Fact-checked by Grok 2 weeks ago

CyTOF

CyTOF, also known as or , is a technology that combines the hydrodynamic focusing of with (ICP-TOF-MS) to simultaneously measure dozens of cellular parameters using stable metal isotope tags. In this technique, cells are labeled with antibodies conjugated to distinct metal isotopes, typically from the series, which are then vaporized into ions and analyzed based on their , allowing for high-resolution detection without the spectral overlap inherent in fluorescence-based methods. This enables the quantification of up to 50 or more protein markers per cell, far exceeding the 10–20 parameters typical of conventional . The technology was pioneered in the late 2000s by researchers at DVS Sciences, including Scott D. Tanner, Vladimir I. Baranov, and Dmitry R. Bandura, who published the first prototype description in 2009, adapting ICP-MS principles originally commercialized in the 1980s for biological applications. Following its introduction, DVS Sciences was acquired by Fluidigm (renamed in 2022 and merged with SomaLogic in 2024) in 2014, leading to commercial instruments like the CyTOF2 and systems, which process up to 1,000 cells per second with a detection efficiency of around 30%. Key advancements include the development of metal-chelating polymers for stable conjugation and intercalators like for viability and DNA staining, enhancing data quality and throughput. CyTOF has revolutionized fields such as , , and by facilitating deep phenotyping of immune cell subsets, tumor microenvironments, and signaling pathways in complex tissues. Notable applications include mapping hematopoietic differentiation hierarchies and profiling drug responses in , where its ability to handle rare cell populations (down to 10^6–10^7 total cells analyzed) provides quantitative, high-dimensional insights unattainable with traditional . Compared to , CyTOF offers superior without compensation artifacts, though it requires specialized and generates large datasets necessitating advanced computational tools like t-SNE or FlowSOM for analysis. Ongoing developments, including imaging mass cytometry (IMC) for and expanded reagent libraries, continue to broaden its utility in as of 2025.

Introduction

Definition and Scope

CyTOF, or Cytometry by Time-Of-Flight, is a proprietary platform developed by DVS Sciences—now integrated into —that facilitates flow-based analysis of single cells through . This system represents a specific implementation of technology, trademarked by Canada Inc., designed for high-throughput, multiplexed detection of cellular components at the individual cell level. At its core, CyTOF combines the hydrodynamic focusing and cell interrogation principles of traditional with (ICP-TOF-MS) to enable elemental detection. Cells are labeled with probes conjugated to distinct stable metal isotopes, which are ionized in a and analyzed based on their , providing quantitative data on expression without the optical limitations of fluorescence-based assays. This integration allows for precise, measurement of transient signals from individual cells passing through the instrument at rates up to thousands per second. The scope of CyTOF is centered on single-cell proteomics and phenotyping, supporting the simultaneous analysis of over 50 protein markers per cell using rare earth metal tags, such as lanthanides, to achieve high-dimensional resolution of cellular heterogeneity. This capability is particularly valuable in , , and , where it enables deep profiling of surface and intracellular targets without spectral overlap or compensation requirements. Notably, "CyTOF" specifically denotes the commercial instrument lineup, including models like the , XT, and XT PRO systems (as of 2024). Recent models, such as the CyTOF XT PRO introduced in 2024, further enhance automation and sample throughput for clinical applications.

Basic Principles

CyTOF, or by time-of-flight, relies on the conjugation of biological probes, such as antibodies, to stable isotopes of rare-earth elements for multiplexed detection of cellular targets. These isotopes, spanning the mass range of 139 to 176 units (amu), are attached to antibodies via chelating polymers, enabling specific binding to proteins or other markers on or within cells while minimizing natural background interference due to the rarity of these elements in biological samples. This metal-tagging approach supports simultaneous measurement of dozens of parameters per cell, far exceeding the limitations of traditional fluorescence-based . Labeled cells are aerosolized and injected into an inductively coupled , where they are rapidly vaporized and atomized at of approximately 6000–8000 K, producing a of singly charged ions proportional in number to the abundance of each metal tag. The high ensures complete of cellular material into free atoms, which are then ionized primarily to the +1 charge state, facilitating downstream mass analysis without molecular fragmentation complicating the signal. The cloud is extracted into a time-of-flight (TOF) mass spectrometer, where ions are accelerated by an of voltage V and separated based on their (m/z) according to their flight times over a fixed path length L. The flight time t for an is given by t = \sqrt{ \frac{m L^2}{2 z e V} } where m is the , z is the ion charge (typically 1 for atomic ions), and e is the ; lighter ions arrive at the detector faster than heavier ones, generating a time-resolved mass spectrum. In practice, the relationship is calibrated as t = t_0 + A \sqrt{m/z}, with constants t_0 and A determined empirically. CyTOF systems achieve a mass >500 m/z units (), with values exceeding 900 in standard operation, sufficient to resolve adjacent with minimal overlap, typically less than 1%. This high resolution ensures discrete detection channels for each isotope, eliminating the spectral spillover inherent in fluorophore-based , where overlapping emission spectra require compensation matrices and limit to around 20 parameters.

History

Early Development

The foundational research leading to cytometry by time-of-flight (CyTOF) began in 1994, when Tsutomu Nomizu and colleagues at explored the application of (ICP) atomic emission spectrometry to analyze biological particles, successfully determining calcium content in individual yeast and mammalian cells by introducing cell suspensions into the plasma for time-resolved measurements. This work established the feasibility of using plasma-based ionization for single-cell , overcoming challenges in sample introduction and detection sensitivity for microscopic biological entities. A major breakthrough occurred in 2007, when Scott Tanner's team at the demonstrated the potential of combining with ICP (ICP-TOF-MS) for , using metal isotopes conjugated to antibodies as labels to enable multiplexed detection without spectral overlap issues inherent in fluorescence methods. This innovation addressed key limitations in traditional cytometry by leveraging the high resolution and dynamic range of for simultaneous measurement of dozens of parameters per cell. The technique's viability was rigorously validated in a seminal 2009 publication by Dmitry R. Bandura and colleagues, including , along with Vladimir I. Baranov and Olga I. Ornatsky, in Analytical Chemistry, which described the prototype mass cytometer and demonstrated its capacity for up to 60-plex analysis, with potential for over 100 parameters through stable metal isotope tagging. Central challenges, such as vaporizing and ionizing cells in the ICP without fragmentation while preserving intact ion clouds from single cells, were resolved via optimized nebulization and plasma conditions that achieved detection efficiencies suitable for rare event analysis. Proof-of-concept experiments in this study utilized human cell lines labeled with lanthanide-tagged antibodies, enabling multiplexed detection of surface and intracellular markers to distinguish subtypes with high specificity and minimal background . These efforts marked the transition from conceptual elemental mapping to practical , setting the stage for CyTOF's broader adoption in cellular research.

Commercialization and System Iterations

The commercialization of cytometry by time-of-flight (CyTOF) began with DVS Sciences, a Toronto-based company founded in to develop technology, which unveiled the first CyTOF system in November 2009 as the inaugural commercial platform combining principles with for multiparametric . In February 2014, Fluidigm Corporation acquired DVS Sciences for approximately $207.5 million in cash and stock, integrating the CyTOF platform into its portfolio of tools and enabling broader market expansion for high-parameter protein profiling. Following the acquisition, Fluidigm introduced the system in June 2015 as an advanced CyTOF iteration, supporting up to 45 or more parameters per through expanded metal conjugation capabilities. The upgrade, often referred to as CyTOF 3.0 in subsequent installations starting around 2017, improved event processing for sustained acquisition rates of approximately 500 events per second, facilitating deeper phenotyping in research settings. Fluidigm continued iterating on the platform, releasing the CyTOF XT in May 2021 as a next-generation system with automated sample barcoding and capabilities for up to 50 samples per run, reducing manual handling and enhancing reproducibility in high-throughput studies. In April 2022, Fluidigm rebranded to Inc. following a $250 million strategic capital infusion from investors Casdin Capital and , aimed at accelerating innovation in and technologies. Standard BioTools launched the CyTOF XT Pro in March 2025, featuring enhanced sensitivity via optimized ion optics and quadrupled throughput at up to 2,000 events per second, alongside 21 CFR Part 11 compliance for regulated environments. Key technological iterations across CyTOF models are summarized below, focusing on parameter capacity and event acquisition rates, which have evolved to support increasingly complex single-cell analyses while maintaining .
ModelRelease YearMaximum ParametersSustained Event Rate (events/sec)
CyTOF 12009~20300–500
(CyTOF 3.0)2015/201745+500 (peak up to 2,000)
CyTOF XT202150+500
CyTOF XT Pro202550+2,000
These advancements reflect a progression from foundational single-cell detection to automated, high-volume processing, driven by commercial priorities for accessibility and scalability in biomedical research.

Instrumentation

Core Components

The core components of a CyTOF system form the foundation for by integrating sample introduction, plasma-based , ion manipulation, and detection within a high-vacuum environment. The process begins with the and , which aerosolize the cell suspension into fine droplets using a concentric glass driven by gas flow, enabling the introduction of 200-300 cells per second to ensure without clustering. Central to atomization is the , configured as an () source operating at 1-1.5 kW radio-frequency (RF) power from a 40 MHz , which sustains a high-temperature (approximately 5,000–10,000 K) to vaporize, atomize, and ionize the desolvated cells within its three concentric tubes. Ions generated in the atmospheric-pressure are then extracted through a multi-aperture vacuum interface into the mass analyzer region. Ion selection and separation occur via the quadrupole ion guide and time-of-flight (TOF) analyzer; the RF quadrupole acts as a to remove low-mass interferents (below m/z 80), directing the relevant metal ions toward the dual-stage orthogonal acceleration reflectron TOF analyzer, which sequences ion clouds at 76.8 kHz push for mass-to-charge resolution exceeding 500 (FWHM at m/z 159). The detector, a dynode electron multiplier, captures arriving ions and amplifies the signal for counting, processed through a high-speed digitizer to yield quantitative intensities per event. The entire flight path operates under a multi-stage , with turbo-molecular pumps maintaining pressures around 10^{-6} in the TOF analyzer to minimize scattering and ensure precise time-of-flight measurements. Overall flow proceeds sequentially from sample aerosolization in the , desolvation in a heated spray chamber, in the , extraction and filtering, TOF separation, and final detection, as illustrated in standard schematics of the architecture. This hardware configuration leverages ionization principles to enable metal-tagged detection without spectral overlap.

Technical Specifications

CyTOF systems support the simultaneous of up to 50 markers per , in addition to dedicated channels for viability assessment and sample duplexing, such as (191Ir/193Ir) DNA intercalators for nucleated identification and for viability assessment. The available detection channels span 135 distinct masses in the range of 75 to 209 units (amu), primarily utilizing isotopes conjugated to antibodies for multiplexed labeling without spectral overlap. Throughput in CyTOF instruments typically ranges from 300 to 500 per second in standard configurations, enabling the analysis of millions of per run; the advanced CyTOF XT Pro model achieves up to 2000 events per second through optimized sample uptake rates of 30 µL/min and higher concentrations. is defined by a of approximately 100 to 500 metal-tagged antibodies per , with signal-to-noise ratios exceeding 100:1 when using isotopes in the optimal of 153 to 176 amu. resolution, measured as (FWHM), is 300 to 600 at m/z ≈ 160, sufficient to resolve adjacent isotopes like ¹⁴³Nd from ¹⁴⁴Nd; in mode, spatial is at the single- level (≈10-15 µm). Instruments operate on a power supply of approximately 5 to 7 kW (via a 30 A, 220-240 V circuit) and feature a compact benchtop design with a footprint of about 1 m × 1 m and height up to 1.35 m. The following table compares key specifications across major CyTOF models:
ModelMaximum ParametersEvent Rate (cells/sec)Mass Range (amu)Footprint (width × height)
Up to 50250-50075-209≈1 m × 1.35 m
CyTOF XTUp to 50Up to 50075-2090.93 m × 1.35 m
CyTOF XT Pro50+500-200075-209≈0.93 m × 1.35 m

Workflow

Sample Preparation

Sample preparation for cytometry by time-of-flight (CyTOF) involves biological processing steps tailored to metal-tagged probes, enabling high-parameter analysis of single cells while minimizing spectral overlap inherent to fluorescence-based methods. This process typically begins with cell harvesting from sources such as cryopreserved peripheral blood mononuclear cells (PBMCs) or dissociated tissues, yielding 1–3 million viable cells per sample to ensure sufficient events for downstream analysis. Viability assessment is critical and is achieved through staining with agents like cisplatin, which binds to DNA in dead cells. DNA intercalators like iridium-based Cell-ID Intercalator-Ir (191Ir/193Ir) are used post-staining to label all nucleated cells for event gating during acquisition. Fixation follows, often using 1.6% formaldehyde or specialized stabilizers like SmartTube Proteomic Stabilizer for whole blood, to preserve cellular integrity and halt metabolic activity prior to antibody labeling. Antibody conjugation in CyTOF utilizes stable isotopes, primarily lanthanides from the series, covalently linked to monoclonal via polymer-based or direct methods to create probes like Maxpar antibodies from . Recent developments include the Maxpar Direct , offering pre-configured panels that bypass traditional for streamlined high-parameter . These metal tags, spanning such as 139La to 176Yb, are selected to avoid mass interference, with panel design software like Maxpar Panel Designer facilitating the assembly of 30–40 marker panels by optimizing combinations and curves. Conjugation protocols involve reducing antibody bonds with , followed by metal chelator attachment and purification, enabling customization for specific epitopes while maintaining signal intensity comparable to commercial reagents. Panels must account for isotopic abundance and potential overlaps, such as excluding 106Cd or 110Cd when using barcoding. To enable multiplexing, samples are barcoded with unique combinations of metal isotopes prior to pooling, allowing up to 50 samples (or more with custom kits) to be stained and analyzed in a single run, which reduces technical variability and reagent costs. Common approaches include the Cell-ID 20-Plex Barcoding Kit, which employs isotopes like 102Pd to 110Pd for labeling fixed cells, or live-cell barcoding kits supporting over 35 samples while preserving sensitive epitopes. Barcoding is performed post-fixation by incubating cells with distinct metal codes, followed by washing to remove excess labels. The staining protocol commences with resuspending fixed, barcoded cells in Maxpar Cell Staining Buffer, followed by addition of a metal-conjugated antibody cocktail and incubation for 1 hour at 4°C to promote specific binding to surface or intracellular targets. Intracellular staining may require additional permeabilization steps, but surface protocols emphasize room-temperature or low-temperature incubations to optimize antibody affinity. Washing steps, typically involving centrifugation at 800 × g and resuspension in staining buffer, remove unbound probes three to four times to minimize background noise. DNA intercalators, such as 191Ir-based reagents added at 125 nM for 1 hour or overnight at 2–8°C, are incorporated post-staining to label nucleated cells and facilitate event normalization. Quality control measures ensure sample integrity throughout preparation, including cell counting via aliquots post-fixation and verification of staining efficiency through bead-based normalization using EQ Four Element Calibration Beads (for CyTOF2/) or EQ Six Element Calibration Beads (for CyTOF XT), which are spiked into samples to correct for instrumental drift during . These beads, containing known metal intensities, enable consistent signal across runs, with software integration confirming nucleated event gating via iridium channels. Prepared samples are then ready for introduction into the CyTOF instrument for ionization and detection.

Data Acquisition

In CyTOF data acquisition, prepared single-cell suspensions are introduced into the system via nebulization, where the liquid sample is aerosolized into fine droplets, ideally containing one per droplet, and mixed with gas before entering the (ICP) torch. The plasma, operating at temperatures around 6000–10000 K, atomizes and ionizes the s and their metal-tagged antibodies, producing a transient cloud for each . Event detection and triggering occur based on the DNA intercalation signals from 191Ir and 193Ir isotopes, which provide a strong, correlating dual-signal threshold to distinguish intact s from or , ensuring only relevant cellular events are processed. The resulting ion cloud from each cell is extracted through ion optics, where a high-pass filter removes low-mass ions (typically below 75–120 ) to focus on the metal signals of interest. This filtered cloud is then pulsed into the time-of-flight (TOF) mass analyzer at , with ions separated by their and detected across multiple channels corresponding to the panel markers. For moderately expressed markers, the system detects on the order of 100–500 ions per channel, reflecting the low transmission efficiency (approximately 1 in 10^4 to 10^5 ions) but enabling quantitative measurement of antibody-bound metals. Acquisition operates in dual-counting mode, which combines analog and detection to extend and accurately quantify low-abundance signals down to near-single-molecule levels. Typical run parameters include event rates of 200–500 cells per second, allowing collection of up to 1–2 million events per hour depending on the instrument model. To maintain signal stability and correct for instrumental drift, are spiked into the sample at a of approximately 1 every 100–200 cellular events. These s, such as EQ Four Element (for CyTOF2/: e.g., 140Ce, 151Eu, 165Ho, 175Lu) or EQ Six Element (for CyTOF XT: e.g., 89Y, 115In, 140Ce, 159Tb, 175Lu, 209Bi), serve as internal standards for real-time and post-acquisition drift compensation, ensuring consistent quantification across the run. The pulse processing unit (PPU) manages the high transient rates, handling up to 300 kHz of pulses to support efficient data capture without overload. Raw output is generated as .fcs files (FCS 3.1 format), containing per-event data including event ID, acquisition time, and integrated intensity (counts) for each , ready for downstream .

Data Analysis

Preprocessing and Formats

Raw CyTOF data is acquired and exported in the standard (FCS) 3.1 file format by the CyTOF software, ensuring compatibility with analysis tools such as FlowJo. Earlier instrument versions generated intermediate IMD files, which are converted to FCS format using the CyTOF software for downstream processing. These FCS files encapsulate event data, including ion counts across up to 50 channels, supporting datasets with 10^5 to 10^6 events per sample acquisition. Normalization corrects for signal drift inherent in mass cytometry due to instrument variability over time. Bead-based normalization, introduced by Finck et al., uses internal calibration beads (e.g., EQ four-element beads) measured concurrently with cells to compute scaling factors based on bead intensities, stabilizing signals across files. This method models drift by comparing observed bead signals to a reference "passport" standard, applying multiplicative corrections to cell events. Following normalization, an arcsinh transformation is applied for variance stabilization, approximating logarithmic scaling for high-intensity values while remaining linear near zero to preserve low-signal information. A common cofactor of 5 is used in this transformation to enhance comparability with traditional data. Gating and filtering remove artifacts to isolate viable single cells. Doublets are excluded by plotting the area against height (or width) of the 191Ir DNA intercalator signal, identifying aggregates as events with disproportionate area-to-height ratios. Debris is removed by gating on the ratio of DNA intercalator (e.g., 191Ir or 193Ir) to EQ bead marker (140Ce) , discarding events that are DNA-negative and bead-negative as non-cellular fragments. These steps, often performed in software like FlowJo, ensure high-quality cell populations for analysis. Background subtraction addresses channel-specific noise, primarily from electronic offsets and isotopic contributions. Noise is modeled per channel using the poisson-limited nature of ion detection, where variance equals the mean count in low-signal regimes. Dual-counting techniques in the detector mitigate dead-time effects for poisson-distributed signals, subtracting baseline estimated from bead-free regions or control channels. Compensation for isotopic impurities, typically <1% in purified metal tags, is applied via matrix inversion to correct minor spillover between adjacent masses. Dedicated software facilitates these preprocessing steps. The CyTOF software from (formerly Fluidigm) handles initial file generation, normalization, and basic gating during acquisition. For advanced preprocessing, the open-source premessa enables panel editing, bead-based normalization, debarcoding, and FCS file manipulation, promoting reproducible workflows.

Visualization and Interpretation

Visualization and interpretation of CyTOF data involve applying high-dimensional analytical techniques to preprocessed datasets, enabling researchers to explore cellular heterogeneity and derive biological insights from measurements of 40 or more parameters per cell. These methods address the challenges of visualizing complex, non-linear manifolds in mass cytometry data, where traditional bivariate gating falls short, by projecting cells into lower-dimensional spaces while preserving structural relationships. Such approaches facilitate the identification of rare cell populations and subtle phenotypic differences that would otherwise be obscured in high-dimensional space. Dimensionality reduction techniques are foundational for embedding CyTOF data into 2D or 3D visualizations, allowing intuitive exploration of multiparametric relationships. (t-SNE), adapted for cytometry as viSNE, maps high-dimensional single-cell data onto two dimensions by minimizing divergences between high- and low-dimensional probability distributions, with common parameters including perplexity set to 30 to balance local and global structure preservation. Uniform manifold approximation and projection (UMAP) offers a faster alternative, constructing a fuzzy topological representation of the data manifold and optimizing it in low dimensions, often outperforming t-SNE in runtime and consistency for CyTOF datasets while maintaining both local clustering and global topology. These methods handle the non-linear manifolds inherent in CyTOF data, enabling validation against manual subsets for accuracy. Unsupervised clustering algorithms further aid in phenotyping by partitioning cells into discrete subsets based on marker expression patterns. FlowSOM employs self-organizing maps to generate a hierarchical metaclustering structure, rapidly identifying 30 or more cell subsets in high-dimensional CyTOF data through a two-level process that first creates a of clusters and then refines them. Similarly, spanning-tree progression analysis of density-normalized events () downsamples and clusters cells by density, constructing a to visualize hierarchical relationships and reveal functional marker variations across populations. These tools excel in unsupervised discovery, often uncovering novel subsets that align with known immunological categories. Statistical analyses build on these visualizations to quantify differences and enable testing. viSNE supports gating in the , allowing iterative refinement of populations while leveraging the full parameter set for precise subset isolation. Cluster identification, characterization, and (Citrus) performs supervised analysis by hierarchically clustering cells and regressing cluster abundances against experimental covariates, identifying differentially abundant subsets without predefined gates. For interpretation, heatmaps display median marker expression across clusters, highlighting phenotypic distinctions in a compact format, while methods like reconstruct developmental progressions by modeling cells along a pseudotime axis based on distances in high-dimensional . These visualizations provide quantitative context, such as relative expression levels, to guide biological inference. Popular software platforms integrate these methods for seamless workflows. Cytobank, a cloud-based tool, supports viSNE, FlowSOM, , and natively, facilitating collaborative analysis of large CyTOF datasets. FlowJo incorporates a viSNE alongside t-SNE and UMAP capabilities, enabling embedding and clustering directly within its gating for user-friendly exploration. Recent open-source tools, such as ImmCellTyper (as of 2024), provide semi-supervised clustering and cell type annotation for CyTOF data analysis.

Applications

Immunology

Mass cytometry, or CyTOF, has revolutionized immune cell phenotyping by enabling the simultaneous measurement of over 40 markers per cell, allowing for the detailed mapping of more than 30 immune subsets in peripheral blood mononuclear cells (PBMCs). This high-dimensional approach surpasses traditional in resolving complex cellular heterogeneity without spectral overlap, facilitating unbiased identification of cell types based on surface markers and functional attributes. For instance, in T-cell phenotyping, CyTOF panels incorporating markers such as CD3, , , and intracellular cytokines have delineated at least 14 distinct T-cell subsets, including naive, memory, effector, and cytokine-producing populations, providing insights into adaptive immune dynamics. In applications to innate immunity, CyTOF has elucidated natural killer (NK) cell heterogeneity, particularly in the context of cancer, where it reveals functional diversity among NK subsets based on receptors like NKG2A, NKp46, and CD16. A seminal study by Newell et al. (2012) utilized CyTOF to identify 10 distinct NK cell subsets through combinatorial analysis of surface markers and cytokine expression, highlighting niche-specific behaviors without reliance on fluorochromes. Similarly, in adaptive immunity during chronic infections, CyTOF profiling of exhaustion markers such as PD-1 and TIM-3 on CD8+ T cells has uncovered progressive functional impairment, with co-expression correlating to reduced cytokine production and proliferative capacity. These markers define exhausted subsets that emerge in sustained antigenic environments, aiding in the dissection of T-cell dysfunction. CyTOF also supports tracking B-cell maturation in vaccine responses, where it monitors transitions from naive to plasmablast and memory B-cell stages post-immunization, using markers like , CD27, , and IgD. Studies have shown that vaccine-induced shifts in B-cell subsets, such as increased class-switched memory cells, correlate with antibody titers and long-term immunity. A key advantage of CyTOF in is its sensitivity for detecting rare populations, such as antigen-specific cells comprising less than 0.1% of total PBMCs, without requiring prior hypotheses or enrichment steps, thus enabling discovery of low-frequency effectors in immune responses.

Oncology and Beyond

CyTOF has significantly advanced the understanding of tumor-immune interactions in by enabling high-dimensional profiling of immune s within the (TME). For instance, in , has revealed the immunosuppressive role of myeloid-derived suppressor s (MDSCs), which accumulate in the TME and inhibit antitumor immunity through mechanisms such as arginase-1 expression and T- suppression. Using , researchers identified distinct MDSC subsets, including granulocytic and monocytic types, that correlate with poor and therapy resistance in human breast tumors, highlighting their heterogeneity and functional states across patient samples. A seminal study by Chevrier et al. in 2017 utilized CyTOF to create an immune atlas of clear cell renal cell carcinoma (ccRCC), analyzing over 70 patient samples with a 40-parameter panel to map immune cell infiltration and organization. This work uncovered the presence of tertiary lymphoid structures (TLS) in the TME, which are ectopic lymphoid aggregates associated with improved survival and responsiveness to immunotherapy, as they facilitate local T- and B-cell priming against tumor antigens. The analysis demonstrated that TLS maturation stages, marked by follicular dendritic cells and high endothelial venules, vary across tumor grades and predict clinical outcomes, providing a framework for TLS as prognostic biomarkers in renal cancer. Beyond oncology, CyTOF has been instrumental in mapping hematopoiesis, offering single-cell resolution of progenitor differentiation in bone marrow. High-dimensional proteo-genomic maps generated via CyTOF have quantified over 30 surface and intracellular markers to delineate hematopoietic stem cell (HSC) subsets, revealing rare populations like long-term HSCs and their transitions during steady-state and stress conditions, which informs models of blood cell production disorders. In neurology, CyTOF profiling of brain tissue has elucidated neuronal diversity by simultaneously assessing protein markers on neurons, glia, and immune cells in mouse models. A developmental atlas of the mouse brain, constructed using a 40-parameter CyTOF panel, identified over 50 neuronal subtypes based on neurotransmitter receptors and transcription factors, tracking their spatiotemporal emergence from embryonic to adult stages and linking phenotypic changes to circuit formation. For microbiology, CyTOF enables phenotyping of microbial communities at single-cell resolution, particularly in host-pathogen interactions. By conjugating metal isotopes to bacterial-specific antibodies, researchers have quantified heterogeneity in bacterial populations during infections, such as silver nanoparticle uptake in Escherichia coli, distinguishing viable from stressed cells and mapping community dynamics in biofilms without fluorescence interference. Emerging applications of CyTOF extend to infectious and autoimmune diseases, capturing dynamic immune responses. In , whole-blood CyTOF workflows have defined circulating immune signatures, identifying expanded activated T-cell subsets and dysregulated monocytes in severe cases, with recovery trajectories marked by restored naive T-cell frequencies and reduced -producing effectors. For autoimmune profiling, CyTOF has delineated B- and T-cell subsets in systemic lupus erythematosus (SLE), revealing expanded age-associated B cells (CD21low CD11c+) and proliferating + helper T cells in early disease, which correlate with risk and production. At the 2024 CyTOF Summit, studies showcased 50-parameter panels to uncover distinct T-cell functional signatures in responders, emphasizing intracellular profiles like IFN-γ and IL-2 in exhausted versus reinvigorated + T cells across solid tumors.

Variants and Advances

Imaging Mass Cytometry

Imaging (IMC) represents a spatial extension of technology, enabling the simultaneous detection and visualization of up to 40 or more protein markers at subcellular within intact sections. Unlike traditional suspension-based CyTOF, IMC employs a 213 nm (UV) to ablate discrete 1 μm² pixels from formalin-fixed, paraffin-embedded (FFPE) or slides mounted on indium-tin oxide-coated glass. This ablation process vaporizes and ionizes the biological material, creating transient plumes that are transported to the time-of-flight mass spectrometer (ICP-TOF-MS) for quantitative analysis of metal isotopes conjugated to antibodies, thereby generating high-dimensional images without the spectral overlap or autofluorescence issues inherent to fluorescence-based methods. The workflow for IMC begins with standard tissue preparation, including sectioning to 4-5 μm thickness, followed by multiplexed with metal-tagged antibodies specific to cellular targets. The stained slide is then raster-scanned by the UV in the Hyperion Imaging , operating at rates of approximately 200 pixels per second, which ensures minimal cross-contamination (<2%) between adjacent pixels while covering regions of interest up to several square millimeters. This process yields raw data in the form of maps for each marker, allowing reconstruction of spatial protein distributions at 1 μm across entire slides. The Hyperion supports whole-slide , facilitating the analysis of complex architectures like tumor microenvironments with 40+ concurrent markers. In June 2025, launched new panels for and , expanding applications in spatial . Data analysis in IMC involves several key steps to extract biologically meaningful insights from the multiplexed s. Initial preprocessing includes to correct for minor misalignments across channels and to account for efficiency variations. segmentation is typically performed using algorithms such as watershed transformation, which leverages and markers to delineate individual cells and subcellular compartments, often enhanced by Gaussian blurring to reduce noise. Subsequent neighborhood analysis quantifies spatial interactions, such as cell-cell contacts or proximity metrics, revealing phenotypic heterogeneity and functional relationships within tissues. These computational approaches enable phenotyping of thousands of cells per while preserving spatial context. A pivotal advancement in IMC occurred in 2023 with the launch of the Hyperion XTi Imaging System, which introduces automated, high-throughput capabilities by processing up to 40 slides per day—five times faster than prior models—while maintaining superior limits of detection and subcellular resolution. This system streamlines batch staining and acquisition, accelerating applications in spatial biology. In October 2025, high-resolution IMC (HR-IMC) was introduced, achieving enhanced subcellular mapping of structures with improved resolution capabilities. IMC's ability to deliver fluorescence-like multiplexed imaging without optical limitations has made it a in initiatives like the Human Tumor Atlas Network, where it maps intricate tumor ecosystems and immune interactions in patient-derived samples.

High-Throughput Developments

In March 2025, launched the CyTOF XT PRO system, a fifth-generation mass cytometer designed to enhance throughput and support applications. This platform achieves up to four times faster acquisition speeds compared to prior models, enabling high-dimensional immune profiling with improved sensitivity and repeatability. The system supports simultaneous analysis of over 50 and incorporates automated sample handling to facilitate large-scale studies, such as processing up to 100 samples in automated runs, which streamlines workflows for monitoring and biomarker discovery. Advancements in software have complemented these hardware improvements, with tools like Maxpar Pathsetter providing streamlined and for CyTOF-generated datasets. Presented at the AACR Annual Meeting 2025, updates to the CyTOF ecosystem emphasized integration with compliant protocols under 21 CFR Part 11, aiding regulatory submissions in clinical trials. These developments reduce overall run times from hours to under an hour for high-parameter panels, enabling the analysis of millions to billions of cells across population-scale cohorts. The 2024 CyTOF Summit, hosted by , highlighted emerging applications of these high-throughput capabilities, including multi-omics integration for predictive modeling. Discussions focused on AI-assisted to automate gating and phenotyping, accelerating insights into complex immune dynamics. The 2025 CyTOF Summit in , , further showcased the latest advancements in CyTOF and Imaging Mass Cytometry. Contract research organizations (CROs) have increasingly adopted standardized CyTOF protocols for clinical trials, with providers like Caprion Biosciences offering services tailored to immuno-oncology endpoints, such as functional immune profiling. This integration supports reproducible, high-volume data generation essential for advancing personalized therapies.

Advantages and Limitations

Advantages

One of the primary advantages of (CyTOF), also known as , is its high multiplexing capability, allowing simultaneous measurement of over 50 parameters per cell without the need for spectral compensation, in contrast to conventional , which is typically limited to 10-20 parameters due to fluorophore overlap. This enables comprehensive phenotyping of complex cellular heterogeneity in a single sample, facilitating the identification of rare cell populations and intricate signaling networks that would require multiple panels in fluorescence-based methods. CyTOF also provides low , as it eliminates autofluorescence inherent in biological samples and minimizes signal spillover between channels, thanks to the distinct masses of metal tags resolved by . Furthermore, metal-tagged samples maintain signal stability for up to two weeks post-staining, offering greater flexibility in experimental workflows compared to fluorescent labels, which degrade more rapidly. At the single-cell level, CyTOF delivers quantitative measurements with high , supporting absolute cell counts through with reference standards or beads, and is compatible with of stained samples, preserving viability and marker expression for downstream analysis. This precision arises from the uniform incorporation of metal tags, resulting in low coefficients of variation (typically <10% intra-assay), enhanced by barcoding techniques that pool multiple samples to reduce batch effects and improve reproducibility across experiments. These strengths have enabled landmark discoveries in , such as the delineation of over 100 distinct immune cell subsets and novel functional states in human peripheral blood, revealing previously unrecognized heterogeneity in immune responses.

Limitations

Despite its capabilities, by time-of-flight (CyTOF) faces significant practical limitations that can constrain its broader adoption in research and clinical settings. One primary challenge is its relatively low throughput compared to traditional fluorescence-based . Current CyTOF systems achieve acquisition rates of up to 2,000 cells per second (e.g., with the CyTOF XT PRO), in contrast to over 10,000 cells per second possible with conventional flow cytometers, which can restrict experiments to approximately 10^7–10^8 cells per run depending on the system, duration, and setup. Another inherent drawback is the destructive nature of the analysis process, where cells are vaporized by the during ionization, preventing any downstream sorting, recovery, or further manipulation of the analyzed sample. This one-time-use aspect limits CyTOF to applications where archival or iterative analysis of the same cells is not required. CyTOF also incurs high operational costs, with instruments priced at over $500,000 and metal-conjugated costing around $300–$400 per vial, further compounded by potential vulnerabilities for rare earth metals used in tagging. Additionally, the risk of spectral contamination arises from isobaric interferences—such as or argide formations—if antibody panels are not meticulously designed to avoid overlapping masses, which can lead to inaccurate signal attribution and reduced data quality. While 2025 updates, such as the CyTOF XT PRO system, have introduced improvements like up to fourfold higher throughput through enhanced automation, CyTOF remains approximately 10 times slower than advanced systems.

References

  1. [1]
    An introduction to mass cytometry: fundamentals and applications
    Mass cytometry is a novel adaptation of atomic mass spectrometry that enables uniquely high-dimensional flow cytometry applications.
  2. [2]
    CyTOF® for the Masses - Frontiers
    A comprehensive resource guide. Included in this review are experiment and panel design, antibody conjugations, sample staining, sample acquisition, and data ...Introduction · Experimental Design · Panel Design & Antibody... · Sample Staining
  3. [3]
    Mass Cytometry - an overview | ScienceDirect Topics
    Mass cytometry (or CyTOF for cytometry by TOF) is a hybrid technology combining principles of flow cytometry with inductively coupled plasma mass spectrometry.
  4. [4]
  5. [5]
    CYTOF Trademark | Trademarkia
    CYTOF is a registered trademark (Registration #3441318) owned by STANDARD BIOTOOLS CANADA INC., a MARKHAM, ONTARIO based entity . The trademark was filed on ...
  6. [6]
    Fluidigm Completes Acquisition Of DVS Science
    Feb 13, 2014 · DVS's principal product is the CyTOF® 2 Mass Cytometer, which analyzes antibody/metal complexes using atomic mass spectrometry. The technology ...
  7. [7]
    [PDF] Deep Immune Profiling with the Maxpar Direct Immune Profiling ...
    Mass cytometry, which utilizes CyTOF® technology, is a single-cell analysis platform that uses metal-tagged antibodies to resolve over 50 markers in a single ...
  8. [8]
    CyTOF XT - Standard BioTools
    Mass cytometry, powered by CyTOF technology, is the world's most advanced cytometric platform. It has opened new doors to discovery and clinical research by ...Missing: DVS | Show results with:DVS
  9. [9]
    [PDF] Helios, a CyTOF System - User Guide
    The relationship between time of ion flight to the detector and their mass-to-charge ratio (m/z) is: in which t0 and A are derived from the mass calibration ...
  10. [10]
    None
    ### Summary of Flight Time Equation for TOF MS
  11. [11]
    Determination of Calcium Content in Individual Biological Cells by ...
    Time-resolved particle image velocimetry and 3D simulations of single particles in the new conical ICP torch. Journal of Analytical Atomic Spectrometry 2019 ...Missing: early concept
  12. [12]
    DVS Sciences Unveils First-in-Class Mass Cytometer for Use in ...
    Nov 26, 2009 · The CyTOF™ Mass Cytometer addresses the challenges of flow cytometry using inductively coupled plasma mass spectrometry, thereby combining two ...<|control11|><|separator|>
  13. [13]
    Mass Cytometry - an overview | ScienceDirect Topics
    ... DVS Sciences, Inc. Cytometry by time of flight (CyTOF) was initially commercialized by DVS Sciences in 2009. In 2014, Fluidigm acquired DVS Sciences to ...
  14. [14]
    Fluidigm Introduces Advanced Mass Cytometry System - The Scientist
    Jun 25, 2015 · Fluidigm Corporation (NASDAQ:FLDM) today introduced HeliosTM, a CyTOF® platform that enables system-level biology at single-cell resolution.
  15. [15]
    Instrumentation | Mass Cytometry Center of Excellence
    The Helios CyTOF 3.0 instrument arrived in October 2017, replacing Vanderbilt's original CyTOF 1 (#17 globally!) Center labs have trained operators that can ...
  16. [16]
    Next-Generation CyTOF XT Redefines Cytometry with Advances in ...
    May 25, 2021 · This new CyTOF platform offers significant improvements in throughput, automation, time to results and total cost of ownership.
  17. [17]
    Fluidigm Completes $250 Million Strategic Capital Infusion and ...
    Apr 4, 2022 · Fluidigm has been renamed Standard BioTools Inc. (Standard BioTools) and its common stock is expected to begin trading on Nasdaq under the symbol “LAB.”
  18. [18]
    CyTOF XT PRO System Launches with Innovations for Clinical Trial ...
    The CyTOF XT PRO offers wide immune coverage, improved throughput, 21 CFR Part 11 compliance, advanced barcoding, and high reproducibility.
  19. [19]
    [PDF] CyTOF XT PRO
    The CyTOF XT PRO system can resolve multiple elemental probes at high acquisition rates, maximizing the per-cell information obtained from a single sample. The ...
  20. [20]
    [PDF] CyTOF XT - ALAB
    Flow Rate (L/min). CyTOF XT instrument. 85 ±5 psi (100 psi max.) 15. Exhaust. Hose Diameter. mm (in). Flow Rate cfm(L/sec) Anemometer m/sec (ft/min). Torch Box.
  21. [21]
    The anatomy of single cell mass cytometry data - Wiley Online Library
    Oct 2, 2018 · For CyTOF, the rate is often closer to 200–300 cells per second, which means that the instrument will need to run for longer to acquire a ...
  22. [22]
    CyTOF® for the Masses - PMC - NIH
    Introduction. Mass cytometry, also termed cytometry by Time-Of-Flight (CyTOF®), is a powerful tool for high-dimensional and high-throughput single-cell assays. ...
  23. [23]
    [PDF] Rapid and Reproducible Results for Accelerated Insights on the ...
    The improvements in injector design and event processing have enabled higher event rates without compromising data integrity, making the CyTOF XT. PRO system ...
  24. [24]
    Immune monitoring technology primer: flow and mass cytometry
    In general, the best fluorochromes have a detection limit of about 40 molecules per cell, while the detection limit for mass cytometry is about 400–500 ...
  25. [25]
    CyTOForum • View topic - Sensitivity of Mass cytometry
    Aug 3, 2017 · I think the limit of detection is supposed to be on the order of ~400 molecules per cell. In your case, the cytokines you are detecting look ...Extending the axis for CyTOF dataTuning sensitivity and data qualityMore results from cytoforum.stanford.edu
  26. [26]
    Mass cytometry: technique for real time single cell ... - PubMed
    A novel instrument for real time analysis of individual biological cells or other microparticles is described.Missing: original paper
  27. [27]
    [PDF] Maxpar Cell Surface Staining with Fresh Fix Protocol (400276 Rev 07)
    IMPORTANT Run cells on the same day they are washed from intercalation solution. Stained samples can be acquired on CyTOF 2, Helios (HT or WB Injector), or ...
  28. [28]
  29. [29]
    Metal-isotope-tagged monoclonal antibodies for high-dimensional ...
    To date, 48 isotopes coming from 18 heavy metals have been conjugated to monoclonal IgG antibodies and are widely used in single-cell CyTOF immunoassays. The ...
  30. [30]
    Sample Multiplexing - Standard BioTools
    Cell barcoding is a powerful method for multiplexing samples with mass cytometry (CyTOF technology). By staining and acquiring samples in a single tube,
  31. [31]
    Guidelines for the use of flow cytometry and cell sorting in ...
    From there, a yield sort was carried out, with a flow rate of 4 mL/h. The resulting cell processing rate was 110 000 total cells per second. With a target ...
  32. [32]
    Normalization of mass cytometry data with bead standards - NIH
    Mar 19, 2013 · The protocol described here includes simultaneous measurements of beads and cells on the mass cytometer, subsequent extraction of the bead-based signature,
  33. [33]
    [PDF] CyTOF XT (FLDM-00461)
    CyTOF XT has a 0.7 m2 footprint, 135 mass channels (75-209 amu), 12 5mL and 1 15mL tube capacity, and uses 15 L/min argon.Missing: power | Show results with:power
  34. [34]
    Choosing File Types
    Jul 21, 2021 · CyTOF Software v8.0 generates flow cytometry standard (FCS 3.1) files. Events FCS files contain events that meet the event detection criteria specified in the ...
  35. [35]
  36. [36]
    Normalization of mass cytometry data with bead standards - PubMed
    The protocol described here includes simultaneous measurements of beads and cells on the mass cytometer, subsequent extraction of the bead-based signature.
  37. [37]
    transformData: Transform data in diffcyt - rdrr.io
    The arcsinh transform is widely used for CyTOF data. It behaves similarly to a log transform at high values, but is approximately linear near zero; so unlike ...
  38. [38]
    CyTOF workflow: differential discovery in high-throughput cytometry
    Conventional flow cytometers and mass cytometers produce .fcs files that can be manually analyzed using programs such as FlowJo [TriStar] or Cytobank (Kotecha, ...
  39. [39]
    Automated Data Cleanup for Mass Cytometry - Wiley Online Library
    Nov 18, 2019 · Since the iridium intercalator contains both 191Ir and 193Ir, users typically couple the two for their cleanup selection strategies. Care must ...
  40. [40]
    Data scientist's primer to analysis of mass cytometry data
    Because CyTOF instrument performance may vary over time (both within a single run, but more prominently between different runs), it is important to normalize ...Missing: rates | Show results with:rates
  41. [41]
    Validation of CyTOF Against Flow Cytometry for Immunological ...
    May 17, 2019 · We developed a 40+ parameter CyTOF panel and demonstrate that compared to flow cytometry, CyTOF yields analogous quantification of cell lineages ...Cytof And Flow Cytometry... · Results · Validating Cytof In Tumor...
  42. [42]
    Comments on “Determination of Background, Signal‐to‐Noise, and ...
    Dec 10, 2019 · The model implied in Figure 1B represents a typical noise distribution. ... For low count numbers, this will be a Poisson distribution and at ...
  43. [43]
    Determination of background, signal‐to‐noise, and dynamic range ...
    Sep 27, 2017 · The first parameter is the signal-to-noise ratio (SNR) which is a measure that compares the level of a desired signal to the level of a ...<|separator|>
  44. [44]
    [PDF] A deep profiler's guide to cytometry
    Mass cytometry is a new technology that promises to extend these capa- bilities significantly. Immunophenotyping by mass spec- trometry provides the ability to ...
  45. [45]
    Software - Standard BioTools
    CyTOF Software v9.3 ... CyTOF™ Software v9.3 offers instrument control and data acquisition functionality for both imaging and flow applications with the Hyperion ...
  46. [46]
    ParkerICI/premessa: R package for pre-processing of mass ... - GitHub
    premessa is an R package for pre-processing of flow and mass cytometry data, that includes panel editing/renaming for FCS files, bead-based normalization and ...
  47. [47]
    viSNE enables visualization of high dimensional single-cell data ...
    May 19, 2013 · Here we present viSNE, a tool that allows one to map high-dimensional cytometry data onto two dimensions, yet conserve the high-dimensional structure of the ...
  48. [48]
    Comparative analysis of dimension reduction methods for cytometry ...
    Apr 1, 2023 · CyTOF data are notably different from scRNA-seq data in many aspects. This calls for the evaluation and development of computational methods ...
  49. [49]
    FlowSOM: Using self‐organizing maps for visualization and ...
    Jan 8, 2015 · In this article, we introduce a new visualization technique, called FlowSOM, which analyzes Flow or mass cytometry data using a Self-Organizing Map.Missing: Bioinformatics | Show results with:Bioinformatics
  50. [50]
    Extracting a cellular hierarchy from high-dimensional cytometry data ...
    Oct 2, 2011 · We present a versatile computational approach, spanning-tree progression analysis of density-normalized events (SPADE).
  51. [51]
    Comparison of clustering methods for high‐dimensional single‐cell ...
    Dec 19, 2016 · Modern flow cytometers can routinely detect 15–20 parameters (protein markers) per cell 1, 2, at throughput rates above 10,000 cells per second.Clustering Methods · Results · Discussion
  52. [52]
    Automated identification of stratifying signatures in cellular ... - PNAS
    Jun 16, 2014 · We present a data-driven method termed Citrus that identifies cell subsets associated with an experimental endpoint of interest.Results · Summary Of Citrus · Evaluation Of Citrus Vs...
  53. [53]
    Cytobank Premium - Beckman Coulter
    The cloud-based Cytobank data analysis platform is an ideal solution for immunologists and cytometrists who need to quickly gain insights from large datasets.Missing: CyTOF | Show results with:CyTOF
  54. [54]
    tSNE - FlowJo v10 Documentation
    tSNE is an algorithm for dimensionality reduction, visualizing complex data in fewer dimensions while maintaining its structure, useful for high dimensional ...Missing: viSNE | Show results with:viSNE
  55. [55]
    CyTOF Measurement of Immunocompetence across Major Immune ...
    Using mass cytometry (CyTOF), we can simultaneously detect ~40 markers to identify various cell subsets and determine their function.
  56. [56]
    CyTOF mass cytometry analysis of human memory CD4+ T cells and ...
    Jun 17, 2022 · Here, we describe a mass-cytometry-based protocol to examine memory CD4+ T cell and memory B cell (MBC) responses in human peripheral blood.
  57. [57]
    Mass cytometry-based peripheral blood analysis as a novel tool for ...
    Our study proved that the alterations of peripheral immune cell subsets could assist tumour detection, and provide a ready-to-use detection model for HCC and ...
  58. [58]
    Cytometry by time-of-flight shows combinatorial cytokine expression ...
    Jan 27, 2012 · Cytometry by time-of-flight shows combinatorial cytokine expression and virus-specific cell niches within a continuum of CD8+ T cell phenotypes.
  59. [59]
    Use of Mass Cytometry to Profile Human T Cell Exhaustion - PMC
    Jan 22, 2020 · In this review, we discuss how mass cytometry can be used to reveal the role of T EX subsets in humans by combining exhaustion-directed phenotyping with ...
  60. [60]
    Application and utility of mass cytometry in vaccine development
    Nov 1, 2017 · With mass cytometry, we have been able to simultaneously interrogate samples for broad changes in T-cell, B-cell, dendritic cell, granulocyte, ...
  61. [61]
    Predicting Cell Populations in Single Cell Mass Cytometry Data - PMC
    Furthermore, results on the BMMC dataset show that LDA can detect rare cell populations having frequencies <0.5% of the total number of cells, like MPP, HSC ...
  62. [62]
    An Immune Atlas of Clear Cell Renal Cell Carcinoma - PubMed - NIH
    We used mass cytometry with extensive antibody panels to perform in-depth immune profiling of samples from 73 clear cell renal cell carcinoma (ccRCC) patients ...Missing: tertiary lymphoid structures
  63. [63]
    Single-cell proteo-genomic reference maps of the hematopoietic ...
    Nov 22, 2021 · Here, we have generated high-content single-cell proteo-genomic reference maps of human blood and bone marrow that quantitatively link the ...
  64. [64]
    A streamlined whole blood CyTOF workflow defines a circulating ...
    Jan 26, 2021 · Mass cytometry (CyTOF) represents one of the most powerful tools in immune phenotyping, allowing high throughput quantification of over 40 ...<|separator|>
  65. [65]
    Longitudinal immune cell profiling in early systemic lupus ... - NIH
    Nov 1, 2023 · Two major helper T cell subsets and unique Ki67 + proliferating immune cell subsets were expanded in the early phase of SLE, and the immunologic features ...
  66. [66]
    50-Parameter CyTOF Intracellular Cytokine Panel Reveals Distinct ...
    Oct 15, 2025 · Title: "50-Parameter CyTOF Intracellular Cytokine Panel Reveals Unprecedentedly Distinct Functional Signatures of T Cells" Speaker: Anna ...
  67. [67]
    Imaging Mass Cytometry - Chang - 2017 - Wiley Online Library
    Feb 3, 2017 · Imaging Mass Cytometry (IMC) is an expansion of mass cytometry, but ... laser operating at 213 nm and focused to a 1 µm diameter spot size.
  68. [68]
    Hyperion XTi Imaging System - Standard BioTools
    Imaging Mass Cytometry technology enables 40-plus markers that can be simultaneously stained, acquired and visualized. Other fluorescence-based approaches ...
  69. [69]
    Different approaches to Imaging Mass Cytometry data analysis
    Apr 3, 2023 · This review aims to give a systematic synopsis of all the available classical image analysis tools and pipelines useful to be employed for IMC data analysis.
  70. [70]
    Standard BioTools introduces the Hyperion XTi Imaging System ...
    Apr 17, 2023 · With Hyperion™ XTi, researchers can now image up to 40 slides per day, processing larger numbers of samples in shorter periods of time. Powered ...
  71. [71]
    The Human Tumor Atlas Network: Charting Tumor Transitions ... - NIH
    Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods 11, 417–422. [DOI] [PubMed] [Google Scholar]; Goltsev ...
  72. [72]
    CyTOF XT PRO - Standard BioTools
    The CyTOF XT PRO is a mass cytometry system for analyzing immune responses, with 4x faster speeds, 50+ target capture, and 21 CFR Part 11 compliance.
  73. [73]
    [PDF] Capture the immune markers that matter
    The CyTOF XT PRO system is a powerful cytometry platform with proven sensitivity, dynamic range and resolution to capture 50-plus targets simultaneously, ...
  74. [74]
    Revolutionary 3.7K Protein Assay Leads Standard BioTools ...
    Apr 24, 2025 · New SomaScan Select measures 3700 proteins, covers 70% FDA biomarkers. Explore breakthrough CyTOF XT PRO system with 4x faster throughput.
  75. [75]
    Chicago 2025 - Standard BioTools
    Join us at American Association for Cancer Research® (AACR) Annual Meeting 2025 I Booth # ... Single-cell Analysis Services with CyTOF · Spatial Proteomics ...Missing: cloud- | Show results with:cloud-
  76. [76]
  77. [77]
    Standard BioTools Announces New Product Innovations in Plasma ...
    Apr 24, 2025 · CyTOF XT PRO System: a next-generation platform designed to maximize clinical trial efficiency by delivering clearer, more complete and more ...
  78. [78]
    CyTOF Summit 2024 Video Library - Standard BioTools
    Hear from 10 of our fantastic speakers on their latest findings and insights using CyTOF® and Imaging Mass Cytometry™ technologies.Missing: signatures | Show results with:signatures
  79. [79]
    Caprion Biosciences Offers Mass Cytometry CRO Services to ...
    Jan 8, 2019 · Mass cytometry services for pharmaceutical and biotechnology companies to accelerate development of new health insights.Missing: trials | Show results with:trials
  80. [80]
    CyTOF Solutions for Clinical Trials | Standard BioTools
    CyTOF technology is the only single-cell platform that can simultaneously and deeply profile a broad range of surface and functional markers on the same cell, ...Missing: DVS | Show results with:DVS
  81. [81]
    Mass Cytometry - Standard BioTools
    Cytometry by time-of-flight or mass cytometry (on which CyTOF® technology is based) is a cytometric technique that measures metal-conjugated antibodies ...
  82. [82]
    Mass Cytometry vs Flow Cytometry | Biocompare
    Feb 23, 2023 · Currently, a typical flow cytometry experiment will detect around 8–10 different markers, although it is becoming easier for researchers to ...
  83. [83]
    Mass Cytometry: Single Cells, Many Features - PMC - NIH
    The deep parameterization is sufficient to identify the major cell subsets in a sample with sufficient parameters “left over” for studies of cellular behavior.
  84. [84]
    Mass Cytometry (CyTOF) - Bio-Techne
    To quantify the expression level of proteins on individual cells, metal isotopes can be conjugated to primary antibodies for mass cytometry analysis. Fixed, ...Missing: principles | Show results with:principles
  85. [85]
    Mass cytometry for the multiplexed quantification and ...
    Mar 26, 2023 · Samples were then run as a single barcoded tube on event mode at a rate of 250–500 events per second on a CyTOF® 2 until ~100,000 events per ...
  86. [86]
    Effect of cryopreservation on delineation of immune cell ...
    Feb 2, 2017 · This study examined an analytical and operational feasibility of MC for an in-depth immunophenotyping analysis of the tumor microenvironment.Missing: absolute | Show results with:absolute
  87. [87]
    Multi-site reproducibility of a human immunophenotyping assay in ...
    The study showed consistent cell population frequencies with an average %CV of 14.4% for whole blood and 17.7% for PBMC across multiple sites.Missing: tags | Show results with:tags
  88. [88]
    Minimizing Batch Effects in Mass Cytometry Data - PubMed Central
    Oct 15, 2019 · A barcoding approach allows for multiple samples to be stained together in one tube, reducing the intra-barcode technical variability, and ...
  89. [89]
    About the Data | The COVIDome Project
    Precise mapping of immune cell lineages and subtypes. Single-cell mass cytometry (CyTOF) allowed us to measure over 100 immune cell subtypes, including major ...
  90. [90]
    Mass Cytometry | flow
    The Fluidigm CyTOF is a mass spectrometry cytometer ana lyzer capable of generating data of 48 parameters for each event passing through the detector.Missing: hardware components injector quadrupole vacuum
  91. [91]
    CyTOF - ExpertCytometry
    At the heart of the CyTOF is a very sensitive Time of Flight (TOF) mass spectrometer. This detection system can resolve 143Nd from 144Nd or 152Sm from 153Eu and ...<|separator|>
  92. [92]
    Cytometry by Time of Flight - an overview | ScienceDirect Topics
    Cytometry by time-of-flight (CyTOF) is a recently introduced technology that measures the abundance of metal isotope labels on antibodies and other tags.<|control11|><|separator|>
  93. [93]
    Impact of recent innovations in the use of mass cytometry in support ...
    The cloud of atomic ions for each single cell is extracted into the ion optics and TOF regions of the mass cytometer, where the ions are separated by mass.
  94. [94]
    Mass Cytometry for Clinical Research | Biocompare
    Oct 12, 2021 · The CyTOF is able to do many more cells,” says Karen McKinnon, Assistant Professor and Director of the Immune Monitoring and Genomics Facility ...
  95. [95]
    Mass cytometry panel optimization through the designed distribution ...
    These studies visualize how signal overlap can complicate mass cytometry data interpretation and demonstrate how the rational distribution of interference can ...Missing: isobaric | Show results with:isobaric