPAAC reaction to dibenzocyclooctyne- (DBCO) modified oligonucleotides. The reaction conditions were optimised to achieve maximum yield and the best performance. The resulting protein-oligonucleotide conjugates (HRP-DNA) were characterised by electrophoresis and mass spectrometry (MS). Native-PAGE experiments demonstrated different migration patterns for HRP-DNA and the azido-modified protein allowing zymogram experiments. Structure-activity relationships of novel HRP-DNA conjugates were assessed using molecular dynamics simulations, characterising the molecular interactions that define the structural and dynamical properties of the obtained protein-oligonucleotide conjugates (POC).
Collapse
Affiliation(s)
- Željka Ban
- Division of Organic Chemistry & Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Antun Barišić
- Department of Chemistry, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Ivo Crnolatac
- Division of Organic Chemistry & Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia.
| | - Saša Kazazić
- Division of Organic Chemistry & Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Sanja Škulj
- Department of Chemistry, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | | | - Branimir Bertoša
- Department of Chemistry, Faculty of Science, University of Zagreb, Zagreb, Croatia.
| | - Ivan Barišić
- AIT Austrian Institute of Technology,Vienna, Austria.
| | - Ivo Piantanida
- Division of Organic Chemistry & Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
11
|
Sheng W, Zhang C, Mohiuddin TM, Al-Rawe M, Zeppernick F, Falcone FH, Meinhold-Heerlein I, Hussain AF. Multiplex Immunofluorescence: A Powerful Tool in Cancer Immunotherapy. Int J Mol Sci 2023; 24:ijms24043086. [PMID: 36834500 PMCID: PMC9959383 DOI: 10.3390/ijms24043086] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Traditional immunohistochemistry (IHC) has already become an essential method of diagnosis and therapy in cancer management. However, this antibody-based technique is limited to detecting a single marker per tissue section. Since immunotherapy has revolutionized the antineoplastic therapy, developing new immunohistochemistry strategies to detect multiple markers simultaneously to better understand tumor environment and predict or assess response to immunotherapy is necessary and urgent. Multiplex immunohistochemistry (mIHC)/multiplex immunofluorescence (mIF), such as multiplex chromogenic IHC and multiplex fluorescent immunohistochemistry (mfIHC), is a new and emerging technology to label multiple biomarkers in a single pathological section. The mfIHC shows a higher performance in cancer immunotherapy. This review summarizes the technologies, which are applied for mfIHC, and discusses how they are employed for immunotherapy research.
Collapse
Affiliation(s)
- Wenjie Sheng
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Chaoyu Zhang
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - T. M. Mohiuddin
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Marwah Al-Rawe
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Felix Zeppernick
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Franco H. Falcone
- Institute for Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Ivo Meinhold-Heerlein
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Ahmad Fawzi Hussain
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
12
|
McMahon NP, Jones JA, Anderson AN, Dietz MS, Wong MH, Gibbs SL. Flexible Cyclic Immunofluorescence (cyCIF) Using Oligonucleotide Barcoded Antibodies. Cancers (Basel) 2023; 15:827. [PMID: 36765785 PMCID: PMC9913741 DOI: 10.3390/cancers15030827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Advances in our understanding of the complex, multifaceted interactions between tumor epithelia, immune infiltrate, and tumor microenvironmental cells have been driven by highly multiplexed imaging technologies. These techniques are capable of labeling many more biomarkers than conventional immunostaining methods. However, multiplexed imaging techniques suffer from low detection sensitivity, cell loss-particularly in fragile samples-, and challenges with antibody labeling. Herein, we developed and optimized an oligonucleotide antibody barcoding strategy for cyclic immunofluorescence (cyCIF) that can be amplified to increase the detection efficiency of low-abundance antigens. Stained fluorescence signals can be readily removed using ultraviolet light treatment, preserving tissue and fragile cell sample integrity. We also extended the oligonucleotide barcoding strategy to secondary antibodies to enable the inclusion of difficult-to-label primary antibodies in a cyCIF panel. Using both the amplification oligonucleotides to label DNA barcoded antibodies and in situ hybridization of multiple fluorescently labeled oligonucleotides resulted in signal amplification and increased signal-to-background ratios. This procedure was optimized through the examination of staining parameters including staining oligonucleotide concentration, staining temperature, and oligonucleotide sequence design, resulting in a robust amplification technique. As a proof-of-concept, we demonstrate the flexibility of our cyCIF strategy by simultaneously imaging with the original oligonucleotide conjugated antibody (Ab-oligo) cyCIF strategy, the novel Ab-oligo cyCIF amplification strategy, as well as direct and indirect immunofluorescence to generate highly multiplexed images.
Collapse
Affiliation(s)
- Nathan P. McMahon
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Jocelyn A. Jones
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Ashley N. Anderson
- Department of Cell, Development & Cancer Biology Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Matthew S. Dietz
- Department of Cell, Development & Cancer Biology Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Melissa H. Wong
- Department of Cell, Development & Cancer Biology Department, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Summer L. Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
13
|
Martel R, Shen ML, DeCorwin-Martin P, de Araujo LO, Juncker D. Extracellular Vesicle Antibody Microarray for Multiplexed Inner and Outer Protein Analysis. ACS Sens 2022; 7:3817-3828. [PMID: 36515500 PMCID: PMC9791990 DOI: 10.1021/acssensors.2c01750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteins are found both outside and inside of extracellular vesicles (EVs) and govern the properties and functions of EVs, while also constituting a signature of the cell of origin and of biological function and disease. Outer proteins on EVs can be directly bound by antibodies to either enrich EVs, or probe the expression of a protein on EVs, including in a combinatorial manner. However, co-profiling of inner proteins remains challenging. Here, we present the high-throughput, multiplexed analysis of EV inner and outer proteins (EVPio). We describe the optimization of fixation and heat-induced protein epitope retrieval for EVs, along with oligo-barcoded antibodies and branched DNA signal amplification for sensitive, multiplexed, and high-throughput assays. We captured four subpopulations of EVs from colorectal cancer (CRC) cell lines HT29 and SW403 based on EpCAM, CD9, CD63, and CD81 expression, and quantified the co-expression of eight outer [integrins (ITGs) and tetraspanins] and four inner (heat shock, endosomal, and inner leaflet) proteins. The differences in co-expression patterns were consistent with the literature and known biological function. In conclusion, EVPio analysis can simultaneously detect multiple inner and outer proteins in EVs immobilized on a surface, opening the way to extensive combinatorial protein profiles for both discovery and clinical translation.
Collapse
Affiliation(s)
- Rosalie Martel
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Molly L. Shen
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Philippe DeCorwin-Martin
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Lorenna Oliveira
Fernandes de Araujo
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - David Juncker
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada,
| |
Collapse
|
14
|
Parappilly MS, Chin Y, Whalen RM, Anderson AN, Robinson TS, Strgar L, Sutton TL, Conley P, Klocke C, Gibbs SL, Chang YH, Wu G, Wong MH, Skalet AH. Circulating Neoplastic-Immune Hybrid Cells Predict Metastatic Progression in Uveal Melanoma. Cancers (Basel) 2022; 14:cancers14194617. [PMID: 36230539 PMCID: PMC9564048 DOI: 10.3390/cancers14194617] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 01/25/2023] Open
Abstract
Background: Uveal melanoma is an aggressive cancer with high metastatic risk. Recently, we identified a circulating cancer cell population that co-expresses neoplastic and leukocyte antigens, termed circulating hybrid cells (CHCs). In other cancers, CHCs are more numerous and better predict oncologic outcomes compared to circulating tumor cells (CTCs). We sought to investigate the potential of CHCs as a prognostic biomarker in uveal melanoma. Methods: We isolated peripheral blood monocular cells from uveal melanoma patients at the time of primary treatment and used antibodies against leukocyte and melanoma markers to identify and enumerate CHCs and CTCs by immunocytochemistry. Results: Using a multi-marker approach to capture the heterogeneous disseminated tumor cell population, detection of CHCs was highly sensitive in uveal melanoma patients regardless of disease stage. CHCs were detected in 100% of stage I-III uveal melanoma patients (entire cohort, n = 68), whereas CTCs were detected in 58.8% of patients. CHCs were detected at levels statically higher than CTCs across all stages (p = 0.05). Moreover, CHC levels, but not CTCs, predicted 3 year progression-free survival (p < 0.03) and overall survival (p < 0.04). Conclusion: CHCs are a novel and promising prognostic biomarker in uveal melanoma.
Collapse
Affiliation(s)
- Michael S. Parappilly
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Yuki Chin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Riley M. Whalen
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Ashley N. Anderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Trinity S. Robinson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Luke Strgar
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Computational Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Thomas L. Sutton
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Patrick Conley
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher Klocke
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Summer L. Gibbs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Computational Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Guanming Wu
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Melissa H. Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Correspondence: (M.H.W.); (A.H.S.)
| | - Alison H. Skalet
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Correspondence: (M.H.W.); (A.H.S.)
| |
Collapse
|
15
|
Giannikopoulos P, Parham DM. Pediatric Sarcomas: The Next Generation of Molecular Studies. Cancers (Basel) 2022; 14:2515. [PMID: 35626119 PMCID: PMC9139929 DOI: 10.3390/cancers14102515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Pediatric sarcomas constitute one of the largest groups of childhood cancers, following hematopoietic, neural, and renal lesions. Partly because of their diversity, they continue to offer challenges in diagnosis and treatment. In spite of the diagnostic, nosologic, and therapeutic gains made with genetic technology, newer means for investigation are needed. This article reviews emerging technology being used to study human neoplasia and how these methods might be applicable to pediatric sarcomas. Methods reviewed include single cell RNA sequencing (scRNAseq), spatial multi-omics, high-throughput functional genomics, and clustered regularly interspersed short palindromic sequence-Cas9 (CRISPR-Cas9) technology. In spite of these advances, the field continues to be challenged by a dearth of properly annotated materials, particularly from recurrences and metastases and pre- and post-treatment samples.
Collapse
Affiliation(s)
| | - David M. Parham
- Department of Anatomic Pathology, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
16
|
A framework for multiplex imaging optimization and reproducible analysis. Commun Biol 2022; 5:438. [PMID: 35545666 PMCID: PMC9095647 DOI: 10.1038/s42003-022-03368-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/14/2022] [Indexed: 01/05/2023] Open
Abstract
Multiplex imaging technologies are increasingly used for single-cell phenotyping and spatial characterization of tissues; however, transparent methods are needed for comparing the performance of platforms, protocols and analytical pipelines. We developed a python software, mplexable, for reproducible image processing and utilize Jupyter notebooks to share our optimization of signal removal, antibody specificity, background correction and batch normalization of the multiplex imaging with a focus on cyclic immunofluorescence (CyCIF). Our work both improves the CyCIF methodology and provides a framework for multiplexed image analytics that can be easily shared and reproduced. An approach for tissue image analysis applicable to highly multiplexed immunofluorescence imaging of the spatial distribution of multiple protein biomarkers is proposed, here applied to the analysis of multiplex IF using the multiplex imaging platform, CyCIF.
Collapse
|
17
|
Oligonucleotide conjugated antibody strategies for cyclic immunostaining. Sci Rep 2021; 11:23844. [PMID: 34903759 PMCID: PMC8668956 DOI: 10.1038/s41598-021-03135-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/26/2021] [Indexed: 11/09/2022] Open
Abstract
A number of highly multiplexed immunostaining and imaging methods have advanced spatial proteomics of cancer for improved treatment strategies. While a variety of methods have been developed, the most widely used methods are limited by harmful signal removal techniques, difficulties with reagent production and antigen sensitivity. Multiplexed immunostaining employing oligonucleotide (oligos)-barcoded antibodies is an alternative approach that is growing in popularity. However, challenges remain in consistent conjugation of oligos to antibodies with maintained antigenicity as well as non-destructive, robust and cost-effective signal removal methods. Herein, a variety of oligo conjugation and signal removal methods were evaluated in the development of a robust oligo conjugated antibody cyclic immunofluorescence (Ab-oligo cyCIF) methodology. Both non- and site-specific conjugation strategies were assessed to label antibodies, where site-specific conjugation resulted in higher retained binding affinity and antigen-specific staining. A variety of fluorescence signal removal methods were also evaluated, where incorporation of a photocleavable link (PCL) resulted in full fluorescence signal removal with minimal tissue disruption. In summary, this work resulted in an optimized Ab-oligo cyCIF platform capable of generating high dimensional images to characterize the spatial proteomics of the hallmarks of cancer.
Collapse
|
18
|
Durkee MS, Abraham R, Clark MR, Giger ML. Artificial Intelligence and Cellular Segmentation in Tissue Microscopy Images. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1693-1701. [PMID: 34129842 PMCID: PMC8485056 DOI: 10.1016/j.ajpath.2021.05.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
With applications in object detection, image feature extraction, image classification, and image segmentation, artificial intelligence is facilitating high-throughput analysis of image data in a variety of biomedical imaging disciplines, ranging from radiology and pathology to cancer biology and immunology. Specifically, a growth in research on deep learning has led to the widespread application of computer-visualization techniques for analyzing and mining data from biomedical images. The availability of open-source software packages and the development of novel, trainable deep neural network architectures has led to increased accuracy in cell detection and segmentation algorithms. By automating cell segmentation, it is now possible to mine quantifiable cellular and spatio-cellular features from microscopy images, providing insight into the organization of cells in various pathologies. This mini-review provides an overview of the current state of the art in deep learning- and artificial intelligence-based methods of segmentation and data mining of cells in microscopy images of tissue.
Collapse
Affiliation(s)
- Madeleine S Durkee
- Department of Radiology and the Committee on Medical Physics, University of Chicago, Chicago, Illinois.
| | - Rebecca Abraham
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, Illinois
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, Illinois
| | - Maryellen L Giger
- Department of Radiology and the Committee on Medical Physics, University of Chicago, Chicago, Illinois.
| |
Collapse
|
19
|
Kennedy‐Darling J, Bhate SS, Hickey JW, Black S, Barlow GL, Vazquez G, Venkataraaman VG, Samusik N, Goltsev Y, Schürch CM, Nolan GP. Highly multiplexed tissue imaging using repeated oligonucleotide exchange reaction. Eur J Immunol 2021; 51:1262-1277. [PMID: 33548142 PMCID: PMC8251877 DOI: 10.1002/eji.202048891] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/21/2020] [Accepted: 02/04/2021] [Indexed: 01/04/2023]
Abstract
Multiparameter tissue imaging enables analysis of cell-cell interactions in situ, the cellular basis for tissue structure, and novel cell types that are spatially restricted, giving clues to biological mechanisms behind tissue homeostasis and disease. Here, we streamlined and simplified the multiplexed imaging method CO-Detection by indEXing (CODEX) by validating 58 unique oligonucleotide barcodes that can be conjugated to antibodies. We showed that barcoded antibodies retained their specificity for staining cognate targets in human tissue. Antibodies were visualized one at a time by adding a fluorescently labeled oligonucleotide complementary to oligonucleotide barcode, imaging, stripping, and repeating this cycle. With this we developed a panel of 46 antibodies that was used to stain five human lymphoid tissues: three tonsils, a spleen, and a LN. To analyze the data produced, an image processing and analysis pipeline was developed that enabled single-cell analysis on the data, including unsupervised clustering, that revealed 31 cell types across all tissues. We compared cell-type compositions within and directly surrounding follicles from the different lymphoid organs and evaluated cell-cell density correlations. This sequential oligonucleotide exchange technique enables a facile imaging of tissues that leverages pre-existing imaging infrastructure to decrease the barriers to broad use of multiplexed imaging.
Collapse
Affiliation(s)
- Julia Kennedy‐Darling
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Akoya Biosciences1505 O'Brien DriveMenlo ParkCAUSA
| | - Salil S. Bhate
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
- Department of BioengineeringStanford UniversityStanfordCAUSA
| | - John W. Hickey
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Sarah Black
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Graham L. Barlow
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Gustavo Vazquez
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Vishal G. Venkataraaman
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Nikolay Samusik
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Becton DickinsonSan JoseCAUSA
| | - Yury Goltsev
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Christian M. Schürch
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
- Department of Pathology and NeuropathologyUniversity Hospital and Comprehensive Cancer Center TübingenTübingenGermany
| | - Garry P. Nolan
- Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| |
Collapse
|
20
|
McMahon NP, Solanki A, Wang LG, Montaño AR, Jones JA, Samkoe KS, Tichauer KM, Gibbs SL. TRIPODD: a Novel Fluorescence Imaging Platform for In Situ Quantification of Drug Distribution and Therapeutic Response. Mol Imaging Biol 2021; 23:650-664. [PMID: 33751366 DOI: 10.1007/s11307-021-01589-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 02/08/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Personalized medicine has largely failed to produce curative therapies in advanced cancer patients. Evaluation of in situ drug target availability (DTA) concomitant with local protein expression is critical to an accurate assessment of therapeutic efficacy, but tools capable of both are currently lacking. PROCEDURE We developed and optimized a fluorescence imaging platform termed TRIPODD (Therapeutic Response Imaging through Proteomic and Optical Drug Distribution), resulting in the only methodology capable of simultaneous quantification of single-cell DTA and protein expression with preserved spatial context within a tumor. Using TRIPODD, we demonstrate the feasibility of combining two complementary fluorescence imaging techniques, intracellular paired agent imaging (iPAI) and cyclic immunofluorescence (cyCIF), conducted with oligonucleotide-conjugated antibodies (Ab-oligos) on tissue samples. RESULTS We successfully performed sequential imaging on a single tissue section of iPAI to capture single-cell DTA and local protein expression heterogeneity using Ab-oligo cyCIF. Fluorescence imaging data acquisition was followed by spatial registration resulting in high dimensional data correlating DTA to protein expression at the single-cell level where uptake of a targeted probe alone was not well correlated to protein expression. CONCLUSION Herein, we demonstrated the utility of TRIPODD as a powerful imaging platform capable of interpreting tumor heterogeneity for a mechanistic understanding of therapeutic response and resistance through quantification of drug target availability and proteomic response with preserved spatial context at single-cell resolution.
Collapse
Affiliation(s)
- Nathan P McMahon
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Allison Solanki
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Lei G Wang
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Antonio R Montaño
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Jocelyn A Jones
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering at Dartmouth College, Dartmouth College, Hanover, NH, USA.,Department of Surgery, Geisel School of Medicine at Dartmouth College, Dartmouth College, Hanover, NH, 03755, USA
| | - Kenneth M Tichauer
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616, USA
| | - Summer L Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR, USA. .,Knight Cancer Institute, Oregon Health & Science University, Collaborative Life Sciences Building, 2730 S Moody Ave, Mail Code: CL3SG, Portland, OR, 97201, USA.
| |
Collapse
|