The largest database of trusted experimental protocols
> Chemicals & Drugs > Organic Chemical > Rhodamine-dextran-amine dye

Rhodamine-dextran-amine dye

Rhodamine-dextran-amine dye is a fluorescent labeling compound composed of the dye rhodamine conjugated to dextran, a polysaccharide, and an amine group.
This dye is commonly used in biological research for tracing and visualizing cellular and molecular structures.
Rhodamine-dextran-amine dye exhibits bright fluorescence and can be easily detected using fluorescence microscopy or flow cytometry.
It is a versatile tool for studying a variety of biological processes, such as cell lineage tracing, endocytosis, and protein trafficking.
The dextran component helps to improve the solubility and stability of the dye, while the amine group allows for covalent conjugation to target molecules of interest.
Researchers can utilize rhodamine-dextran-amine dye to gain valuable insights into the dynamics and behavior of cells and tissues in a wide range of applications.

Most cited protocols related to «Rhodamine-dextran-amine dye»

To visualize the projections from VCN to MNTB, axons from VCN were labeled in two ways: with lipophilic dyes in fixed tissue or with dextran dyes in fresh tissue slices made in an acute in vitro preparation. In fixed tissue, we used the lipophilic dye NeuroVue Red (PTI Research, Inc., Exton, PA), as previously described (Hsieh and Cramer, 2006 (link); Hsieh et al., 2007 (link)). After perfusion with 4% PFA, the cerebellum was carefully dissected away so that VCN was clearly visible. With fine forceps, a small piece of NeuroVue Red filter paper (100–200 μm2) was placed in VCN. To allow dye transport, brainstems were incubated in 4% PFA at 37° C for 10–14 days. Coronal vibratome sections were then cut at 100 μm, mounted onto slides, and coverslipped with Glycergel mounting medium (Dako).
In fresh tissue, we used dextran dyes in acute brainstem slices maintained in vitro. First, mice were euthanized with an overdose of isoflurane and perfused transcardially with artificial cerebrospinal fluid (ACSF; 130 mM NaCl, 3 mM KCl, 1.2 mM KH2PO4, 2.4 mM CaCl2, 1.3 mM MgSO4, 20 mM NaHCO3, 3 mM HEPES, and 10 mM glucose saturated with 95% O2/5% CO2). Brainstems were quickly removed and thick brainstem slices (0.5–1.0 mm) were cut and maintained in oxygenated ACSF. Dextran amine dyes (Rhodamine or Alexa 488, 3,000 MW; Invitrogen) were used in a 6.25% solution containing 0.4% Triton X-100 in PBS. Brainstems were injected either unilaterally with a single dye or bilaterally with two different dyes. Dye solutions were delivered to VCN using pressure injection through a pulled micropipette attached to a Picospritzer. Current was subsequently applied at the dye injection site. Dye was allowed to transport for 1–3 hours. Tissue was fixed overnight in 4% PFA in PBS at 4°C, rinsed, then sectioned on a vibratome at 100 μm, mounted on slides and coverslipped with Glycergel mounting medium.
In some cases we delivered small deposits of rhodamine dextran amine (RDA) or horseradish peroxidase (HRP) in order to label small numbers of identifiable individual axons in P6–28 ephrin-B2lacZ/+ mice. Using a pulled glass pipette coated with concentrated RDA or HRP, three to four deposits were made in the ventral acoustic stria, just ventromedial to the VCN. Dye was allowed to transport for 2 hours and brainstems were fixed in 4% PFA at 4°C overnight. Brainstems were sectioned coronally at 150–200 μm on a vibratome, reacted for DAB (for HRP deposits), mounted on slides, and coverslipped.
Publication 2010
Acoustics Amines Axon Bicarbonate, Sodium Brain Stem Cerebellum Cerebrospinal Fluid Dextran Drug Overdose Ephrins Forceps Glucose HEPES Horseradish Peroxidase Isoflurane Mus Perfusion Pressure Rhodamine rhodamine dextran Sodium Chloride Striae Distensae Sulfate, Magnesium Tissues Triton X-100
In 11 animals at P8 and 13 animals at P13 we performed neuronal tracing to fill calyces of Held in the MNTB. Mice were perfused transcardially with artificial cerebrospinal fluid (aCSF; 130 mM NaCl, 3 mM KCl, 1.2 mM KH2PO4, 20mM NaHCO3, 3 mM HEPES, 10 mM glucose, 2 mM CaCl2, 1.3 mM MgSO4 perfused with 95% O2 and 5% CO2). Brains were quickly dissected and placed in a chamber with oxygenated aCSF.
The brain was temporarily transferred to a Petri dish with aCSF and axonal projections from the AVCN to the MNTB were filled using a rhodamine dextran amine (RDA; MW 3000, Invitrogen) solution (6.35% RDA with 0.4% Triton-X100 in PBS). A pulled glass micropipette was filled with RDA and pulses of RDA were delivered close to the midline in the ventral acoustic stria (VAS) using an Electro Square Porator (ECM830; BTX) at a rate of 5 pulses per second (pps) at 55 V for 50 ms. These pulses resulted in sparse labeling of GBC axons and calyces of Held on both sides of the brainstem (Figures 1C,D). In some cases, sequential labeling was performed with Alexa 488 dextran amine (DA488; 6.35% with 0.4% Triton-X100 in PBS) so that a single brainstem would have AVCN axons sparsely labeled with two different fluorophores. The brain was then placed back into the aCSF chamber for approximately 2 h under continuous oxygenation to allow for dye transport. The tissue was then transferred to 4% PFA solution for another 2 h followed by incubation in 30% sucrose solution in 0.1 M PBS. Brainstems were cryosectioned in the coronal plane at 18 μm and mounted on an alternating series of four slides. Slides with adequately labeled axons and calyces of Held were immunolabeled for parvalbumin (PV) at P8 and for vesicular glutamate transporters 1 and 2 (VGluT1/2) at P13.
Full text: Click here
Publication 2019
Acoustics Amines Animals ARID1A protein, human Axon Bicarbonate, Sodium Brain Brain Stem Cell Respiration Cerebrospinal Fluid Dextran Glucose HEPES Hyperostosis, Diffuse Idiopathic Skeletal Kidney Calices Mice, Laboratory Neurons Parvalbumins Pulse Rate Pulses rhodamine dextran Sodium Chloride Striae Distensae Sucrose Sulfate, Magnesium Tissues Triton X-100 Vesicular Glutamate Transport Protein 1
Four types of MNPs were used: (i) Maghemite (γ-Fe2O3) fluorinated magnetic nanoparticles synthesized by nucleation, followed by controlled growth of γ-Fe2O3 thin films onto gelatin RITC-iron oxide nuclei (RITC, Rhodamine Isothiocyanate) according to the description in previous publication [31 ]. (ii–iv) Magnetite (Fe304) core particles with different coatings (Chemicell, Berlin, Germany). We studied nano-screenMAG–UC/C (uncoated, cationic), nano-screenMAG-D (coated with starch) and nano-screenMAG-DXS (coated with dextran sulfate) particles. The nano-screenMAG particles consist of a magnetite core surrounded by a lipophilic fluorescent dye covered by a hydrophilic matrix (starch or dextran). The nanoparticles have a red fluorescence (excitation: 578; emission: 613) (Table 1).

Summary of magnetic nanoparticle core and coating properties

Particle typeHydrodynamic diameter (nm)Dry diameter (nm)ChargeCoatingFunctional group
Uncoated-magnetite MNPs10010CationicNo coating
Starch-magnetite MNPs10010NeutralStarchHydroxyl groups
Dextran-magnetite MNPs10010NeutralDextranSodium sulfate
Uncoated-maghemite MNPs10020AnionicNo coatingCarboxyl and amine groups
Full text: Click here
Publication 2016
Amines Cations Cell Nucleus Dextran ferric oxide Fluorescence Fluorescent Dyes gamma-ferric oxide Gelatins Magnetite rhodamine isothiocyanate Starch Sulfate, Dextran
The CT was accessed through the tympanic bulla in order to apply a dextran tracer. A longitudinal incision on the ventral neck revealed the digastric and masseter muscles. The tympanic bulla sits roughly between the two muscles though much farther dorsal. A small hole was made through the bulla where the CT was visualized between the ossicles and cut. The nerve stump proximal to the brain was labeled generously with crystals of 3kD biotinylated rhodamine-labeled dextran amine (D-7162: Invitrogen; Carlsbad, CA). A small piece of parafilm was placed over the opening in bulla before closing the incision (adapted from Mangold and Hill, 2008 (link)).
However, this method brought limitations to the current studies primarily because the optimal time to observe anterograde dextran tracer (i.e., CT afferents in the nTS) is two to three days. As the dye slowly dissipates with longer survival times, it becomes impractical to localize the CT projections in longer-term survivals. Further, in studying glial reactivity it was not desirable to introduce another source of damage. Therefore, the antibodies to purinergic receptors P2X2 and P2X3 were tested and subsequently utilized to define the area of termination of primary CT afferents (see Figure 1).
Publication 2012
Amines Amputation Stumps Antibodies Brain Dextran Muscles, Masseter Muscle Tissue Neck Nervousness Neuroglia Receptors, Purinergic P2X2 rhodamine dextran Tympanic Cavity
In 19 animals at P14, we performed neuronal tracing to fill calyces of Held in the MNTB. Mice were perfused transcardially with artificial cerebrospinal fluid (aCSF; 130 mM NaCl, 3 mM KCl, 1.2 mM KH2PO4, 20 mM NaHCO3, 3 mM HEPES, 10 mM glucose, 2 mM CaCl2, 1.3 mM MgSO4 perfused with 95% O2 and 5% CO2). Brains were quickly dissected and placed in a chamber with oxygenated aCSF. Axonal projections from the anteroventral VCN (AVCN) to the MNTB were filled using a pulled glass micropipette filled with rhodamine dextran amine (RDA; MW 3000, Invitrogen) solution (6.35% RDA with 0.4% Triton-X100 in PBS), which was then electroporated (Electro Square Porator (ECM830; BTX) with 5 pulses per second (pps) at 55 V for 50 ms into the ventral acoustic stria close to the midline. These pulses resulted in sparse labeling of globular bushy cell (GBC) axons and their terminal calyces of Held on both sides of the brainstem. The brain was then placed back into the aCSF chamber for approximately 2 h under continuous oxygenation to allow for dye transport. The tissue was then transferred to 4% PFA solution for another 2 h followed by incubation in 30% sucrose solution in 0.1 M PBS. Brainstems were cryosectioned in the coronal plane at 18 μm and mounted on an alternating series of five slides. Slides with adequately labeled axons and calyces of Held were immunolabeled for VGLUT1/2 as described above.
Publication 2021
Acoustics Amines Animals ARID1A protein, human Axon Bicarbonate, Sodium Brain Brain Stem Cell Respiration Cells Cerebrospinal Fluid Glucose HEPES Kidney Calices Mice, Laboratory Neurons Pulses rhodamine dextran Sodium Chloride Striae Distensae Sucrose Sulfate, Magnesium Tissues Triton X-100

Most recents protocols related to «Rhodamine-dextran-amine dye»

Spinal cord slice preparations. Experiments were performed on spinal sections isolated from 23 neonatal (P1-P16) Sim1 Cre/+ ;Rosa26 floxstopTdTom/+ /Gt(Rosa)26 floxstopH134R/EYFP/+ (referred to as Sim1TdTom/ChR2) mice. Isolation and preparation of sections, and electrophysiological recording methods have been previously described (Chopek, Nascimento et al. 2018) . Briefly, animals were anaesthetized with isoflurane, decapitated at the medulla-spinal cord junction and spinal cords dissected out in ice-cold dissecting artificial cerebrospinal fluid (aCSF), composed of (mM): KCl (3.5), NaHCO3 (35), KH2PO4 (1.2), MgSO4 (1.3), CaCl2 (1.2), glucose (10), sucrose (212.5), MgCl2 (2.2), and equilibrated to pH 7.4 with 95% 02 & 5% C02. Once dissected free, thoracic spinal cords were immediately secured in agarose and sectioned at 350 µm using a vibratome (Leica VT1200S, Leica) and then incubated in warm (30 o C) aCSF for a minimum of 30 minutes before performing electrophysiological experiments. Incubation and recording aCSF was composed of (mM): NaCl (111.0), KCl (3.085), D-glucose (10.99), NaHCO3 (25.0), MgSO4 . 7H2O (0.31), CaCl2 (2.52), KH2PO4
(1.1), equilibrated to pH 7.4 with 95% O2 & 5% CO2.
Intact spinal cord preparations. Spinal cords were dissected free from Sim1TdTom/ChR2 mice as described above in 'Slice preparations' but remained longitudinally intact with dorsal and ventral roots attached. Once free, connective tissue was carefully removed from spinal cord tissue and secured ventral side up with fine insect pins. To retrogradely label SPNs, rhodamine dextran amine dye (RDA) was applied to cut T6-T8 ventral roots on one or both sides of the spinal cord using glass suction pipettes with internal diameters of 100-120 µm. Ventral roots were cut close to their exit from the spinal cord to minimize labeling time. Retrograde labeling of thoracic SPNs continued in the dark at room temperature for at least 3 hours (Szokol, Glover et al. 2008) . A block of agar was prepared in advance with one side of the block cut with a scalpel to provide a 30-degree incline. The spinal cord was then mounted on the agar block and fixed in place with acrylic glue to expose the corresponding labeled thoracic spinal segment. The spinal cord at the level of the agar block was then sectioned with a vibratome. This portion of the spinal cord was then glued to a sylgard-coated (Sylgard, Dow Corning, MI, USA) recording chamber designed and 3D printed in-house specifically for these experiments. In particular, the obliquely cut surface of the spinal cord was placed on a sylgard 'ramp' to enable visualisation and patch-clamp recordings of SPNs under fluorescence while preserving and maintaining continuity with the lumbar spinal region for optical stimulation.
Whole cell patch-clamp recordings and optogenetic stimulation. Slices or spinal cord preparations were transferred to a recording chamber mounted on a Zeiss AxioExaminer microscope and perfused with oxygenated room-temperature aCSF. Cells were visualized using a 20x wide aperture (1.2 nA) water-immersion objective lens, a CCD camera (CoolSNAP EX OCD Camera, Photometrics, AZ) and Slidebook 6.0 software (Intelligent Imaging Innovations, CO, USA, RRID:SCR_014300). Patch pipettes were pulled with a P-97 Sutter puller and those with 4-6 MΩ resistances were filled with the following (mM): K-gluconate (128); NaCl (4); CaCl2 (0.0001); Hepes (10), glucose (1); Mg-ATP (5); and GTP-Li (0.3). Whole cell patch-clamp recordings were made under current-clamp using a Multiclamp 700B amplifier (Molecular Devices, California, USA, RRID: SCR_014300). Recordings were low pass filtered at10 kHz and acquired at 25 kHz with CED Power 1401 AD board and displayed and recorded using Signal software (Cambridge Electronic Design, Cambridge UK). Before performing an optical stimulation protocol, rheobase (defined as the minimum current to elicit a single AP was collected to determine cell excitability). Before the optical stimulation protocol, we recorded rheobase and repetitive firing in response to 1 second depolarizing current steps from each SPN. In slice preparations, presumed SPNs (small neurons visualized in the IML, with TdTom fluorescence noted near the soma) were patched and responses recorded. Using a spatial light modulator system as previously described (Chopek, Nascimento et al. 2018 , Chopek, Zhang et al. 2021 ), a region of interest slightly greater than the SPN soma was created for optical stimulation of presumed V3 terminals apposing the patched SPN. Blue light was delivered in five 500 ms pulses at 5 Hz at a laser power of ~2.5 mW. For intact cord preparations with thoracic surface exposed, SPNs that were retrogradely labelled with RDA were patched and responses recorded. A region of interest covering the ventral L2 segment was created for optical stimulation of presumed ventral L2 V3 neurons and likely axons of passage from caudal lumbar segments. Blue light was delivered in five 500 ms pulses at 5 Hz at a laser power of ~2.5 mW. In a subset of slice experiments, lumbar V3 neurons were patched and optically activated to confirm that optical stimulation resulted in AP generation in V3 neurons.
Publication 2023

Example 1

In this example the binding molecules are pMHC complexes, the linker is a dextran-streptavidin-fluorochrome conjugate, and the label is a DNA oligonucleotide.

The sample is a HPBMC from humans, the isolating and/or detecting is done by flow cytometry (FACS), and the determining of the identity of the label is done by quantitative PCR (QPCR).

This is an example where the Sample (1) was blood from one CMV positive and HIV negative donor which was modified (1b) to generate Peripheral blood mononuclear cells (PBMCs).

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The binding molecules (3) were peptide-MHC (pMHC) complexes displaying either CMV (positive antigen) or HIV (negative antigen) derived peptide-antigens. The binding molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules were purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of a control T-cell population.

The Labels (4) were oligonucleotides applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag. The labels were combined oligonucleotide labels arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label. The detection molecule (5) was synthetized (5a) by attaching binding molecules in the form of biotinylated pMHC and labels in the form of biotin-modified double stranded oligonucleotides onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. Two different detection molecules were generated wherein the two individual detection molecules containing different pMHC were encoded by corresponding individual oligonucleotide labels.

An amount of sample, PBMC's (1b) was incubated with an amount of mixed detection molecules (5) under conditions (6c) that allowed binding of detection molecules to T cells in the sample.

The cell-bound detection molecules were separated from the non-cell bound detection molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind detection molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells were subjected to quantitative PCR analysis of the oligonucleotide label associated with the detection molecules bound to the isolated cells to reveal the identity of the detection molecules that bound to the T cells present in the sample. This example thus revealed the presence of T cells in the blood expressing a T cell receptor that binds to peptide-MHC molecules represented within the library of Detection Molecules. It also revealed the feasibility of enriching for Detection Molecules based on the presence of T cells specific for the positive peptide-antigen (CMV) over the negative peptide-antigen (HIV) antigens.

    • 1. Sample preparation. The cell sample used in this example was obtained by preparing PBMC's from blood drawn from a donor that was CMV positive as well as HIV negative as determined by conventional MHC-multimer staining.
      • a. Acquiring sample: Blood was obtained from the Danish Blood Bank, in the form of buffy coats (BC). A peripheral blood mononuclear cell preparation obtained following standard donor blood preparations.
      • b. Modifying sample: Peripheral blood mononuclear cells (PBMCs) were isolated from whole blood by density gradient centrifugation. The density gradient medium, Lymphoprep (Axis-Shield), which consists of carbohydrate polymers and a dense iodine compound, facilitate separation of the individual constituents of blood. Blood samples were diluted 1:1 in RPMI (RPMI 1640, GlutaMAX, 25 mM Hepes; gibco-Life technologies) and carefully layered onto the Lymphoprep. After centrifugation, 30 min, 490 g, PBMCs together with platelets were harvested from the middle layer of cells. The isolated cells, the buffy coat (BC), was washed twice in RPMI and cryopreserved at −150° C. in fetal calf serum (FCS; gibco-Life technologies) containing 10% dimethyl sulfoxide (DMSO; Sigma-Aldrich). BC's used in this example are listed in table 1 along with peptide-antigen specific T cells identified within these samples by conventional MHC multimer staining.
    • 2. Linker preparation: The linker used in this example was a dextran molecule, to which was attached streptavidin and fluorochromes. The streptavidin served as attachment sites for biotinylated oligonucleotides (Label) and biotinylated peptide-MHC complexes (Binding Molecules). The fluorochrome allowed separation of cells bound to detection molecules from cells not bound to detection molecules.
      • i. In this example linkers were linear and branched dextran molecules with covalently attached streptavidin (5-10 per linker) and fluorochromes (2-20 per linker) in the form of PE. Linkers were essentially Dextramer backbone as described by Immudex.
    • 3. Binding molecules preparation: The binding molecules used in this example were two different class I MHC-peptide complexes. MHC heavy chains (HLA-A0201 and HLA-B0702) and B2M were expressed in E.coli as previously described (Hadrup et al. 2009) and each MHC-I complex generated with two peptide antigens. The individual specificities (allele and peptide combination) were generated in the following way:
      • a. Synthesis: Binding molecules in this example were specific pMHC monomers that were produced from UV-exchange of selected HLA-I monomers carrying a photolabile 9-residue peptide-ligand (p*). When exposed to UV-light (366 nm) the photolabile ligand will be cleaved and leave the binding groove empty. Due to the instability of empty MHC-I molecules, the complexes will quickly degrade if they are not rescued by replacement with another peptide that match that HLA-type. In this way specific pMHC monomers were produced by mixing excess of desired HLA ligands with p*MHC monomers. p*MHC monomers were refolded, biotinylated and purified as previously described (Hadrup et al. 2009).
        • i. HIV derived peptide ILKEPVHGV from antigen HIV polymerase and CMV derived peptide TPRVTGGGAM from antigen pp65 TPR (Pepscan Presto, NL) were diluted in phosphate buffered saline (DPBS; Lonza) and mixed to final concentrations 100 μg/ml:200 μM (HLA-A0201: ILKEPVHGV and HLA-B0702:TPRVTGGGAM). The mixtures were exposed to 366 nm UV light (UV cabinet; CAMAG) for one hour and optionally stored for up to 24 h at 4° C.
      • b. Modification: No further modifications
      • c. Purification: Before applying the peptide exchanged HLA monomer Binding Molecules for preparation of detection molecules these were centrifuged 5 min, 3300 g, to sediment any aggregated MHC molecules.
    • 4. Label preparation: In this example, two different double stranded oligonucleotides, DNA barcodes, of the same length but partially different sequence, were generated. Each of the DNA barcodes would become attached to a specific pMHC, and thus functioned as a Label for this specific Binding Molecule. The oligonucleotides were biotinylated, allowing easy attachment to the dextran-streptavidin conjugate linker.
      • a. Synthesis: The Labels were generated from partially complementary oligonucleotides which were purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM oligonucleotides were made in nuclease free water and stored at −20° C.
        • i. The Label system used in this example was named 2OS and was developed to increase the complexity of a limited number of oligonucleotide sequences. This was enabled by applying a combinatorial strategy where two partially complementary oligonucleotides (an A oligonucleotide with a 5′ biotin tag and a B oligonucleotide) where annealed and then elongated to produce new unique oligonucleotide-sequences (AxBy) which were applied as a DNA-barcodes (Labels) (see FIG. 9 for design of the DNA-barcodes).By combining 2 unique oligonucleotide-sequences (A label precursor) that were partly complementary to 2 other unique oligonucleotide sequences (B label precursor) a combinatorial library of 4 different (AxBy) Labels were produced. Only 2 of these Labels were used in this example (A1B1 and A2B2). Refer to table 2 for an overview of the different 2OS A and B nucleotide sequences. Briefly:
        • Partly complementary A and B oligonucleotides were annealed to produce two combined A+B DNA barcodes (A1+B1 to produce A1B1 and A2+B2 to produce A2B2). The respective A and B oligonucleotides were mixed as stated in table 3, heated to 65° C. for 2 min and cooled slowly to <35° C. in 15-30 min. The annealed A and B oligonucleotides were then elongated as stated in table 3. Elongation reagents were mixed <15 min before use. After mixing, the reactions were incubated 5 min, RT, to allow elongation of the annealed oligonucleotides. The reagents used for annealing (left) and elongation (right) of partly complementary oligonucleotides is described in table 3. Reagents marked in italic were from the Sequenase Version 2.0 DNA Sequencing Kit (Affymetrix #70770).
      • b. Modification: All labels were diluted to working concentrations (2.17 uM) in nuclease free water with 0.1% Tween and stored at −20° C.
      • c. Purification: Labels were not purified further.
    • 5. Detection Molecules preparation: The Binding Molecules (pMHCs) and Labels (DNA-barcodes) were attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Synthesis: For preparation of Detection Molecules the 2OS DNA-barcodes were attached to the dextramer prior to addition of pMHC. 1 ul dextramer was used for every 3 ul of prepared Detection Molecule. Briefly, for generation of Detection Molecules:
        • i. The Label:linker conjugate were generated by addition of label in two fold excess over linker (label:linker, 2:1) i.e. 1×0.16 uM linker (dex), were mixed with 0.15×2.17 uM label (2OS DNA-barcode) and incubated at least 30 min, 4° C.
        • ii. Binding molecules, in the form of biotinylated UV-exchanged peptide-MHC monomer, were added to the label:linker conjugate to reach a concentration of 44 ug/ml of the given pMHC in 3 ul, and incubated 30 min, RT.
        • The final volume was reached by addition of 0.02% NaN2 in PBS together with D-biotin (Avidity Bio200) in a final concentration of 12.6×10−6M. The detection molecule preparation was incubated 30 min, 4° C., and optionally stored for up to 4 weeks at 4° C. (overview of the amounts used can be found in table 4).
        • Two sets of two detection molecules were generated. Each set with the two specificities individually labeled. The label was inverted between the two sets as described below.
          • 1. 1×CMV specific pMHCs (HLA-A0201: ILKEPVHGV) coupled to 2OS-A1B1, 1×HIV specific pMHCs (HLA-B0702:TPRVTGGGAM) coupled to 2OS-A2B2.
          • 2. 1×CMV specific pMHCs (HLA-A0201: ILKEPVHGV) coupled to 2OS-A2B2, 1×HIV specific pMHCs (HLA-B0702:TPRVTGGGAM) coupled to 2OS-A1B1.
      • b. Modification: No further modifications were performed
      • c. Purification: The Detection Molecules were centrifuged 5 min, 3300 g, to sediment any aggregates before being added to the cell sample,
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were mixed in one container to allow Detection Molecules to bind to T cells.
      • a. Amount of sample: 1×10E6-2×10E6 cells in the form of BC's, were used.
      • b. Amount of detection molecule: According to table 4. for staining in 100 ul 1.32 ug/ml calculated in relation to each binding molecule (specific peptide-MHC) on the Detection Molecule was required per incubation, i.e. 3 ul of each Detection Molecule.
      • c. Conditions: BCs were thawed in 10 ml, 37° C., RPMI with 10% fetal bovine serum (FBS), centrifuged 5 min, 490 g, and washed twice in 10 ml RPMI with 10% FBS. All washing of cells refer to centrifugation 5 min, 490 g, with subsequent removal of supernatant. 1×10E6-2×10E6 cells were washed in 200 ul barcode-buffer (PBS/0.5% BSA/2 mM EDTA/100 μg/ml herring DNA) and resuspended in this buffer to approximately 20 μl per sample. The barcode buffer is optimized to increase the stability of the oligonucleotides associated with the Detection molecules. The cells were incubated with 50 nM dasatinib, 30 min, 37° C., prior to incubation of cells with Detection Molecules.
        • i. The Detection molecules were added in the required amount. If necessary barcode-buffer was added to reach a total volume of 100 ul and cells were incubated 15 min, 37° C. 20 ul antibody mixture containing PerCP-conjugated anti-CD8 antibody and dump channel FITC-conjugated antibodies (table 5) along with 0.1 μl near-IR-viability dye (Invitrogen L10119) was added for every 100 ul of PBMC:detection molecule sample. The samples were incubated 30 min, 4° C., and cells were washed twice in 200 ul barcode-buffer. Optionally cells were fixed in 1% paraformaldehyde in DPBS overnight, 4° C., and washed twice in barcode-buffer. Fixed cells were stored for up to a week at 4° C.
    • 7. Enrichment of detection molecules with desired characteristics: The Detection Molecules were enriched by using flow cytometry, more specifically, Fluorescence-Activated-Cell-Sorting (FACS). Since all Detection Molecules carried a PE-fluorescent label, the cells that bound to Detection Molecules would fluoresce according to this fluorochromes emission peak. By applying a FACS sorter this feature could be used to separate cells that bound to detection molecules from cells that did not bind Detection Molecules i.e. to separate those cells that did fluoresce from those cells that did not fluoresce. As a result the Detection Molecules that bound to cells, and hence the associated Labels, were enriched for.
      • a. Apply: Cells were sorted on a BD FACSAria, equipped with three lasers (488 nm blue, 633 nm red and 405 nm violet). The flow cytometry data analyses were performed using the BD FACSDiva software version 6.1.2. The following gating strategy was applied: Lymphocytes were identified in a FSC/SSC plot. Additional gating on single cells (FSC-A/FSC-H), live cells (near-IR-viability dye negative), and CD4, CD14, CD16, CD19, CD40 negative (FITC)/CD8 positive cells (PerCP) were used to define the CD8 T cell population (table 5). The PE positive population, i.e. the cells that bound to detection molecules, were defined within the PerCP positive population
      • b. Wash: not applied
      • c. Separate: The PE positive cells were sorted by FACS, as described in 7a, into tubes that contained 200 μl barcode-buffer and that had been pre-saturated for 2 h-O.N. in 2% BSA. The sorted cells were centrifuged 5 min, 5000 g, to allow removal of excess buffer (<10 ul should reside in the container). Cells were stored at −80° C.
    • 8. Identification of enriched Detection Molecules: Since the Detection Molecules were enriched based on specific binding of the Binding Molecule (the pMHC) to cells, the identification of the associated Labels (oligonucleotide barcodes) amongst the sorted cells would also reveal the pMHCs that had bound to cells in the PBMC sample. In this example the enriched Detection Molecules were identified by quantitative PCR (QPCR) using Label-specific fluorescent reporter probes.
      • a. Apply: Labels derived from sorted cells were analyzed by QPCR with the Brilliant II QRT-PCR Low ROX Master Mix Kit (Agilent technologies, #600837) according to table 6. PCR was run on the thermal cycler: Mx3000P qPCR system (Agilent Technologies). The thermal profile is listed in table 7 and the label-specific fluorescent reporter probes were: Probe123: 5′6-FAM/GCCTGTAGTCCCACGCGATCTAACA/3′BHQ_1 for detection of label 2OS-A1B1 and Probe124: 5′HEX/CAACCATTGATTGGGGACAACTGGG/3′BHQ_1 for detection of label 2OS-A2B2. The label specific reporter probes were purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM oligonucleotides were made in nuclease free water and stored at −20° C. Primers used for amplification in this experiment forward: GAAGTTCCAGCCAGCGTC, and reverse: CTGTGACTATGTGAGGCTTTC.
      • b. Analysis: combined with the above.

After sorting of PE labeled cells and QPCR the resultant Ct values confirmed that Detection Molecules were enriched, i.e. 2OS DNA-barcode Labels were successfully recovered, only when associated with the CMV epitope, while they were not detected when associated with the HIV epitope (FIG. 10). This was observed even when labels were inverted between the two Binding Molecules, implying that the recovery was not Label specific, but truly specific for the Binding molecule.

It was verified that the 2OS labels were recovered after cellular interaction, sorting and QPCR only if they were associated with positive control Detection molecules, and not if they were associated with negative control Detection molecules.

Example 2

In this example the binding molecules are pMHC complexes, the linker is a dextran-streptavidin-fluorochrome conjugate, and the label is a DNA oligonucleotide.

The sample is a HPBMC, the isolating and/or detecting is by FACS, and the determining of the identity of the label is by QPCR.

This is an example where the Sample (1) was blood from one CMV positive and HIV negative donor which was modified (1b) to generate Peripheral blood mononuclear cells (PBMCs).

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The example is similar to example 1 except that a 1000 fold excess of Detection Molecules with irrelevant Binding Molecules but without label were included. The Binding Molecules used (3) are peptide-MHC (pMHC) complexes displaying either CMV (positive antigen) or HIV (negative antigen) derived peptide-antigens or pMHC complexes displaying irrelevant peptide antigen. The Binding Molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding Molecules were purified (2c) by HPLC.

The Labels (4) were single stranded oligonucleotides applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag.

The Detection Molecule (5) was synthetized (5a) by attaching binding molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides, DNA-barcodes, onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. Three different detection molecules were generated wherein the two of these individual detection molecules containing CMV- and HIV-directed pMHC were encoded for by corresponding individual DNA-barcodes. Detection Molecules with irrelevant Binding Molecules (p*MHC) were not encoded for with a DNA-barcode.

An amount of sample, PBMC's (1b) was incubated with an amount of mixed detection molecules (5) in a ratio of 1:1:998 (Labeled-CMV:Labeled-HIV:unlabeled-irrelevant directed Binding Molecules) under conditions (6c) that allowed binding of detection molecules to T cells in the sample.

The cell-bound detection molecules were separated from the non-cell bound detection molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind detection molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells were subjected to quantitative PCR analysis of the oligonucleotide label associated with the detection molecules bound to the isolated cells to reveal the identity of the detection molecules that bound to the T cells present in the sample. This example thus revealed the presence of T cells in the blood expressing a T cell receptor that binds to peptide-MHC molecules represented within the library of Detection Molecules. It also revealed the feasibility of enriching for Detection Molecules based on the presence of T cells specific for the positive peptide-antigen (CMV) over the negative peptide-antigen (HIV) antigens. In this setting, it was proved that it was possible to specifically enrich Detection Molecules associated with positive peptide-antigen when excess of irrelevant Detection Molecule was included in the sample incubation.

    • 1. Sample preparation. The cell sample used in this example was obtained in the same way as described in example 1
    • 2. Linker preparation: The linker used in this example was prepared as in example 1
    • 3. Binding Molecules preparation: The binding molecules used in this example were two different class I MHC-peptide complexes. MHC heavy chains (HLA-A02 and HLA-B07) and B2M were expressed in E.coli as previously described (Hadrup et al. 2009) and each MHC-I complex generated with two peptide antigens or left with an irrelevant photolabile 9-mer peptide. The individual specificities (allele and peptide combination) were generated in the following way:
      • a. Synthesis: As in example 1.
        • i. HIV derived peptide ILKEPVHGV from antigen HIV polymerase and CMV derived peptide TPRVTGGGAM from antigen pp65 TPR (Pepscan Presto, NL) were diluted in phosphate buffered saline (DPBS; Lonza) and mixed to final concentrations 100 μg/ml:200 μM (HLA-A0201: ILKEPVHGV and HLA-B0702:TPRVTGGGAM). The mixtures were exposed to 366 nm UV light (UV cabinet; CAMAG) for one hour and optionally stored for up to 24 h at 4° C. The irrelevant Binding Molecules, p*HLA-A0201 and p*HLA-B0702 were mixed in equal amounts (pg/ml) and diluted to 100 μg/ml in DPBS.
      • b. Modification: No further modifications
      • c. Purification: Before applying the peptide exchanged HLA monomer Binding Molecules for preparation of detection molecules these were centrifuged 5 min, 3300 g, to sediment any aggregated MHC molecules. The p*MHC monomers were not purified further.
    • 4. Label preparation: In this example Labels were synthetic oligonucleotides modified with biotin for coupling to the Linker.
      • a. Synthesis: The Labels were oligonucleotides, DNA barcodes, which were purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM oligonucleotide were made in nuclease free water and stored at −20° C.
        • i. The Label system used in this example was named 1OS and comprised of single stranded oligonucleotides which were applied as a DNA barcodes (Oligo 4: 5GAGATACGTTGACCTCGTTGAANNNNNNTCTTGAACTATGA ATCGTCTCACTTAAGCTCTTGGTTGCAT and Oligo 5: 5GAGATACGTTGACCTCGTTGAANNNNNNTCTATAGGTGTC TACTACCTCACTTAAGCTCTTGGTTGCAT were used in this experiment, 5 indicates a 5′ biotin modification).
      • b. Modification: All Labels were diluted to working concentrations (2.17 uM) in nuclease free water with 0.1% Tween and stored at −20° C.
      • c. Purification: Labels were not purified further.
    • 5. Detection Molecules preparation: The Binding Molecules (pMHCs) and Labels (oligonucleotides) were attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Synthesis: In this example the Detection Molecules were prepared by attaching 1OS DNA-barcodes prior to addition of pMHC. Detection Molecules were essentially generated in the same way as described in example 1, with the difference that two different sets of two detection molecules were generated.
        • Each set had two specificities individually labeled. The Labels were inverted between the two sets as described below. Moreover a third Detection Molecule was generated without any Label, but comprised of the Linker and a Binding Molecule (p*MHC), this detection molecule was included in both sets of Detection Molecules as described below in the indicated stoichiometry:
        • 1. 1×CMV specific pMHCs (HLA-A0201: ILKEPVHGV) coupled to Oligo4, 1×HIV specific pMHCs (HLA-B0702:TPRVTGGGAM) coupled to Oligo5 and 998× non-labeled p*MHC (HLA-A0201:p* and HLA-B0702:p*).
          • 2. 1×CMV specific pMHCs (HLA-A0201: ILKEPVHGV) coupled to Oligo5, 1×HIV specific pMHCs (HLA-B0702:TPRVTGGGAM) coupled to Oligo4 and 998× non-labeled p*MHC (HLA-A0201:p* and HLA-B0702:p*).
      • b. Modification: Since the total volume of each set of Detection Molecules exceeded 100 μl this volume was reduced to reach a desired concentration of specific Binding Molecules in the pooled solution of Detection Molecules.
        • i. Size exclusion spin columns (Nanosep 300K Omega, Pall Corporation) with a cut-off at 300 kDa were saturated by adding 500 μl 2% BSA/DPBS and centrifuging 5000 g, until the volume had passed through. Subsequently, the columns were washed twice by adding 500 μl DPBS and centrifuging 5000 g until no volume was left in the columns. Each pooled set of detection molecules were added to a spin column and centrifuged 5000 g, 4° C., until the desired volume resided in the column (80 μl per incubation with sample). The reduced volume of each set of Detection Molecules was moved to new containers.
      • c. Purification: The Detection Molecules were centrifuged 5 min, 3300 g, to sediment any aggregates before being added to the sample.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were incubated in the same way as described in example 1.
    • 7. Enrichment of Detection Molecules with desired characteristics: The Detection Molecules were enriched for in the same way as described in example 1.
    • 8. Identification of enriched Detection Molecules: Enriched Detection Molecules were essentially identified in the same way as described in example 1, with the difference that two different label-specific fluorescent reporter probes were applied: LNA-4: 5′6-FAM/TCT[+T][+G][+A]AC[+T][+A]TG[+A][+A][+T]CGTC/3′BHQ-1-plus for detection of Oligo4 and LNA-5: 5′HEX/TCT[+A][+T][+A]GG[+T][+G]TC[+T][+A][+C]TACC/3′BHQ-1-plus for detection of Oligo5 ([+X] indicating locked nucleic acids (LNAs)). Primers used for amplification of Oligo 4 and Oligo 5, forward: GAGATACGTTGACCTCGTTG and reverse: ATGCAACCAAGAGCTTAAGT.

After sorting of PE labeled cells and QPCR the resultant Ct values confirmed that Detection Molecules were enriched, i.e. 1OS DNA-barcode Labels were successfully recovered, only when associated with the CMV epitope, while they were not detected when associated with the HIV epitope (FIG. 11). This was observed even when labels were inverted between the two Binding Molecules, implying that the recovery was not Label specific, but truly specific for the Binding molecule. Moreover the example demonstrated that a great amount of irrelevant Detection molecule equipped with the same fluorescent label as all Detection molecules in the incubation would not be detrimental for enrichment of cells that would fluoresce due to specific binding with a Detection molecule.

It was verified that the 1OS Labels were recovered after cellular interaction, sorting and QPCR only if they were associated with positive control Detection molecules, and not if they were associated with negative control Detection molecules, even in the presence of a high amount of irrelevant Detection molecule.

Example 3

In this example the binding molecules are pMHC representing 6 different HLA-alleles, the linker is dextran-streptavidin-PE conjugate and the label is a DNA oligonucleotide.

The sample is HPBMC, the isolating and/or detecting is by FACS, and the determining of the identity of the label is by sequencing.

This is an example where the Samples (1) were blood from one donor that were HLA-B0702:CMV pp65 TPR positive and another donor that were HLA-B0702 negative which were modified (1b) to generate Peripheral blood mononuclear cells (PBMCs). These samples were mixed in different ratios to generate new samples with different but known frequencies of T cells specific toward the HLA-B0702:CMV epitope.

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) were peptide-MHC (pMHC) complexes displaying one out of 110 different peptide-antigens comprised within 6 different HLA-types. The MHC molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules were purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T-cell populations.

The Labels (4) were single stranded oligonucleotide applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag.

The Detection Molecule (5) was synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. A library of 110 different Detection Molecules were generated wherein individual Binding Molecules, comprised of different pMHC, were encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, PBMC's (1b) was incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules were separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells were subjected to PCR for specific amplification of the DNA-barcode Label associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product revealed the identity of Detection Molecules that bound to T cells present in the sample.

This example thus revealed the presence of T cells in the blood expressing a T cell receptor that binds to pMHC molecules represented within the library of Detection Molecules. The number of sequencing reads mapped to a given DNA-barcode and its corresponding Binding Molecule would mirror the frequency of the T cells found by conventional MHC multimer stainings using the same Binding Molecule.

    • 1. Sample preparation. The cell samples used in this example was obtained by preparing PBMC's from blood drawn from one donor that were HLA-B0702:CMV pp65 TPR positive and from another donor that were HLA-B0702 negative, as determined by conventional pMHC-multimer and antibody stainings.
      • a. Acquiring sample: As in example 1
      • b. Modifying sample: PBMCs were isolated from whole blood as described in example 1.
        • i. Mixing of PBMCs from the two donors provided a titration of the HLA-B0702 CMV pp65 TPR responses in a B0702 negative donor sample. 5 fold dilutions of BC260 into BC262 were applied to generate seven samples with: 100, 20, 4, 0.8, 0.16, 0.032 and 0.0064% of cells derived from BC260. This corresponded to theoretical frequencies of HLA-B0702 CMV pp65 TPR specific T cells of 5%, 1%, 0.2%, 0.04%, 0.008%, 0.0016% and 0.00032%. The samples were in turn applied to evaluate the sensitivity of the Detection Molecules for detecting antigen-specific T cells in a sample. The relevance of the results obtained after applying the Detection Molecules could be evaluated by comparison of results obtained by conventional pMHC-multimer staining when applying a corresponding sample.
    • 2. Linker preparation: The linker used in this example was prepared as in example 1-2.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example were class I MHC-peptide complexes. The individual specificities (allele and peptide combination) were generated as described in example 1. A library of 110 different pMHCs, comprised of 6 different HLA-types, were generated, these are listed in table 9.
      • a. Synthesis: As described in example 1-2
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-2
    • 4. Label preparation: In this example Labels were synthetic oligonucleotides modified with biotin for coupling to the Linker. 110 different Labels from the 1OS system were applied (table 8).
      • a. Synthesis: As described in example 2
        • i. The Label system used in this example were named 1OS and comprised of single stranded oligonucleotides which were applied as a DNA barcodes.
      • b. Modification: As described in example 1-2.
      • c. Purification: As described in example 1-2.
    • 5. Detection Molecules preparation: 110 different Detection Molecules were generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (1OS DNA-barcodes) were attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Detection Molecules were essentially generated in the same way as described in example 2, with the difference that 110 different Detection Molecules were generated. The given combination of Label (DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are presented in table 9.
      • b. Modification: Since the total volume of 110 pooled Detection Molecules exceeded 100 μl this volume was reduced to reach a desired concentration of specific Binding Molecules. This was done as described in example 2
      • c. Purification: As described in example 1-2.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were mixed in one container to allow Detection Molecules to bind to T cells.
      • a. Amount of sample: Duplicates of seven samples each comprised of 2×10E6 cells in the form of BC's, i.e. 2×(1×100% BC260 and 6× five-fold dilutions of BC260 into BC262) (as described in 1.b)
      • b. Amount of Detection Molecule: As described in example 1
      • c. Conditions: BC260 and BC262 were thawed individually in 10 ml, 37° C., RPMI with 10% fetal bovine serum (FBS), centrifuged 5 min, 490 g, and washed twice in 10 ml RPMI with 10% FBS. All washing of cells refer to centrifugation 5 min, 490 g, with subsequent removal of supernatant. BC's were incubated individually in 50 nM dasatinib, 30 min, 37° C. and resuspended in 10 ml per 2×10E6 cells of the respective BC. Five-fold dilutions of BC260 were produced by adding 2.5 ml (0.5×10E6) of the former cell sample into 10 ml (2×10E6 cells) of the BC262 sample. The mixed cellular samples were washed in 200 ul barcode-buffer (PBS/0.5% BSA/2 mM EDTA/100 μg/ml herring DNA) and resuspended in this buffer to approximately 20 μl per sample prior to incubation of cells with Detection Molecules.
        • i. The Detection molecules were added in the required amount and samples were incubated as described in example 1-2.
    • 7. Enrichment of Detection Molecules with desired characteristics: The Detection Molecules were enriched for in the same way as described in example 1-2.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules were enriched based on specific binding of the Binding Molecule (the pMHC) to cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells would also reveal the pMHCs that had bound to cells in the PBMC sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, were amplified by PCR and identified by high-throughput sequencing.
      • a. Apply. The sorted cell sample which contained DNA-barcodes derived from the enriched Detection Molecules were amplified by PCR. See table 10 for composition of the PCR and table 11 for the thermal profile. The Taq PCR Master Mix Kit (Qiagen, #201443) was applied and PCR was run on the thermal cycler: GeneAmp, PCR System 9700 (Applied Biosystem). PCR products were visualized after gel electrophoresis on a Bio-Rad Gel Doc EZ Imager. DNA was sequenced using the Ion Torrent PGM platform (Life Technologies)
        • i. Primers were purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM were made in nuclease free water and stored at −20° C. The primers included adaptors for Ion Torrent sequencing, i.e. an A-key and a P1-key on the forward and reverse primer respectively. Additionally the forward primers had unique DNA sequences besides the primer region and the A-key (the primer sequences are listed in table 12). These primers were used to assign DNA-barcodes derived from the same sample with a sample-identification sequence (Sample-ID barcode) (see FIG. 9 for a schematic presentation of this design). This enabled distribution of DNA-barcode sequence reads according to their originating sample, when DNA-barcodes from multiple samples were sequenced in the same sequencing reaction. The non-enriched library of the 110 different Detection Molecules (diluted 100.000× after being reduced in volume) were also assigned with a sample-ID barcode through PCR (referred to as the Detection Molecule input). Information about the distribution of Labels within the library of Detection Molecules before enrichment would allow normalization of the sequence output. Pooled PCR products derived from the sample input and from multiple incubations of Detection Molecule and sample were purified with the MinElute PCR purification Kit (Qiagen, #28006) according to standard procedure.
        • ii. Purified DNA was sequenced by GeneDx (U.S.A) on an Ion Torrent PGM 314 chip.
      • b. Analysis. Positive sequence reads were aligned to sequences that read from the sample-barcode-identity at the 5′-end all the way through the DNA-barcode-identity. The number of reads was normalized according to the total number of reads that mapped to the same sample-ID barcode and according to the Detection Molecule input reads.
        • i. Mapping sequencing reads to 1OS DNA-barcodes: A sequence database was created consisting of the possible combinations of 15 sample-identification barcodes and 110 1OS DNA-barcodes together with the primer sequences from the 1OS system. This accumulated to 1650 sequences that could be expected from a sequencing run. Each sequencing read was then used to search the database for alignments, using the nucleotide BLAST algorithm, with a match reward of 1, mismatch reward of −2 and a gap cost of 2 for both opening and extending a gap. In this way sequencing errors were penalized equally, whether a base was miscalled or inserted/deleted in the sequencing read compared to the actual sequence. Alignments were discarded by the following criteria:
          • 1. E-value >1e-12; insufficient length of alignment (should be greater than 60 for the 1OS barcodes).
          • 2. Start position in subject sequence larger than 2, i.e. fewer than 5 out of 6 bases in the unique part of the sample-identification barcode was included in the alignment.
        • ii. If multiple alignments could still be found for any sequencing read, only the alignment with the best percent identity was kept. Finally, the number of reads mapping to each DNA barcode in the database was counted.
        • iii. Identifying overrepresented DNA barcodes: Relative read counts were calculated by normalizing each read to the total read count mapping to the same sample-ID barcode. The relative read counts were then used to calculate the fold change per DNA-barcode compared to the control DNA-barcode Detection Molecule input (the non-enriched Detection Molecule library). Significantly overrepresented DNA-barcodes were identified using a 2-sample test for equality of proportions on the raw read counts in a sample versus the DNA-barcode input-sample, and p-values were corrected for multiple testing using the Benjamini-Hochberg FDR method.

FACS sorting of fluorescent labeled cells, specific amplification of DNA-barcode Labels and high-throughput sequencing verified that it was possible to enrich and detect 1OS barcodes from a library of multiple different Detection molecules composed of 110 different 2OS DNA-barcode Labels encoding for 110 different antigen specificities distributed on 6 different HLA-types (FIG. 12). Moreover the number of sequence reads recovered from a given 1OS barcode was sensitive to the frequency of antigen specific T cells in the sample.

This example demonstrates that it is possible to detect antigen specific T cell responses of different frequencies in a panel of 110 different 1OS labeled Detection Molecules.

Example 4

In this example the binding molecules are class I pMHC complexes comprising 6 different HLA alleles, the linker is dextran-streptavidin-PE conjugate and the label is a DNA oligonucleotide.

The sample is HPBMC, the isolating and/or detecting is by FACS, and the determining of the identity of the label is by sequencing.

This example was essentially the same as example 3 only another Label system was applied.

This is an example where the Samples (1) were blood from one donor that were HLA-B0702:CMV pp65 TPR positive and another donor that were HLA-B0702 negative which were modified (1b) to generate Peripheral blood mononuclear cells (PBMCs). These samples were mixed in different ratios to generate new samples with different but known frequencies of T cells specific toward the HLA-B0702:CMV epitope. The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) were peptide-MHC (pMHC) complexes displaying one out of 110 different peptide-antigens comprised within 6 different HLA-types. The MHC molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules were purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T-cell populations.

The Labels (4) were oligonucleotides applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag. The labels were combined oligonucleotide labels arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label. The Detection Molecule (5) was synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. A library of 110 different Detection Molecules were generated wherein individual Binding Molecules, comprised of different pMHC, were encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, PBMC's (1b) was incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules were separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells were subjected to PCR for specific amplification of the DNA-barcode associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product revealed the identity of Detection Molecules that bound to T cells present in the sample.

This example thus revealed the presence of T cells in the blood expressing a T cell receptor that binds to pMHC molecules represented within the library of Detection Molecules. The number of sequencing reads mapped to a given DNA-barcode and its corresponding Binding Molecule would mirror the frequency of the T cells found by conventional MHC multimer stainings using the same Binding Molecule. Moreover it revealed that Labels, i.e. Detection Molecules, associated with T cells in a sample would be sufficiently enriched to reveal the presence of low frequent T cells binding such Detection Molecules.

    • 1. Sample preparation. The cell samples used in this example were obtained by preparing PBMC's from blood drawn from one donor that were HLA-B0702:CMV pp65 TPR positive and from another donor that were HLA-B0702 negative, as determined by conventional pMHC-multimer and antibody staining. They were acquired (a.) and modified (b.) in the same way as described in example 3.
    • 2. Linker preparation: The linker used in this example was prepared as in example 1-3.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example were class I MHC-peptide complexes. The individual specificities (allele and peptide combination) were generated as described in example 1-3. A library of 110 different pMHCs were generated, comprised of 6 different HLA-types, these are listed in table 9.
      • a. Synthesis: As described in example 1-3
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-3
    • 4. Label preparation: In this example Labels were synthetic oligonucleotides modified with biotin for coupling to the Linker. 110 different Labels from the 2OS system were applied (table 2).
      • a. Synthesis: As described in example 1.
        • i. The Label system used in this example was named 2OS and was developed to increase the complexity of a limited number of oligonucleotide sequences. This was enabled by applying a combinatorial strategy where two partially complementary oligonucleotides (an A oligonucleotide with a 5′ biotin tag and a B oligonucleotide) where annealed and then elongated to produce new unique oligonucleotide-sequences (AxBy) which were applied as a DNA-barcodes (Labels) (FIG. 9). By combining 6 unique oligonucleotide-sequences (A label precursor) that were all partly complementary to 20 other unique oligonucleotide sequences (B label precursor) a combinatorial library of 120 different (AxBy) Labels were produced. Only 110 of these Labels were used in this example (table 9).
      • b. Modification: As described in example 1-3.
      • c. Purification: As described in example 1-3.
    • 5. Detection Molecules preparation: 110 different Detection Molecules were generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (2OS DNA-barcodes) were attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Detection Molecules were essentially generated in the same way as described in example 3. The given combination of Label (2OS DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are presented in table 9.
      • b. Modification: Since the total volume of 110 pooled Detection Molecules exceeded 100 μl this volume was reduced to reach a desired concentration of specific Binding Molecules. This was done as described in example 2-3.
      • c. Purification: As described in example 1-3.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were mixed in one container to allow Detection Molecules to bind to T cells.
      • a. Amount of sample: Samples were equivalent to those used in example 3.
      • b. Amount of Detection Molecule: As described in example 1-3
      • c. Conditions: Samples and Detection Molecules were treated under the same conditions as described in example 3.
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules were enriched for in the same way as described in example 1-3.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules were enriched based on specific interaction of the Binding Molecule (the pMHC) with cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells would also reveal the pMHCs that had bound to cells in the PBMC sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, were amplified by PCR and identified by high-throughput sequencing.
      • a. Apply. The sorted cell sample which contained DNA-barcodes derived from the enriched Detection Molecules were amplified by PCR. See table 10 for composition of the PCR and table 11 for the thermal profile. The Taq PCR Master Mix Kit (Qiagen, #201443) was applied and PCR was run on the thermal cycler: GeneAmp, PCR System 9700 (Applied Biosystem). PCR products were visualized after gel electrophoresis on a Bio-Rad Gel Doc EZ Imager. DNA was sequenced using the Ion Torrent PGM platform (Life Technologies)
        • i. Primers were purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM were made in nuclease free water and stored at −20° C. The primers included adaptors for Ion Torrent sequencing, i.e. an A-key and a P1-key on the forward and reverse primer respectively. Additionally the forward primers had unique DNA sequences besides the primer region and the A-key. These primers were used to assign DNA-barcodes derived from the same sample with a sample-identification sequence (Sample-ID barcode) (the primer sequences are listed in table 13). This enabled distribution of DNA-barcode sequence reads according to their originating sample, when DNA-barcodes from multiple samples were sequenced in the same sequencing reaction. The non-enriched library of the 110 different Detection Molecules (diluted 100.000× after being reduced in volume) were also assigned with a sample-ID barcode through PCR (referred to as the Detection Molecule input)(see FIG. 9 for a schematic overview of the primer design). Information about the distribution of Labels within the library of Detection Molecules before enrichment would allow normalization of the sequence output. Pooled PCR products derived from the sample input and from multiple incubations of Detection Molecule and sample were purified with the MinElute PCR purification Kit (Qiagen, #28006) according to standard procedure.
        • ii. The purified DNA was sequenced by GeneDx (U.S.A) on an Ion Torrent PGM 314 chip.
      • b. Analysis. Positive sequence reads were aligned to sequences that read from the sample-barcode-identity at the 5′-end all the way through the DNA-barcode-identity. The number of reads was normalized according to the total number of reads that mapped to the same sample-ID barcode and according to the Detection Molecule input reads. The Analysis was essentially as in example 3 except that another database of 2OS sequences was generated and sequences were mapped according to corresponding 2OS DNA-barcodes.
        • i. Mapping sequencing reads to 2OS DNA-barcodes: A sequence database was created consisting of the possible combinations of 15 sample-identification barcodes and 120 2OS DNA barcodes (together with the primer and annealing sequences from the 2OS system). This accumulated to 1800 sequences that could be expected from a sequencing run. Each sequencing read was then used to search the database for alignments, using the nucleotide BLAST algorithm, with a match reward of 1, mismatch reward of −2 and a gap cost of 2 for both opening and extending a gap. In this way sequencing errors were penalized equally, whether a base was miscalled or inserted/deleted in the sequencing read compared to the actual sequence. Alignments were discarded by the following criteria:
          • 1. E-value >1e-12; insufficient length of alignment (should be greater than 102 for the 2OS barcodes).
          • 2. Start position in subject sequence larger than 2, i.e. fewer than 5 out of 6 bases in the unique part of the sample-identification barcode was included in the alignment.
        • ii. If multiple alignments could still be found for any sequencing read, only the alignment with the best percent identity was kept. Finally, the number of reads mapping to each DNA barcode in the database was counted.
        • iii. Identifying overrepresented DNA barcodes: Relative read counts were calculated by normalizing each read to the total read count mapping to the same sample-ID barcode. The relative read counts were then used to calculate the fold change per DNA-barcode compared to the control DNA-barcode Detection Molecule input (the non-enriched Detection Molecule library). Significantly overrepresented DNA-barcodes were identified using a 2-sample test for equality of proportions on the raw read counts in a sample versus the DNA-barcode input-sample, and p-values were corrected for multiple testing using the Benjamini-Hochberg FDR method.

FACS sorting of fluorescent labeled cells, specific amplification of DNA-barcode Labels and high-throughput sequencing verified that it was possible to enrich and detect 2OS barcodes from a library of multiple different Detection molecules composed of 110 different 2OS DNA-barcode Labels encoding for 110 different antigen specificities distributed on 6 different HLA-types (FIG. 13). Moreover the number of sequence reads recovered from a given 2OS barcode was sensitive to the frequency of antigen specific T cells in the sample, also indicating that it will be possible to detect Labels associated with responses of very low frequency (<0.002).

This example demonstrates that it is possible to detect antigen specific T cell responses of different and of low frequencies in a panel of 110 different 2OS labeled Detection Molecules.

Example 5

In this example the binding molecules are class I pMHCs, the linker is streptavidin conjugate and the label is DNA.

The isolating and/or detecting is by FACS, and the determining of the identity of the label is by sequencing.

This is an example where the Samples (1) were blood from six different donors with different HLA-types which were modified (1b) to generate Peripheral blood mononuclear cells (PBMCs).

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) were peptide-MHC (pMHC) complexes displaying one out of 110 different peptide-antigens comprised within 6 different HLA-types. The MHC molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules were purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T-cell populations.

The Labels (4) were single stranded oligonucleotide applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag.

The Detection Molecule (5) was synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. A library of 110 different Detection Molecules were generated wherein individual Binding Molecules, comprised of different pMHC, were encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, PBMC's (1b) was incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules were separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells were subjected to PCR for specific amplification of the DNA-barcode associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product revealed the identity of Detection Molecules that bound to T cells present in the sample.

This example thus revealed the presence of T cells in the blood expressing a T cell receptor that binds to pMHC molecules represented within the library of Detection Molecules. The application of multiple (110) Detection Molecules in one sample enabled detection of T cells with different T cell receptor specificities in parallel.

    • 1. Sample preparation. The cell samples used in this example were obtained by preparing PBMC's from blood drawn from six different donors with a number of different peptide-antigen responsive T cells, as determined by conventional pMHC-multimer staining.
      • a. Acquiring sample: Blood was obtained from the Danish Blood Bank, as example 1-4
      • b. Modifying sample: PBMCs were isolated from whole blood as described in example 1-4.
    • 2. Linker preparation: The linker used in this example was prepared as in example 1-4.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example were class I MHC-peptide complexes. The individual specificities (allele and peptide combination) were generated as described in example 1. A library of 110 different pMHCs, comprised of 6 different HLA-types, were generated, these are listed in table 9.
      • a. Synthesis: As described in example 1-4
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-4
    • 4. Label preparation: In this example Labels were synthetic oligonucleotides modified with biotin for coupling to the Linker. 110 different Labels from the 1OS system were applied (table 8).
      • a. Synthesis: As described in example 2-3.
        • i. The Label system used in this example were named 1OS and comprised of single stranded oligonucleotides which were applied as a DNA barcodes.
      • b. Modification: As described in example 1-4.
      • c. Purification: As described in example 1-4.
    • 5. Detection Molecules preparation: 110 different Detection Molecules were generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (1OS DNA-barcodes) were attached to the Linker (dextran-streptavidin-PE conjugate) to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Detection Molecules were generated in the same way as described in example 3. The given combination of Label (DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are presented in table 9.
      • b. Modification: Since the total volume of 110 pooled Detection Molecules exceeded 100 μl this volume was reduced to reach a desired concentration of specific Binding Molecules. This was done as described in example 2-4.
      • c. Purification: As described in example 1-4.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were incubated in the same way as described in example 1-2.
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules were enriched for in the same way as described in example 1-6.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules were enriched based on specific interaction of the Binding Molecule (the pMHC) with cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells would also reveal the pMHCs that had bound to cells in the PBMC sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, were amplified by PCR and identified by high-throughput sequencing. The enriched Labels, of the 1OS DNA-barcode system, were identified as in example 3.

FACS sorting of fluorescent labeled cells, specific amplification of DNA-barcode Labels and high-throughput sequencing verified that it was possible to enrich and detect 1OS barcodes from a library of multiple different Detection molecules composed of 110 different 1OS DNA-barcode Labels encoding for 110 different antigen specificities distributed on 6 different HLA-types (FIG. 14). It was verified that several DNA-barcodes, encoding different antigen specificities on different HLA-types, could be enriched for and detected in parallel, indicative for the presence of multiple antigen-specific T cell responses in that sample.

This example demonstrates that it is possible to detect several antigen-specific T cell responses in parallel when applying a library of Detection molecules of increasing complexity.

Example 6

In this example 110 different binding molecules are used, the linker is dextran for all of the detection molecules and 110 different labels are used.

This example was essentially the same as example 5 only another Label system was applied.

This is an example where the Samples (1) were blood from six different donors with different HLA-types which were modified (1b) to generate Peripheral blood mononuclear cells (PBMCs).

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) were peptide-MHC (pMHC) complexes displaying one out of 110 different peptide-antigens comprised within 6 different HLA-types. The MHC molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules were purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T cell populations.

The Labels (4) were oligonucleotides applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag. The labels were combined oligonucleotide labels arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label. The Detection Molecule (5) was synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. A library of 110 different Detection Molecules were generated wherein individual Binding Molecules, comprised of different pMHC, were encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, PBMC's (1b) was incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules were separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells were subjected to PCR for specific amplification of the DNA-barcode associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product revealed the identity of Detection Molecules that bound to T cells present in the sample.

This example thus revealed the presence of T cells in the blood expressing a T cell receptor that binds to pMHC molecules represented within the library of Detection Molecules. The application of multiple (110) Detection Molecules in one sample enabled detection of T cells with different T cell receptor specificities in parallel.

    • 1. Sample preparation. The cell samples used in this example were obtained by preparing PBMC's from blood drawn from six different donors with a number of different peptide-antigen responsive T cells, as determined by conventional pMHC-multimer.
      • a. Acquiring sample: Blood was obtained from the Danish Blood Bank.
      • b. Modifying sample: PBMCs were isolated from whole blood as described in example 1-2, 5.
    • 2. Linker preparation: The linker used in this example was prepared as in example 1-5.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example were class I MHC-peptide complexes. The individual specificities (allele and peptide combination) were generated as described in example 1-5. A library of 110 different pMHCs, comprised of 6 different HLA-types, were generated, these are listed in table 9.
      • a. Synthesis: As described in example 1-5.
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-5
    • 4. Label preparation: In this example Labels were synthetic oligonucleotides modified with biotin for coupling to the Linker. 110 different Labels from the 2OS system were applied (table 2).
      • a. Synthesis: As described in example 1, 4.
        • i. The Label system used in this example was named 2OS and was developed to increase the complexity of a limited number of oligonucleotide sequences. This was enabled by applying a combinatorial strategy where two partially complementary oligonucleotides (an A oligonucleotide with a 5′ biotin tag and a B oligonucleotide) where annealed and then elongated to produce new unique oligonucleotide-sequences (AxBy) which were applied as a molecular barcodes (Labels) (FIG. 9). By combining 6 unique oligonucleotide-sequences (A label precursor) that were all partly complementary to 20 other unique oligonucleotide sequences (B label precursor) a combinatorial library of 120 different (AxBy) Labels were produced. Only 110 of these Labels were used in this example (table 9).
      • b. Modification: As described in example 1-5.
      • c. Purification: As described in example 1-5.
    • 5. Detection Molecules preparation: 110 different Detection Molecules were generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (2OS DNA-barcodes) were attached to the Linker (dextran-streptavidin-PE conjugate) to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Detection Molecules were generated in the same way as described in example 3, 4, 5. The given combination of Label (2OS DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are presented in table 9.
      • b. Modification: Since the total volume of 110 pooled Detection Molecules exceeded 100 μl this volume was reduced to reach a desired concentration of specific Binding Molecules (done as described in example 2-5).
      • c. Purification: As described in example 1-5.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were incubated in the same way as described in example 1-2, 5.
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules were enriched for in the same way as described in example 1-5.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules were enriched based on specific binding of the Binding Molecule (the pMHC) to cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells would also reveal the pMHCs that had bound to cells in the PBMC sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, were amplified by PCR and identified by high-throughput sequencing. The enriched Labels, of the 2OS DNA-barcode system, were identified as in example 4.

FACS sorting of fluorescent labeled cells, specific amplification of DNA-barcode Labels and high-throughput sequencing verified that it was possible to enrich and detect 2OS barcodes from a library of multiple different Detection molecules composed of 110 different 2OS DNA-barcode Labels encoding for 110 different antigen specificities distributed on 6 different HLA-types (FIG. 15). It was verified that several DNA-barcodes, encoding different antigen specificities of different HLA-types, could be enriched for and detected in parallel, indicative for the presence of multiple antigen-specific T cell responses in that sample.

This example demonstrates that it is possible to detect several antigen-specific T cell responses in parallel when applying a library of Detection molecules of increasing complexity.

Example 7

In this example 175 different pMHC complexes are used as binding molecules. The sample is tumor infiltrating Lymphocytes from a resected tumor lesion of a human being.

This is an example where the Samples (1) were resected tumor lesions from 11 HLA-A0201 positive patients with malignant melanoma. The samples were modified (1b) to generate Tumor Infiltrating Lymphocytes (TILs).

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) were peptide-MHC (pMHC) complexes displaying one out of 175 different peptide-antigens. The MHC molecules were modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules were purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T-cell populations.

The Labels (4) were oligonucleotides applied as DNA-barcodes. The oligonucleotides were synthetized (4a) by DNA Technology A/S (Denmark) and were synthetically modified (4b) with a terminal biotin capture-tag. The labels were combined oligonucleotide labels arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label. The Detection Molecule (5) was synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contained a modification (5b) in the form of a fluorochrome. A library of 175 different Detection Molecules were generated wherein individual Binding Molecules, comprised of different pMHC, were encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, TILs (1b), was incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules were separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the TIL's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the TIL sample.

FACS isolated cells were subjected to PCR for specific amplification of the DNA-barcode Label associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product revealed the identity of Detection Molecules that bound to T cells present in the sample.

This example thus revealed the presence of T cells among the TIL's expressing a T cell receptor that binds to pMHC molecules represented within the library of Detection Molecules. The number of sequencing reads mapped to a given DNA-barcode and its corresponding Binding Molecule would mirror the frequency of the T cells found by conventional MHC multimer stainings using the same Binding Molecules. The application of multiple (175) Detection Molecules in one sample enabled detection of T cells with different T cell receptor specificities in parallel. This is feasible also when the sample, as in present example is TILs that on average basis has a lower avidity of their TCR to their pMHC antigen. Consequently they are more challenging to detect than virus-responsive T cells using traditional fluorescence labelled MHC mulitimers.

    • 1. Sample preparation. The 11 cell samples used in this example were obtained from resected tumor lesions obtained from patients with malignant melanoma.
      • a. Acquiring sample. TILs were derived from tumor fragments from melanoma patients. Tumor lesion were resected following surgical removal of the given tumor lesion.
      • b. Modifying sample. Tumor fragments (1-3 mm3) were cultured individually in complete medium (RPMI with 10% human serum, 100 U/ml penicillin, 100 μg/ml streptomycin, 1.25 μg/ml fungizone [Bristol-Myers Squibb] and 6000 U/ml IL-2) at 37° C. and 5% CO2, allowing TILs to migrate to the medium. TILs were expanded to reach >50×106 total cells originated from approximately 24 fragments, which had expanded to confluent growth in 2-ml wells and eliminated adherent tumor cells (average of approximately 2×106 cells per well from each tumor fragment). TIL cultures were expanded on a clinical scale using a standard rapid expansion protocol (REP). Briefly, TILs were stimulated with 30 ng/ml anti-CD3 antibody (OKT-3; Ortho Biotech) and 6000 U/ml IL-2 in the presence of irradiated (40 Gy) allogenic feeder cells (PBMCs from a healthy donor) at a feeder:TIL ratio of 200:1. Initially, TILs were rapidly expanded in a 1:1 mix of complete medium and REP medium (AIM-V [Invitrogen] with 10% human serum, 1.25 μg/ml fungizone and 6000 U/ml IL-2), but after seven days complete medium and serum were removed stepwise from the culture by adding REP medium without serum to maintain cell densities around 1-2×106 cells/ml. TIL cultures were cryopreserved at −150° C. in human serum containing 10% DMSO. All patients were HLA-A0201 positive.
    • 2. Linker preparation: The linker used in this example was prepared as in example 1-6.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example were class I MHC-peptide complexes (peptide-HLA-A0201). The individual specificities (allele and peptide combination) were generated as described in example 1-6. A library of 175 different pMHCs were generated, these are listed in table 2.
      • a. Synthesis: As described in example 1-6
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-6
    • 4. Label preparation: In this example Labels were synthetic oligonucleotides modified with biotin for coupling to the Linker. 175 different Labels from the 2OS system were applied (table 14).
      • a. Synthesis: As described in example 1.
        • i. The Label system used in this example was named 2OS and was developed to increase the complexity of a limited number of oligonucleotide sequences. This was enabled by applying a combinatorial strategy where two partially complementary oligonucleotides (an A oligonucleotide with a 5′ biotin tag and a B oligonucleotide) where annealed and then elongated to produce new unique oligonucleotide-sequences (AxBy) which were applied as a DNA-barcodes (Labels) (FIG. 9). By combining 22 unique oligonucleotide-sequences (A label precursor) that are all partly complementary to 55 other unique oligonucleotide sequences (B label precursor) a combinatorial library of 1,210 different (AxBy) Labels were produced. Only 175 of these Labels were used in this example (table 14). Refer to table 2 for an overview of the different 2OS A and B nucleotide sequences.
      • b. Modification: As described in example 1-6.
      • c. Purification: As described in example 1-6.
    • 5. Detection Molecules preparation: 175 different Detection Molecules were generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (2OS DNA-barcodes) were attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC was always attached to a given DNA-barcode.
      • a. Detection Molecules were essentially generated in the same way as described in example 1-6. The given combination of Label (2OS DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are presented in table 14.
      • b. Modification: Since the total volume of 175 pooled Detection Molecules exceeded 100 μl this volume was reduced to reach a desired concentration of specific Binding Molecules. This was done as described in example 2-6.
      • c. Purification: As described in example 1-6.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules were mixed in one container to allow Detection Molecules to bind to T cells.
      • a. Amount of sample: Between 0.2×10E6-2.3×10E6 cells in the form of TILs, were used.
      • b. Amount of Detection Molecule: As described in example 1-6
      • c. Conditions: Samples and Detection Molecules were treated as described in example 1-2 and 5-6.
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules were enriched for in the same way as described in example 1-6.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules were enriched based on specific interaction of the Binding Molecule (the pMHC) with cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells would also reveal the pMHCs that had bound to cells in the TIL sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, were amplified by PCR and identified by high-throughput sequencing.
      • a. Apply. The sorted cell samples which contained DNA-barcodes derived from the enriched Detection Molecules were amplified by PCR. See table 10 for composition of the PCR and table 11 for the thermal profile. The Taq PCR Master Mix Kit (Qiagen, #201443) was applied and PCR was run on the thermal cycler: GeneAmp, PCR System 9700 (Applied Biosystem). PCR products were visualized after gel electrophoresis on a Bio-Rad Gel Doc EZ Imager. DNA was sequenced using the Ion Torrent PGM platform (Life Technologies)
        • i. Primers were purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM were made in nuclease free water and stored at −20° C. The primers included adaptors for Ion Torrent sequencing, i.e. an A-key and a P1-key on the forward and reverse primer respectively. Additionally the forward primers had unique DNA sequences besides the primer region and the A-key. These primers were used to assign DNA-barcodes derived from the same sample with a sample-identification sequence (Sample-ID barcode) (refer to table 13 for primer sequences). This enabled distribution of DNA-barcode sequence reads according to their originating sample, when DNA-barcodes from multiple samples were sequenced in the same sequencing reaction (see FIG. 9 for a schematic presentation of the primer design). If >10.000 cells were sorted in the enrichment step, only a volume corresponding to 10.000 cells were applied as template in the PCR. To further examine the potential impact on the number of cells selected with associated Detection molecules, we included Enriched Detection Molecules derived from one sample (sample 8) multiple times, so that triplicate PCRs were run with equivalent to 10.000 sorted cells and 100 sorted cells respectively. Moreover a single PCR was run with 1000 cells from the same sample. The non-enriched library of the 175 different Detection Molecules (diluted 10.000x after being reduced in volume) were also assigned with a sample-ID barcode through PCR (referred to as the Detection Molecule-input). Triplicate PCRs were run with the Detection Molecule-input. Information about the distribution of Labels within the library of Detection Molecules before enrichment would allow normalization of the sequence output. Pooled PCR products derived from the Detection Molecule-input and from multiple incubations of Detection Molecule and sample were separated according to size, by gel-electrophoresis, and the excised gel-fragment containing DNA-fragments of ˜200 bp were purified with the QIAquick PCR Purification Kit (Qiagen, #28104) according to standard procedure.
        • ii. The purified DNA was sequenced by Amplexa (Denmark) on an Ion Torrent PGM 314 chip.
      • b. Analysis. Positive sequence reads were aligned to sequences that read from the sample-barcode-identity at the 5′-end all the way through the DNA-barcode-identity. The number of reads was normalized according to the total number of reads that mapped to the same sample-ID barcode and according to the Detection Molecule input reads. The Analysis was essentially as in example 4 and 6 except that the database of 2OS sequences was expanded to include the new 175 Labels and sequences were mapped according to corresponding 2OS DNA-barcodes.
        • i. Mapping sequencing reads to 2OS DNA-barcodes: A sequence database was created consisting of the possible combinations of 20 sample-identification barcodes and 192 2OS DNA barcodes (together with the primer and annealing sequences from the 2OS system). This accumulated to 3840 sequences that could be expected from a sequencing run. Each sequencing read was then used to search the database for alignments, using the nucleotide BLAST algorithm, with a match reward of 1, mismatch reward of −2 and a gap cost of 2 for both opening and extending a gap. In this way sequencing errors were penalized equally, whether a base was miscalled or inserted/deleted in the sequencing read compared to the actual sequence. Alignments were discarded by the following criteria:
          • 1. E-value >1e-12; insufficient length of alignment (should be greater than 102 for the 2OS barcodes).
          • 2. Start position in subject sequence larger than 2, i.e. fewer than 5 out of 6 bases in the unique part of the sample-identification barcode was included in the alignment.
        • ii. If multiple alignments could still be found for any sequencing read, only the alignment with the best percent identity was kept. Finally, the number of reads mapping to each DNA barcode in the database was counted.
        • iii. Identifying overrepresented DNA barcodes: Two amendments of the analysis performed in example 4 and 6 (as well as 3 and 5 in respect to the 1OS label system) were introduced:
          • a. The N6 sequence (FIG. 9) were applied to reduce the clonality of the amplification product to avoid potential biases derived from amplification of enriched 2OS sequence Labels.
          •  i. Any repetitive reads that would share a N6 and map to a given sample-ID all through the 2OS barcode region were virtually removed ensuring that this sequence was only accounted for once.
          • b. The sequencing data was reanalyzed after virtual removal of the most high-frequent sequences. This ensured that potential biases derived from high-percentage enrichment of a given Detection Molecule, resulting in large read counts from the associated 2OS sequence Label, would not mask the presence of lower percentages yet enriched Detection Molecules.
        • Collectively this strategy is termed clonality reduction. Relative read counts were calculated by normalizing each read to the total read count mapping to the same sample-ID barcode. The relative read counts were then used to calculate the fold change per DNA-barcode compared to the control DNA-barcode Detection Molecule input (the non-enriched Detection Molecule library). Significantly overrepresented DNA-barcodes were identified using a 2-sample test for equality of proportions on the raw read counts in a sample versus the DNA-barcode input-sample, and p-values were corrected for multiple testing using the Benjamini-Hochberg FDR method. The algorithm applied for this analysis is available at: www.cbs.dtu.dk/services/Barracoda/

FACS sorting of fluorescent labeled cells, specific amplification of DNA-barcode Labels and high-throughput sequencing verified that it was possible to enrich and detect 2OS barcodes from a library of multiple different Detection molecules composed of 175 different 2OS DNA-barcode Labels encoding for 175 different cancer-specificities (FIG. 16). It implied that several DNA-barcodes, encoding different antigen specificities, could be enriched for and detected in parallel, also when the samples, as in the present example, were TILs that on average has a lower avidity of their TCR to their pMHC antigen. The results indicated that the presence of multiple cancer-specific T cell responses could be detected in parallel in a sample.

This example implies that it is possible to detect several cancer-specific T cell responses in parallel when applying a library of Detection molecules of increasing complexity.

Example 8

In this example 1025 different binding molecules are used in the form of pMHC complexes (each with DNA labels).

The sample is a mixture of two different blood samples, the isolating and/or detecting is done by flow cytometry and the determining of the identity of the label is done by sequencing the DNA label.

This example is essentially the same as example 4 but will apply a larger Detection Molecule library (1025) and will include a greater number of different HLA-types (11) to analyze the same samples.

This is an example where the Samples (1) are blood from one donor that is HLA-B0702:CMV pp65 TPR positive and another donor that is HLA-B0702 negative which are modified (1b) to generate Peripheral blood mononuclear cells (PBMCs). These samples are mixed in different ratios to generate new samples with different but known frequencies of T cells specific toward the HLA-B0702:CMV epitope.

The Linker (2) is a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) are peptide-MHC (pMHC) complexes displaying one out of 1025 different peptide-antigens. The MHC molecules are modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules are purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T-cell populations.

The Labels (4) are oligonucleotides applied as DNA-barcodes. The oligonucleotides are synthetized (4a) by DNA Technology A/S (Denmark) and are synthetically modified (4b) with a terminal biotin capture-tag. The labels are combined oligonucleotide labels arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label.

The Detection Molecule (5) is synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contains a modification (5b) in the form of a fluorochrome. A library of 1025 different Detection Molecules are generated wherein individual Binding Molecules, comprised of different pMHC, are encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, PBMC's (1b) is incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules are separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells are subjected to PCR for specific amplification of the DNA-barcode associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product will reveal the identity of Detection Molecules that binds to T cells present in the sample.

This example will reveal the presence of T cells in the blood expressing a T cell receptor that binds to pMHC molecules represented within the library of 1025 Detection Molecules. The number of sequencing reads that will map to a given DNA-barcode and its corresponding Binding Molecule will mirror the frequency of the T cells found by conventional MHC multimer stainings using the same Binding Molecules. The increased complexity of the Detection Molecule library, in terms of the number of different Binding Molecules and Labels, will reflect positively upon the sensitivity for detecting a given Label, i.e. a Detection Molecule, associated with low frequencies of T cells binding such Detection Molecules.

    • 1. Sample preparation. The cell samples used in this example are obtained by preparing PBMC's from blood drawn from one donor that is HLA-B0702:CMV pp65 TPR positive and from another donor that is HLA-B0702 negative, as determined by conventional pMHC-multimer and antibody staining. They are acquired (a.) and modified (b.) in the same way as described in example 3-4.
    • 2. Linker preparation: The linker used in this example is prepared as in example 1-7.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example are class I MHC-peptide complexes. The individual specificities (allele and peptide combination) are generated as described in example 1-7. A library of 1025 different pMHCs are generated including 11 different HLA-types.
      • a. Synthesis: As described in example 1-7
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-7
    • 4. Label preparation: In this example Labels are synthetic oligonucleotides modified with biotin for coupling to the Linker. 1025 different Labels from the 2OS system were applied (table 2).
      • a. Synthesis: As described in example 1.
        • i. The Label system used in this example is named 2OS and was developed to increase the complexity of a limited number of oligonucleotide sequences. This is enabled by applying a combinatorial strategy where two partially complementary oligonucleotides (an A oligonucleotide with a 5′ biotin tag and a B oligonucleotide) are annealed and then elongated to produce new unique oligonucleotide-sequences (AxBy) which are applied as a DNA-barcodes (Labels) (FIG. 2). By combining 22 unique oligonucleotide-sequences (A label precursor) that are all partly complementary to 55 other unique oligonucleotide sequences (B label precursor) a combinatorial library of 1210 different (AxBy) Labels are produced. Only 1025 of these Labels are used in this example.
      • b. Modification: As described in example 1-7.
      • c. Purification: As described in example 1-7.
    • 5. Detection Molecules preparation: 1025 different Detection Molecules are generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (2OS DNA-barcodes) are attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC is always attached to a given DNA-barcode.
      • a. Detection Molecules are essentially generated in the same way as described in example 1-7. The given combination of Label (2OS DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are registered.
      • b. Modification: Since the total volume of 1025 pooled Detection Molecules exceeds 100 μl this volume is reduced to reach a desired concentration of specific Binding Molecules. This is done as described in example 2-7.
      • c. Purification: As described in example 1-7.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules are mixed in one container to allow Detection Molecules to bind to T cells.
      • a. Amount of sample: Samples are equivalent to those used in example 3-4.
      • b. Amount of Detection Molecule: As described in example 1-7
      • c. Conditions: Samples and Detection Molecules are treated under the same conditions as described in example 3-4.
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules are enriched for in the same way as described in example 1-7.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules are enriched based on specific interaction of the Binding Molecule (the pMHC) with cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells will also reveal the pMHCs that bind to cells in the PBMC sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, are amplified by PCR and identified by high-throughput sequencing.
      • a. Apply. The sorted cell samples which contain DNA-barcodes derived from the enriched Detection Molecules are amplified by PCR. See table 10 for composition of the PCR and table 11 for the thermal profile. The Taq PCR Master Mix Kit (Qiagen, #201443) is applied and PCR is run on the thermal cycler: GeneAmp, PCR System 9700 (Applied Biosystem). PCR products are visualized after gel electrophoresis on a Bio-Rad Gel Doc EZ Imager. DNA is sequenced using the Ion Torrent PGM platform (Life Technologies)
        • i. Primers are purchased from DNA Technology (Denmark) and delivered as lyophilized powder. Stock dilutions of 100 μM are made in nuclease free water and stored at −20° C. The primers include adaptors for Ion Torrent sequencing, i.e. an A-key and a P1-key on the forward and reverse primer respectively. Additionally the forward primers have unique DNA sequences besides the primer region and the A-key. These primers are used to assign DNA-barcodes derived from the same sample with a sample-identification sequence (Sample-ID barcode) (refer to table 13 for primer sequences). This enables distribution of DNA-barcode sequence reads according to their originating sample, when DNA-barcodes from multiple samples are sequenced in the same sequencing reaction (see FIG. 9 for a schematic presentation of the primer design). The non-enriched library of the 1025 different Detection Molecules (diluted 100.000× after being reduced in volume) are also assigned with a sample-ID barcode through PCR (referred to as the Detection Molecule input). Triplicate PCRs are run with the Detection Molecule-input. Information about the distribution of Labels within the library of Detection Molecules before enrichment will allow normalization of the sequence output. Pooled PCR products derived from the sample input and from multiple incubations of Detection Molecule and sample are separated according to size, by gel-electrophoresis, and the excised gel-fragment containing DNA-fragments of ˜200 bp are purified with the QIAquick PCR Purification Kit (Qiagen, #28104) according to standard procedure.
        • ii. The purified DNA was sequenced on an Ion Torrent PGM 314 chip.
      • b. Analysis. Positive sequence reads are aligned to sequences that read from the sample-barcode-identity at the 5′-end all the way through the DNA-barcode-identity. The number of reads is normalized according to the total number of reads that maps to the same sample-ID barcode and according to the Detection Molecule input reads. The Analysis is essentially as in example 7 except that the database of 2OS sequences now includes 1025 Labels and sequences are mapped according to corresponding 2OS DNA-barcodes.
        • i. Mapping sequencing reads to 2OS DNA-barcodes: A sequence database is created consisting of the possible combinations of 10 sample-identification barcodes and 1025 2OS DNA barcodes (together with the primer and annealing sequences from the 2OS system). This accumulates to 10250 sequences that can be expected from a sequencing run. Each sequencing read is then used to search the database for alignments, using the nucleotide BLAST algorithm, with a match reward of 1, mismatch reward of −2 and a gap cost of 2 for both opening and extending a gap. In this way sequencing errors are penalized equally, whether a base was miscalled or inserted/deleted in the sequencing read compared to the actual sequence. Alignments are discarded by the following criteria:
          • 1. E-value >1e-12; insufficient length of alignment (should be greater than 102 for the 2OS barcodes).
          • 2. Start position in subject sequence larger than 2, i.e. fewer than 5 out of 6 bases in the unique part of the sample-identification barcode was included in the alignment.
        • If multiple alignments can still be found for any sequencing read, only the alignment with the best percent identity is kept. Finally, the number of reads mapping to each DNA barcode in the database is counted.
        • ii. Identifying overrepresented DNA barcodes: Is essentially performed as described in example 7 with the strategy of applying clonality reduction.
          • Relative read counts are calculated by normalizing each read to the total read count mapping to the same sample-ID barcode. The relative read counts are then used to calculate the fold change per DNA-barcode compared to the control DNA-barcode Detection Molecule input (the non-enriched Detection Molecule library). Significantly overrepresented DNA-barcodes are identified using a 2-sample test for equality of proportions on the raw read counts in a sample versus the DNA-barcode input-sample, and p-values are corrected for multiple testing using the Benjamini-Hochberg FDR method. The algorithm applied for this analysis is available at: www.cbs.dtu.dk/services/Barracoda/

The expected outcome of this example is knowledge on the sensitivity for detecting antigen-specific T cell responses of decreasing frequency (<0.002% of CD8 T cells) in a number of similar samples. The sensitivity will expectantly increase with increasing numbers of Detection molecules incubated with a given sample, because any sequencing reads that are caused by background will be distributed on a greater number of Labels, in this example 1025.

Example 9

In this example 1025 different pMHC complexes, comprising 11 different HLA-alleles, are used as binding molecules. The sample is a mixture of blood from 10 different donors.

This example is essentially the same as example 6 but a greater number of samples (10) will be analyzed with a larger Detection Molecule library (1025), which will include a greater number of different HLA-types (11).

This is an example where the Samples (1) are blood from 10 different donors with different HLA-types which are modified (1b) to generate Peripheral blood mononuclear cells (PBMCs).

The Linker (2) is a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) are peptide-MHC (pMHC) complexes displaying one out of 1025 different peptide-antigens. The MHC molecules are modified (3b) by biotinylation to provide a biotin capture-tag for the Linker. The binding molecules are purified (2c) by HPLC and quality controlled in terms of the formation of functional pMHC multimers for staining of control T-cell populations.

The Labels (4) are oligonucleotides applied as DNA-barcodes. The oligonucleotides are synthetized (4a) by DNA Technology A/S (Denmark) and are synthetically modified (4b) with a terminal biotin capture-tag. The labels are combined oligonucleotide labels arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label.

The Detection Molecule (5) is synthetized (5a) by attaching Binding Molecules in the form of biotinylated pMHC and Labels in the form of biotin-modified oligonucleotides (DNA-barcodes) onto a streptavidin-modified dextran linker. The detection molecule further contains a modification (5b) in the form of a fluorochrome. A library of 1025 different Detection Molecules are generated wherein individual Binding Molecules, comprised of different pMHC, are encoded for by corresponding individual Labels, comprised of different DNA-barcodes.

An amount of sample, PBMC's (1b) is incubated with an amount of mixed Detection Molecules (5) under conditions (6c) allowing binding of Detection Molecules to T cells in the sample.

The cell-bound Detection Molecules are separated from the non-cell bound Detection Molecules (7) by first a few rounds of washing the PBMC's through centrifugation sedimentation of cells and resuspension in wash buffer followed by Fluorescence Activated Cell Sorting (FACS) of fluorochrome labeled cells. T cells that can efficiently bind Detection Molecules will fluoresce because of the fluorochrome comprised within the detection molecules; T cells that cannot bind detection molecules will not fluoresce. FACS-sorting leads to enrichment of fluorescent cells, and hence, enrichment of the detection molecules along with the associated labels that bind T cells of the PBMC sample.

FACS isolated cells are subjected to PCR for specific amplification of the DNA-barcode associated with the Detection Molecules bound to the isolated cells. High throughput sequencing of the resultant PCR product will reveal the identity of Detection Molecules that binds to T cells present in the sample.

This example will reveal the presence of T cells in the blood expressing a T cell receptor that binds to pMHC molecules represented within the library of 1025 Detection Molecules. The number of sequencing reads that will map to a given DNA-barcode and its corresponding Binding Molecule will mirror the frequency of the T cells found by conventional MHC multimer stainings using the same Binding Molecules. The increased complexity of the Detection Molecule library, in terms of the number of different Binding Molecules and Labels, will enable detection of T cells of 1025 different T cell receptor specificities in parallel and will reflect positively upon the sensitivity for detecting a given Label, i.e. a Detection Molecule, associated with low frequencies of T cells binding such Detection Molecules.

    • 1. Sample preparation. The cell samples used in this example are obtained by preparing PBMC's from blood drawn from 10 different donors with a number of different peptide-antigen responsive T cells, as determined by conventional pMHC-multimer.
      • a. Acquiring sample: Blood is obtained from the Danish Blood Bank.
      • b. Modifying sample: PBMCs are isolated from whole blood as described in example 1-2, 5-6.
    • 2. Linker preparation: The linker used in this example is prepared as in example 1-7 and example 8.
    • 3. Binding Molecules preparation: The Binding Molecules used in this example are class I MHC-peptide complexes. The individual specificities (allele and peptide combination) are generated as described in example 1-7 and example 8. A library of 1025 different pMHCs are generated including 10 different HLA-types.
      • a. Synthesis: As described in example 1-7 and example 8.
      • b. Modification: No further modifications
      • c. Purification: As described in example 1-7 and example 8.
    • 4. Label preparation: In this example Labels are synthetic oligonucleotides modified with biotin for coupling to the Linker. 1025 different Labels from the 2OS system were applied (table 2).
      • a. Synthesis: As described in example 1.
        • i. The Label system used in this example is named 2OS and was developed to increase the complexity of a limited number of oligonucleotide sequences. This is enabled by applying a combinatorial strategy where two partially complementary oligonucleotides (an A oligonucleotide with a 5′ biotin tag and a B oligonucleotide) are annealed and then elongated to produce new unique oligonucleotide-sequences (AxBy) which are applied as a DNA-barcodes (Labels) (FIG. 9). By combining 22 unique oligonucleotide-sequences (A label precursor) that are all partly complementary to 55 other unique oligonucleotide sequences (B label precursor) a combinatorial library of 1210 different (AxBy) Labels are produced. Only 1025 of these Labels are used in this example.
      • b. Modification: As described in example 1-7 and example 8.
      • c. Purification: As described in example 1-7 and example 8.
    • 5. Detection Molecules preparation: 1025 different Detection Molecules are generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (2OS DNA-barcodes) are attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC is always attached to a given DNA-barcode.
      • a. Detection Molecules are essentially generated in the same way as described in example 1-7 and example 8. The given combination of Label (2OS DNA-barcode) and Binding Molecule (pMHC) of each Detection Molecule are registered.
      • b. Modification: Since the total volume of 1025 pooled Detection Molecules exceeds 100 μl this volume is reduced to reach a desired concentration of specific Binding Molecules. This is done as described in example 2-7 and example 8.
      • c. Purification: As described in example 1-7 and example 8.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules are incubated in the same way as described in example 1-2, 5-7. The cell sample and the Detection Molecules are mixed in one container to allow Detection Molecules to bind to T cells.
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules are enriched for in the same way as described in example 1-7 and example 8.
    • 8. Identification of enriched Detection Molecules: Because the Detection Molecules are enriched based on specific interaction of the Binding Molecule (the pMHC) with cells, the identification of the associated Labels (DNA-barcodes) amongst the sorted cells will also reveal the pMHCs that bind to cells in the PBMC sample. In this example the DNA-barcodes associated with the enriched Detection Molecules, are amplified by PCR and identified by high-throughput sequencing. The enriched Labels, of the 2OS DNA-barcode system, are identified as in example 8.

The expected outcome of this example is knowledge of the potential complexity for detecting multiple antigen-specific T cell responses in parallel in a single sample. Since the sensitivity is expected to increase with increasing numbers of Detection molecules incubated with a given sample (as described in example 8) it is expected that multiple more T cell responses will also be detected in parallel when using a Detection molecule library of the said complexity. The example will thus prove that it is possible to detect multiple antigen-specific T cell responses in parallel when applying a Detection molecule library comprised of 1025 different 2OS Labels encoding 1025 different Binding molecules distributed on 11 HLA-types.

Example 10

In this example it is shown how multiple, single-cell analyses can be rapidly performed using the present invention. The isolating and/or detecting is done by FACS, leading to the identification of a T cell and its TCR, as well as the pMHC complex that recognizes the T cell by binding to the TCR.

This example will describe how the detection of Binding Molecules can be linked to the T cell receptor (TCR) sequence following single cell sorting of T-cells associated with Detection Molecules.

This is an example where the Sample (1) will be a mixture of T cells comprising TCRs of interest. This could e.g. be tumor infiltrating lymphocytes from a cancer patient. The Linker (2) is a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) is peptide-MHC (pMHC) complexes displaying a of library peptides that are potentially recognized by the T cells in the Sample. This could be a library of melanoma-associated peptides, as used in example 7, or it could be a library of personally-defined potential epitope sequences based on the characteristics of the given patient's tumor. This could e.g. be mutation-derived T cell epitopes and/or epitopes selected based on the expression pattern in the individual tumor. The Binding Molecules will be modified (3b) by biotinylation and quality controlled as described in example 1-6.

The Labels (4) are oligonucleotides as in example 7-9.

The detection molecule (5) will be synthetized (5a) and modified (5b) as in example 7-9.

An amount of Sample, Tumor infiltrating lymphocytes (1b) will be mixed with detection molecules (5) under conditions (6c) that allow binding of detection molecules to T cells in the sample.

Cells will be FACS sorted based on the attachment of Detection Molecules, transferred to a FLUIDIGM C1 unit (or similar device) for single-cell amplification of nucleotide labels and T cell receptor genes.

This example thus reveals the possibility to identify, on a single-cell level, both the antigen-specificity and the T cell receptor sequence. This being done in a mixture of multiple different Binding Molecules (potentially, but not exclusively, >1000). This technology provides a mean for high-throughput identification of TCRs combined with a description of the antigen specificity of the TCR.

    • 1. Sample preparation. The cell sample to be used in this example is a collection of T cells with a recognition profile of interest for determining the sequences of the TCRs associated with this recognition. The sample could e.g. be tumor infiltration lymphocytes from a cancer patient. As described in example 7.
      • c. Acquiring sample: As in example 7.
      • d. Modifying sample: As in example 7.
    • 2. Linker preparation: The linker used in this example is prepared as in example 1-10. The fluorochrome co-attachment is important for the selection process in the example.
    • 3. Binding molecule preparation: The binding molecules used in this example will be a collection of peptide-MHC molecules, designed to match the T cell reactivity in the sample. This could be a library of melanoma-associated peptides, as used in example 7, or it could be a library of personally-defined potential epitope sequences based on the characteristics of the given patient's tumor. This could e.g. be mutation-derived T cell epitopes and/or epitopes selected based on the expression pattern if the individual tumor.
      • a. Synthesis: as in example 7-9.
      • b. Modification: No further modifications
      • c. Purification: as in example 7-9.
    • 4. Label preparation: as in example 7-9.
    • 5. Detection Molecules preparation: multiple (<1000) different Detection Molecules are generated, each with a different Binding Molecule encoded by a unique Label. The Binding Molecules (pMHCs) and Labels (2OS DNA-barcodes) are attached to the Linker (dextran-streptavidin-PE conjugate), to form Detection Molecules, in such a way that a given pMHC is always attached to a given DNA-barcode.
      • a. Detection Molecules are essentially generated in the same way as described in example 1-8.
      • b. Modification: Since the total volume of >1000 pooled Detection Molecules exceeds 100 μl this volume is reduced to reach a desired concentration of specific Binding Molecules. This is done as described in example 2-8.
      • c. Purification: As described in example 1-8.
    • 6. Incubation of sample and Detection Molecules: The cell sample and the Detection Molecules are incubated in the same way as described in example 1-2, 5-7. The cell sample and the Detection Molecules are mixed in one container to allow Detection Molecules to bind to T cells
    • 7. Enrichment of MHC molecules with desired characteristics: The Detection Molecules are enriched for in the same way as described in example 1-7. Cells associated with Detection Molecules are sorted by FACS, through means of the PE-fluorescence signal. The population of cells holding Detection Molecules are following injected to the FLUIDIGM C1 unit allowing for single cell distribution in a micro-well system. Other similar devices or platforms for single cell amplification can also be used.
    • 8. Identification of enriched Detection Molecules: For each cell in the FLUIDIGM C1 unit we will amplify a) the label (=DNA oligonucleotide barcode) and b) the TCR V-alpha and -beta chains. In each well specific primers holding cell identification keys, will be used to amplify DNA oligonucleotide Label. Likewise specific primers holding cell identification keys will be used to amplify the TCR-associate genes, the TCR V-alpha and Beta chains. The complete sequence of the TCR chains will allow the assembly of a fully functional TCR sequence. In parallel knowledge about the Binding Molecules associated with the given T cell, will provide insight o the antigen recognition of the given TCR. Thus we can obtain paired samples of TCR sequences and antigen specificities, using the strategy explained in this example.

The expected outcome of this example is knowledge of the TCR receptor coupled to the knowledge of the recognition of Binding Molecules of this given TCR. Single-cell sorting will enable the generation of a correctly paired and fully functional TCR. The association of the labels will explain the recognition motif of this T cell receptor and provide valuable information in this regard. The technique described here will allow the parallel assessment of multiple different T cells while enabling the capture of specific T cells receptor sequences and have paired knowledge about the Binding molecules associated to this. The overall principle of example 10 is schematized in FIG. 6C.

Example 11

In this example all cells carry the same TCR (T cell receptor). A large number of different pMHC complexes (binding molecules) are employed, in order to examine the recognition breadth and affinity of a given T cell receptor (TCR).

This example describes how the detection of Binding Molecules can be used to determine the recognition breadth and affinity of a given T cell receptor (TCR).

This is an example where the Sample (1) is a T cell clone or a culture of T cell receptor transduced T cells. Importantly for this example all cells will hold the same T cell receptor.

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) will be peptide-MHC (pMHC) complexes displaying a library of peptides that are potentially recognized by the T cell receptor in question. This could ideally be a set of alanine-scanning substituted peptides modified based on the known recognition sequence of the T cell receptor in question. The Binding Molecules will be modified (3b) by biotinylation and quality controlled as described in example 1-6.

The Labels (4) are oligonucleotides as in example 7-9.

The detection molecule (5) will be synthetized (5a) and modified (5b) as in example 7-9. Multiple different detection molecules will be generated wherein the individual detection molecules containing different pMHC were encoded by corresponding individual oligonucleotide labels, as in example 7-9.

An amount of Sample, T cell clone (1b) will be mixed with detection molecules (5) under conditions (6c) that allow binding of detection molecules to the T cells in the sample.

Cells will be washed to remove excess Detection Molecules, and following the identity of the Binding Molecules will be revealed through sequencing of the Label.

This example thus reveals the possibility to identify the specificity and breadth of specificity of a given T cell receptor. This knowledge is essential for the development of T cell receptor gene therapy strategies, and adoptive transfer of T cell population carrying specific T cell receptors.

    • 1. Sample preparation. The cell sample to be used in this example is a T cell clone (carrying a single, defined T cell receptor) or a culture of T cells transduced with a given T cell receptor.
      • e. Acquiring sample: Any PBMC sample can be used as a source to transduce the T cells with a characterized T cell receptor (TCR), expressed in a retroviral vector.
      • f. Modifying sample: PBMCs modified to express a given TCR following retroviral transduction using a vector expressing the selected TCR.
    • 2. Linker preparation: The linker used in this example is prepared as in example 1-10. The fluorochrome has no particular use in this example. Consequently, the fluorochrome attachment is not needed.
    • 3. Binding molecule preparation: The binding molecules used in this example will be a collection of peptide-MHC molecules, designed to assess the breadth to recognition of the given TCR. The peptide library could be alanine substitution libraries of the known peptide-epitope recognized by the given TCR. Alanine substitution is used to assess the essential amino acids in given positions important for TCR recognition.
      • a. Synthesis: as in example 7-9.
      • b. Modification: No further modifications
      • c. Purification: as in example 7-9.
    • 4. Label preparation: as in example 7-9.
    • 5. Detection molecules preparation: as in example 7-9.
    • 6. Incubation of sample and detection molecules: The cell sample and the Detection Molecules were mixed in one container, to allow the Detection Molecules to bind the T cells that they recognize.
      • a. Amount of sample: 100.000 cells expressing a given TCR
      • b. Amount of detection molecule: as in example 7-9.
      • c. Conditions: The 100.000 cells will be mixed with different quantities of Detection Molecules. This being the standard amount of detection molecules as given in example 7-9, but with parallel detection analysis using e.g. 5× excess for Detection molecules, as well as 5×, 25×, 125× fold less Detection Molecules. Such titration is done to assess the avidity of the selected TCR towards the Binding Molecule library used.
    • 7. Enrichment of detection molecules with desired characteristics: In this example, excess Detection molecules are separated from Sample through centrifugation. All cells are expected to bind Detection molecules to some extent, and consequently all cells are used for characterization of Binding Molecules.
    • 8. Identification of enriched Detection Molecules: The identification of Detection Molecules and analyses of sequencing results is conducted as described in example 7.

The expected outcome of this example is knowledge related to the Detection molecules associated with a given T cell receptor. Through this technology we can gain knowledge on the breadth and the avidity of a given TCR towards a large library of similar, overlapping and/or alanine substituted peptide-epitope sequences. The technique can be used to understand both avidity and ‘fine-specificity’ of a given TCR. This will be of crucial importance for development of TCR receptor-associated therapies, such as TCR gene therapy in cancer treatment. The overall principle of example 11 is schematized in FIG. 6D.

Example 12

In this example it is described how the present invention can be used to identify the specificity of the stimuli that lead to a functional response of certain cells. In the example, the external stimulus is the addition of tumor cells, and the functional response measured is the release of INF-γ.

This example will describe how the detection of Binding Molecules can be associated to a functional response to a given stimuli provided to the Sample in vitro.

This is an example where the Sample (1) will be tumor infiltrating lymphocytes (TIL) from cancer patients which are modified to determine the reactivity toward tumor cells (1b). The Sample is mixed with tumor cells to allow cytokine release from responding T cells in the Sample. The cytokines are trapped intracellular following Golgi-transport blockade and cellular fixation.

The Linker (2) was a dextran conjugate with streptavidin and fluorochrome (Dextramer backbone from Immudex).

The Binding Molecules (3) will be peptide-MHC (pMHC) complexes displaying a library of melanoma-associated peptides (as described in Andersen et al. Dissection of T cell antigen specificity in human melanoma. Cancer Research 2012 Apr. 1; 72(7):1642-50), and used in example 7. The Binding Molecules will be modified (3b) by biotinylation and quality controlled as described in example 1-6.

The Labels (4) were oligonucleotides as in example 7-9.

The detection molecule (5) will be synthetized (5a) and modified (5b) as in example 7-9. 175 different detection molecules were generated wherein the individual detection molecules containing different pMHC were encoded by corresponding individual oligonucleotide labels, as in example 7.

An amount of Sample, TILs (1b) will mixed with detection molecules (5) under conditions (6c) that allow binding of detection molecules to T cells in the sample. Cells will be selected by FACS based on their cytokine release upon stimulation with the tumor cells. After cellular selection, cells encompassing different cytokine profiles will be analyzed for their binding to the Detection Molecules, consequently describing the T cell receptor specificity of the responding T cells, i.e. T cells recognizing tumor cells.

This example thus reveal the possibility to identify potential differences in T cell specificity among cells responding differently to a given stimuli, here provided be the mixture of T-cells with tumor cells.

    • 1. Sample preparation. The cell sample to be used in this example is tumor infiltrating lymphocytes (TILs) obtained from melanoma patients (as example 7).
      • a. Acquiring sample: as in example 7.
      • b. Modifying sample: TILs will be purified and expanded as in example 7. Following, TILs will be mixed with tumor cell to assess cytokine release upon T-cell mediated tumor cell recognition, using the following protocol for intracellular cytokine staining: Tumor cells will be thawed, washed twice in culture media (RPMI), and cultured in RPMI+10% FCS (R10) until they had expanded to a sufficient amount of cells. TILs will be thawed in 10 ml RPMI+2.5 ul DNase and 50 ul MgCl2, washed twice in RPMI and rested overnight or at least 4 hr in X-vivo+5% HS+100 U/ml IL-2. Following rest, the cells will be washed, counted and resuspended in X-vivo+5% HS obtaining a concentration of 3*106 cells/ml. 100 ul of the cell suspension will then be added to at least two wells in a 96 well plate—more replicates will be made if enough cells are available. Thus 3*105 cells will added to each well. Tumor cells will be trypsinated, washed in R10, counted and resuspended in RPMI+10% HS to a concentration of 2*106 cells/ml. 50 ul of cell suspension containing 1*105 cells will be added to the TILs. To every well, 50 ul of Golgi medium, containing 45 ul RPMI+10% HS, 5 ul BV421 conjugated CD107a antibody (BD pharmingen) and 0.2 ul Golgi Plug (BD Bioscience 555029) was added. The cells were then incubated 4-5 hours in 37° C. All samples were cultured in a ratio of 3:1 TIL:tumor cells, with a total of 4*105 cells per well. After end incubation, the cells were spun and all replicates were collected into one well. Cell will be washed in PBS+2% FCS (FACS buffer) and resuspended in 50 uL barcode-buffer (PBS/0.5% BSA/2 mM EDTA/100 μg/ml herring DNA).
    • 2. Linker preparation: The linker used in this example is prepared as in example 1-8. The fluorochrome has no particular use in this example. Consequently, the fluorochrome attachment is not needed.
    • 3. Binding molecule preparation: The binding molecules used in this example will be 175 different class I MHC-peptide complexes, as in example 7.
      • a. Synthesis: as in example 7.
      • b. Modification: No further modifications
      • c. Purification: as in example 7.
    • 4. Label preparation: as in example 7.
    • 5. Detection molecules preparation: as in example 7.
    • 6. Incubation of sample and detection molecules: The cell sample and the Detection Molecules were mixed in one container, to allow the Detection Molecules to bind the T cells that they recognize.
      • a. Amount of sample: 50 uL cell suspension as described in 1b.
      • b. Amount of detection molecule: as in example 7.
      • c. Conditions: The Detection molecules were added in the required amount. If necessary barcode-buffer was added to reach a total volume of 100 ul and cells were incubated 15 min, 37° C. Following incubation, cells were and stained with the following surface antibodies: anti-CD3 antibody, anti-CD8 antibody, anti-CD4 antibody and near-IR-viability dye (Invitrogen L10119). The cells were then incubated for 30 min at 4° C., after which they were washed twice in barcode-buffer and incubated in 200 ul fixation buffer (1:4 concentrate, eBioscience 00-5123-43, to diluent eBioscience 00-5223-56) overnight at 4° C. The following day, cells were washed twice in permeabilization buffer (1:10 buffer to water, eBioscience 00-8333-56), resuspended in 50 ul permeabilization buffer and stained with intracellular antibodies: FITC-conjugated anti-TNF antibody (BD pharmingen 562082), APC-conjugated anti-IFN antibody (BD 341117). After incubating for 30 min at 4° C. with the antibodies, the cells were washed twice in permeabilization buffer, resuspended in 50 ul barcode-buffer.
    • 7. Enrichment of detection molecules with desired characteristics: In this Example, the Sample is selected based on the cytokine secretion mediated following incubation with tumor cells. Cytokines are visualized through intracellular cytokine staining (ICS). The Sample is following stained with the Detection Molecules. Cytokine producing cells will be selected by Fluorescence-Activated-Cell-Sorting (FACS), and the Detection Molecules carried along with a given cytokine profile of a given cell population will be assessed through sequencing of the co-attached oligonucleotide barcodes
      • a. Apply: Cells were sorted on a BD FACSAria, equipped with three lasers (488 nm blue, 633 nm red and 405 violet). The flow cytometry data analyses will be performed using the BD FACSDiva software version 6.1.2. The following gating strategy will be applied. Lymphocytes were identified in a FSC/SSC plot. Additional gating on single cells (FSC-A/FSC-H), live cells (near-IR-viability dye negative), and CD8, CD3 positive cells, and CD4 negative.
        • From this defined cell population two separate subsets will be sorted based ion the cytokine secretion in relation to tumor-cell stimulation. T cells positive for any of the detected cytokines ‘ICS positive’ is sorted in one tube, and T cells negative for any of the detected cytokines ‘ICS negative’ is sorted in another tube.
      • b. Wash: not applicable.
      • c. Separate: Optionally cells were acquired up to one week after fixation in 1% paraformaldehyde. The ‘ICS positive’ and ‘ICS negative’ cells were sorted by FACS, as described in 7a, into tubes that had been pre-saturated for 2 h-O.N. in 2% BSA and contained 200 μl barcode-buffer to increase the stability of the oligonucleotides that followed with the sorted cells. The sorted cells were centrifuged 5 min, 5000 g, to allow removal of all excess buffer. Cells were stored at −80° C.
    • 8. Identification of enriched Detection Molecules: The identification of Detection Molecules and analyses of sequencing results is conducted as described in example 7. The ‘ICS positive’ and ‘ICS negative’ cells are treated as two independents samples. Following these two samples are compared for the association with Binding Molecules.

The expected outcome of this examples is knowledge related to the Detection molecules associated with tumor cell recognition (=‘ICS positive’) as oppose to no recognition (=‘ICS negative’). In other terms, we will gain knowledge on the T cell recognition elements associated with tumor cell recognition, among a large pool of different Detection Molecules, here peptide-MHC molecules. Knowledge of T cell mediated recognition of tumor cells has major impact on the design and development of immunotherapeutic strategies for cancer. The overall principle of example 12 is schematized in FIG. 6A.

Cell could likewise be selected based on e.g. phenotypic characteristics, to assess what T specificities are associated with selected phenotypic characteristics as schematized in FIG. 6B.

Tables for Examples 1-12:

TABLE 1
BCs used in Examples 1-7 with indicated frequencies of
peptide-antigen specific T cells as identified by conventional
MHC multimer staining:
EpitopeFreq. (%)
BC171A11 EBV-EBNA40.32
A3 CMV pp150 TVY0.015
BC254A2 FLU MP 58-66 GIL0.0522
A2 EBV LMP2 FLY0.014
A2 CMV pp65 NLV1.128
BC261A2 FLU MP 58-66 GIL0.125
A3 EBV EBNA 3a RLR0.0258
A2 EBV LMP2 FLY0.0075
BC266A1 CMV pp65 YSE0.0859
A1 FLU BP-VSD0.0628
BC268A2 FLU MP 58-66 GIL0.2523
A2 CMV pp65 NLV0.5445
BC260A2 FLU MP 58-66 GIL0.0456
A2 CMV pp65 NLV0.134
B7 CMV pp65 TPR4.5395
BC262A11 EBV-EBNA40.0872

TABLE 2
Structure and sequences of 2OS A oligonucleotides and 2OS B
oligonucleotides, used to produce 2OS DNA-barcodes:
Oligo name5′ modificationForward primer region6xN regionCoding regionAnnealing region
2OS-1-Oligo-A1Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNCGAGGGCAATGGTTAACTGACACGTGGTCAGCATCATTTCC
2OS-1-Oligo-A2Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNCAGAAAGCAGTCTCGTCGGTTCGAAGGTCAGCATCATTTCC
2OS-1-Oligo-A3Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNTAAGTAGCGGGCATAATGTACGCTCGGTCAGCATCATTTCC
2OS-1-Oligo-A4Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNGGATCCAGTAAGCTACTGCGTTTATGGTCAGCATCATTTCC
2OS-1-Oligo-A5Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNGGGCTGCGGAGCGTTTACTCTGTATGGTCAGCATCATTTCC
2OS-1-Oligo-A6Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNAAACGTATGTGCTTTGTCGGATGCCGGTCAGCATCATTTCC
2OS-1-Oligo-A7Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNATATCATCATAGGCTTAGCGACGTAGGTCAGCATCATTTCC
2OS-1-Oligo-A8Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNAGGAAAATCTGCTACCGCCAATGATGGTCAGCATCATTTCC
2OS-1-Oligo-A9Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNCTGATTGACTGCATGGAGGCTATACGGTCAGCATCATTTCC
2OS-1-Oligo-A10Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNGTGGCGACTTCACGATTATCTGAACGGTCAGCATCATTTCC
2OS-1-Oligo-A11Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNCCTGTATTGAAGGTTCAGTCCTGTTGGTCAGCATCATTTCC
2OS-1-Oligo-A12Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNGGCTCTATAAGGTTTCCTCAAAGGTGGTCAGCATCATTTCC
2OS-1-Oligo-A13Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNTTGGGAGCTTTCCTATGTACAGTCCGGTCAGCATCATTTCC
2OS-1-Oligo-A14Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNAGAGAATATGTCGCTCCCGTTATGTGGTCAGCATCATTTCC
2OS-1-Oligo-A15Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNGCAGTTAGATATGCAGTTACCTGACGGTCAGCATCATTTCC
2OS-1-Oligo-A16Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNCTTCACCCGAACATGCAGTGTTATTGGTCAGCATCATTTCC
2OS-1-Oligo-A17Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNAAAGCCGTTGCAGTATCGTCTGAGCGGTCAGCATCATTTCC
2OS-1-Oligo-A18Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNGCTGGATGTTAATAACTGCGGTCCGGGTCAGCATCATTTCC
2OS-1-Oligo-A19Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNACGAGTTGACATGGACGGATCCCTCGGTCAGCATCATTTCC
2OS-1-Oligo-A20Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNTTCATCACTCATTGTTCTGAGTAGGGGTCAGCATCATTTCC
2OS-1-Oligo-A21Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNATGTTTAATCTAACTTGATGCCTCCGGTCAGCATCATTTCC
2OS-1-Oligo-A22Biotin-C6-GAAGTTCCAGCCAGCGTCACAGTTTNNNNNNTAATACGCCTGAGGTGTTGGGTTGCGGTCAGCATCATTTCC
2OS-1-Oligo-B1CTGTGACTATGTGAGGCTTTCTCGANNNNNNGCCTGTAGTCCCACGCGATCTAACAGGAAATGATGCTGACC
2OS-1-Oligo-B2CTGTGACTATGTGAGGCTTTCTCGANNNNNNCAACCATTGATTGGGGACAACTGGGGGAAATGATGCTGACC
2OS-1-Oligo-B3CTGTGACTATGTGAGGCTTTCTCGANNNNNNACGTTTAAGCATCTGTACTCCAGATGGAAATGATGCTGACC
2OS-1-Oligo-B4CTGTGACTATGTGAGGCTTTCTCGANNNNNNGAATTGAAGCCATCGTTTCGCGCAAGGAAATGATGCTGACC
2OS-1-Oligo-B5CTGTGACTATGTGAGGCTTTCTCGANNNNNNCGTAGCTTTTGTAGCGTCTGAGGGCGGAAATGATGCTGACC
2OS-1-Oligo-B6CTGTGACTATGTGAGGCTTTCTCGANNNNNNAATCGTCAGTCCCTGTTTCGACATCGGAAATGATGCTGACC
2OS-1-Oligo-B7CTGTGACTATGTGAGGCTTTCTCGANNNNNNCGGTGGTAGGTGATACTTCTGTACCGGAAATGATGCTGACC
2OS-1-Oligo-B8CTGTGACTATGTGAGGCTTTCTCGANNNNNNTGACTATCGGGGCGTGACATGAGCTGGAAATGATGCTGACC
2OS-1-Oligo-B9CTGTGACTATGTGAGGCTTTCTCGANNNNNNGTTGGTGAAACTACCGACGCTTTACGGAAATGATGCTGACC
2OS-1-Oligo-B10CTGTGACTATGTGAGGCTTTCTCGANNNNNNAATGGAGGTGCAGGAATACTCTCGTGGAAATGATGCTGACC
2OS-1-Oligo-B11CTGTGACTATGTGAGGCTTTCTCGANNNNNNAAAACGCACCACAACTCGGACGTGAGGAAATGATGCTGACC
2OS-1-Oligo-B12CTGTGACTATGTGAGGCTTTCTCGANNNNNNGCCATATAGCACAGCACGCAATCCGGAAATGATGCTGACC
2OS-1-Oligo-B13CTGTGACTATGTGAGGCTTTCTCGANNNNNNCCTATGCGAACTTGGTTTATCCTGCGGAAATGATGCTGACC
2OS-1-Oligo-B14CTGTGACTATGTGAGGCTTTCTCGANNNNNNAAGCTGCGTATCCTCGAACTAGCAGGGAAATGATGCTGACC
2OS-1-Oligo-B15CTGTGACTATGTGAGGCTTTCTCGANNNNNNATGGCGCAGACATTCTGTAGTCGCAGGAAATGATGCTGACC
2OS-1-Oligo-B16CTGTGACTATGTGAGGCTTTCTCGANNNNNNCTTATGGACTGGTTGGGGACAATCCGGAAATGATGCTGACC
2OS-1-Oligo-B17CTGTGACTATGTGAGGCTTTCTCGANNNNNNGTCCTTCCTTACGAAATATTGGTCGGAAATGATGCTGACC
2OS-1-Oligo-B18CTGTGACTATGTGAGGCTTTCTCGANNNNNNTGATGAACCAATCCTCCGATTTCTTGGAAATGATGCTGACC
2OS-1-Oligo-B19CTGTGACTATGTGAGGCTTTCTCGANNNNNNACCGAATGTGGGCCACGAGTCATTCGGAAATGATGCTGACC
2OS-1-Oligo-B20CTGTGACTATGTGAGGCTTTCTCGANNNNNNCGGGTGAGCATATAACTTGCAATTCGGAAATGATGCTGACC
2OS-1-Oligo-B21CTGTGACTATGTGAGGCTTTCTCGANNNNNNAGAATTGTGCTTGGGGCGATTCATAGGAAATGATGCTGACC
2OS-1-Oligo-B22CTGTGACTATGTGAGGCTTTCTCGANNNNNNAATTGGTGACATGCTTAACTACCGTGGAAATGATGCTGACC
2OS-1-Oligo-B23CTGTGACTATGTGAGGCTTTCTCGANNNNNNGAGACGCCTAGAAAGTGATTAACTCGGAAATGATGCTGACC
2OS-1-Oligo-B24CTGTGACTATGTGAGGCTTTCTCGANNNNNNATTACAGTTACAGTGCTGGTCGCAGGGAAATGATGCTGACC
2OS-1-Oligo-B25CTGTGACTATGTGAGGCTTTCTCGANNNNNNCGTTACGTTGGTGGGCTCTTGGTACGGAAATGATGCTGACC
2OS-1-Oligo-B26CTGTGACTATGTGAGGCTTTCTCGANNNNNNGTTATTATCGGTGTCCCGACTAGTTGGAAATGATGCTGACC
2OS-1-Oligo-B27CTGTGACTATGTGAGGCTTTCTCGANNNNNNAGAAATGATTCCCGAGTCGCCTTTTGGAAATGATGCTGACC
2OS-1-Oligo-B28CTGTGACTATGTGAGGCTTTCTCGANNNNNNTGCTCTCGGATGTGGTTCTATGGATGGAAATGATGCTGACC
2OS-1-Oligo-B29CTGTGACTATGTGAGGCTTTCTCGANNNNNNGAGTTAAAACCGTCGCCGTAGCACTGGAAATGATGCTGACC
2OS-1-Oligo-B30CTGTGACTATGTGAGGCTTTCTCGANNNNNNTGTACGCGATAGTACTCGGGTCCTGGGAAATGATGCTGACC
2OS-1-Oligo-B31CTGTGACTATGTGAGGCTTTCTCGANNNNNNAACTTACGCCCAGCAAGGCATTCATGGAAATGATGCTGACC
2OS-1-Oligo-B32CTGTGACTATGTGAGGCTTTCTCGANNNNNNAGCATGGCACAAGAGGAGCACTTCAGGAAATGATGCTGACC
2OS-1-Oligo-B33CTGTGACTATGTGAGGCTTTCTCGANNNNNNCGATCGTGAGTTTGCAGCGTGACGAGGAAATGATGCTGACC
2OS-1-Oligo-B34CTGTGACTATGTGAGGCTTTCTCGANNNNNNACAGCTCCAGCCTCCCTTTGTTTGTGGAAATGATGCTGACC
2OS-1-Oligo-B35CTGTGACTATGTGAGGCTTTCTCGANNNNNNAAACCTTTGTTCGGGCGTCTACCATGGAAATGATGCTGACC
2OS-1-Oligo-B36CTGTGACTATGTGAGGCTTTCTCGANNNNNNTCTTTCAAAACAGCGGGAGTCATCGGGAAATGATGCTGACC
2OS-1-Oligo-B37CTGTGACTATGTGAGGCTTTCTCGANNNNNNGCGGTTTATCCGAATCTCACGCTAAGGAAATGATGCTGACC
2OS-1-Oligo-B38CTGTGACTATGTGAGGCTTTCTCGANNNNNNGCATATGCTACAGGCTGGGGTGAACGGAAATGATGCTGACC
2OS-1-Oligo-B39CTGTGACTATGTGAGGCTTTCTCGANNNNNNGGAGGTCTAAACGTCCGGAGCTATTGGAAATGATGCTGACC
2OS-1-Oligo-B40CTGTGACTATGTGAGGCTTTCTCGANNNNNNAAGAATAAGATTGCGTGCGCCTTAAGGAAATGATGCTGACC
2OS-1-Oligo-B41CTGTGACTATGTGAGGCTTTCTCGANNNNNNCATCATCGTCGTCCAAATATGTGATGGAAATGATGCTGACC
2OS-1-Oligo-B42CTGTGACTATGTGAGGCTTTCTCGANNNNNNCACGTGTAGCTGTGGGCCAAGTCTAGGAAATGATGCTGACC
2OS-1-Oligo-B43CTGTGACTATGTGAGGCTTTCTCGANNNNNNCAGTTGTCAAATCTCCGCATTGGTAGGAAATGATGCTGACC
2OS-1-Oligo-B44CTGTGACTATGTGAGGCTTTCTCGANNNNNNACTGGTAATGCCATTGGTCTAAATGGGAAATGATGCTGACC
2OS-1-Oligo-B45CTGTGACTATGTGAGGCTTTCTCGANNNNNNGTCTTTGGTCGTAACGAATCTCCGTGGAAATGATGCTGACC
2OS-1-Oligo-B46CTGTGACTATGTGAGGCTTTCTCGANNNNNNCTTAGGCATGACGGGGTTGTCCATGGGAAATGATGCTGACC
2OS-1-Oligo-B47CTGTGACTATGTGAGGCTTTCTCGANNNNNNCCGGTGAATTTTGGGTGTCCATGTAGGAAATGATGCTGACC
2OS-1-Oligo-B48CTGTGACTATGTGAGGCTTTCTCGANNNNNNCCTTTATCTCCTCCACCTATAAGGTGGAAATGATGCTGACC
2OS-1-Oligo-B49CTGTGACTATGTGAGGCTTTCTCGANNNNNNGATACTATATGACGGCCTGTAATCGGGAAATGATGCTGACC
2OS-1-Oligo-B50CTGTGACTATGTGAGGCTTTCTCGANNNNNNATTGGTTGGCCGAAAGACTACATCTGGAAATGATGCTGACC
2OS-1-Oligo-B51CTGTGACTATGTGAGGCTTTCTCGANNNNNNCGTAGTTATGGGGTGGGTCACCTGCGGAAATGATGCTGACC
2OS-1-Oligo-B52CTGTGACTATGTGAGGCTTTCTCGANNNNNNAAGTTTCCAGGCACTGATTCGTTCCGGAAATGATGCTGACC
2OS-1-Oligo-B53CTGTGACTATGTGAGGCTTTCTCGANNNNNNTTCCTTATTTCCCGGTTGAGATACAGGAAATGATGCTGACC
2OS-1-Oligo-B54CTGTGACTATGTGAGGCTTTCTCGANNNNNNAGGTATCATGCGGGCCGAATCTTGGGGAAATGATGCTGACC
2OS-1-Oligo-B55CTGTGACTATGTGAGGCTTTCTCGANNNNNNATACCCGTAGGCCAGTACCCTCTCCGGAAATGATGCTGACC

TABLE 3
The reagents used for annealing (left) and elongation
(right) of partly complementary oligonucleotides. Reagents
marked in italic are from the Sequenase Version 2.0 DNA
Annealing reaction(μl)Elongation reaction(μl)
Oligo A (100 μM)2.6Annealing reaction10μl
Oligo B (100 μM)5.40.1M DTT1μl
Sequenase reaction2H2O0.5μl
buffer
Total108x diluted2μl
Sequenase
polymerase
5x diluted Sequence2μl
extension mixture
Total15.5

TABLE 4
Overview of reagents required for production of Binding
Molecules produced from 100 μg/ml pMHC exchange
reaction. The amounts of Detection Molecule used for staining
1 × 106-2 × 106 cells in 100 μl are also specified.
Amount
Exchanged pMHC/ulper
SA conjugateD-biotinEnd: pMHCstaining
PE dex1.32 μl12.6 μM44 ug/ml3 ul

TABLE 5
The components of the antibody mixture added while Detection
Molecules are incubated with sample. The amount listed is for
incubation of 1 × 106-2 × 106 cells in 100 ul.
TargetConjugateAmount (μl)Source
CD8PerCP2Invitrogen MHCD0831
CD4FITC1.25BD bioscience 345768
CD14FITC3.13BD bioscience 345784
CD16FITC6.25BD bioscience 335035
CD19FITC2.50BD bioscience 345776
CD40FITC1.56Serotec MCA1590F

TABLE 6
The Master mix applied for recovery of DNA-barcodes by
QPCR. The template was drawn from the residual fluid
(5-9.25 ul) containing the sorted cells. Nuclease free
H2O was added to a final volume of 25 ul per PCR
ComponentVolume per sample (μl)
Master mix12.5
Probe/SYBR (10 uM/100x)0.25(0.1 uM/1x)
Forward primer (5 uM)1.5(300 nM)
Reverse primer (5 uM)1.5(300 nM)
Template5-9.25
Nuclease free H2O0-4.25
Total25

TABLE 7
The thermal profile applied for qPCR amplification of barcodes
associated with sorted cells.
Temperature (° C.)TimeNo. of cycles
9510 min1
9530 s
6060 s40

TABLE 8
Structure and sequences of 1OS oligonucleotides applied as
DNA barcodes (1OS-1-Oligo-1 to 1OS-1-Oligo-110)
Oligo name5′ modifForward primer region6xNCoding regionReverse primer region
1OS-1-Oligo-1Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTATGAGGACGAATCTCCCGCTTATAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-2Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTCTTGACAAACGTGTGCTTGTACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-3Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTTTATCGGGCGTGGTGCTCGCATAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-4Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCCGATGTTGACGGACTAATCCTGACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-5Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTAGTAGTTCAGACGCCGTTAAGCGCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-6Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCCGTACCTAGATACACTCAATTTGTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-7Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGGGTTCCGTTTTACATTCCAGGAAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-8Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTATCCCGTGAAGCTTGAGTGGAATCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-9Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTGACGTGTGAGGCGCTAGAGCATAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-10Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTATGGCACGCCTAATCTGGACACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-11Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGATGCATGATCTAGGGCCTCGTCTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-12Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGAGGTCTTTCATGCGTATAGTCACAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-13Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGATTCAATATGTGTCGTCTATCCTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-14Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTAACTGCGCATAGTTGGCTCTATGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-15Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCGTTTAAGGTCACATCGCATGAATGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-16Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCCCGGGAAGTGTGAGGATATACCCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-17Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTCTTAAAACTGGTATCACCTGACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-18Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGGTGGTTAGTGATTTGCCCGTCACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-19Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTAGTTGGTGGGTTTCCCTACCGTGTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-20Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTACAGTAAGTGAGAATCCTCTCTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-21Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTTCTAAGTTTAGCGTAGCCGGTTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-22Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTTTAGGTGGGTGCGATTGCCAGTTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-23Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCCACCTTAACACGCGATGATATTGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-24Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTATTACGAGCGCTTGGATCCCGTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-25Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTATGTTGTGCCTTACGCCTCGATTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-26Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTTAACCGAACTGACGGCCATCAAGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-27Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGGTACATGCGCCTTACTCCTTGTGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-28Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTTCTATTCTAAGCCGGCGGTCATATGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-29Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTTGATGCTTTACAAGATCGCGTTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-30Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCCAAGTTAGCTTACTCCATGCCCCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-31Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGAACTATTTCCTGGCTGTTACGCGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-32Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCGGTTTCAAGGATGATCCGCGCTTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-33Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGAGACTGCCCGACACATCTTAGTGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-34Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTGTTAATTAGGCTCGGTCGGCCTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-35Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGGTAGTCCTATGCGGGCTTTCTCTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-36Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGCTTGGACTATAGTCATCGCGTTTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-37Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCACTGTTTAACAAGCCCGTCAGTAGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-38Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNACGTCGTATTATACCCGCCATGGAAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-39Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTGCTTAATTTACGACCGATGCTGCGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-40Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCCATAGATTTCTCCGTGAGTCTTTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-41Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTGCCGCAGACATTGCATACGATATGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-42Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCAGGTCCTAACCCGCAACCATTTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-43Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTGCACCGTTCATATGTTATCGGGACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-44Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGAGACTTACACCCGTAGACGTCGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-45Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNATAAAAGAAACCCTCCGCATTGTGTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-46Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTCCCATCCGAGCAGATTTGACTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-47Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNATGAGCTGTCTCGAACCGAAGGCACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-48Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCGGGCGGTTCAACTTACTGGTAGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-49Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGGGAAATAACGGATGCGCTCTTGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-50Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNACTTCTTCTCGGTCGCATGAGGCTGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-51Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGATACATATACGCTCGTCGGGACTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-52Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCCGGGAAGTGTCATAACTTGAAGCGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-53Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTCAGCCTGCCTCGCTTCTGATATTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-54Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGGGCCAAGTCGACCTAGATGGCTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-55Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTAGGGCTACTGTTATCCTCCGTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-56Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCGTACGGCTGGAGAGCTGTATGTGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-57Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNACAGGTTGTATTACTTCGCGCCTTGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-58Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTGGGCTCATTACAAGTGTTGCATAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-59Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTAAGTGGCGCCGATTGTTTGTCCAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-60Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTATATTTTGCTCCCGGCGACGAGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-61Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCAATTTGCGCTTGTTCGGCATAGCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-62Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGAGTCGAATATCCACCACCGTATGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-63Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTTGTGGTTTGGGTCCTCAGAGGAGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-64Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGTACCTAGTCTCGTAATCATAGGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-65Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCGGCATGATCTACCTTAAAGCTTGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-66Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCCGGCGCAGAAGTTTGAACGAAAAGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-67Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNATGCACTATTTTACGTATCCCGTGCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-68Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGATAGGGTGACTGCTTTCGCGTACAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-69Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTATCTGGTAGACATCTCGGCACAGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-70Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCGGGGTGCAATAATCACTAGTGCTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-71Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTAGTTCTGGCTATACACACTTCGGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-72Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCATAGAGTTACCCGATGGATTCGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-73Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTTCATGGTACAGGCTTCTTTACGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-74Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCGATCTCGGGCCTGGGTTTTGAGTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-75Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNATTATTCGTGACCCAACTCATCAGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-76Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTGAATGGTGAATAATGCGTTCGCCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-77Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTATTAGTTGCTACCCCAAGAATCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-78Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGAAAGTCTTGGATACACGGCCGGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-79Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTGTGTTCCTATGCACAATTTCATAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-80Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTACATGGTAGGGGTCTCCGAACCGTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-81Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTAGGGATAACTTTCCTCCCACTTGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-82Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCTGGTGTCTCACCCATGGGATGTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-83Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTAACGATTTTCTCGCGGGAGTTTCGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-84Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCGAGCCTGGTTAGCGCCTACAAGAGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-85Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCGTAGTAAGATATGTAGTCCACGTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-86Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTGTTAGTTGCCCCATATCTTTACGCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-87Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTGGATTGTGATTGTCCGGATCCGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-88Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGGAGGACTGCGGTTCAGCTTACAAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-89Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCGTAGGTCTAGTTCAGATTCTATAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-90Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTCTACGTGGTTCTATACCATTCGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-91Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGGCTTTACTACAATGCGTGGGCTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-92Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGCTTGCTGTATGGGTCATGTTCCTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-93Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTGCTCTAAAGACGCGAGGACTACCTGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-94Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTGTACATGTCATACTCAAGGCTTTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-95Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTTGACATGTACGCCATTTGGGTCGCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-96Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCAATTCAGTACGATCGTGTAGCGGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-97Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCGCTGTCCAAAGGTTCTTCGTAACGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-98Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTTAGACGAGCAGGTTTCTTGCCTATGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-99Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNTCGTTTGGAGCCGTTCACACATGAAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-100Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTGATCAACTTGCGCCCAGCGTTATGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-101Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGACGATGTTGCCTGTTTTGATACGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-102Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGGGTAGTCGTGAGGTGAACTCTTCCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-103Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNAGCCATTTTACGATTCTATTCGATGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-104Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGTGGTTTATATAATCCCACCTCCTAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-105Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCGAAGAACATCCCGGCATTTCATGGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-106Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTGGGACAATGCCGAAAACTCTTCGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-107Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNATTCCGTACCAACCCGCGTCTTAGAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-108Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNCTGCAGGAGGCTCTAATGCACTCAAGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-109Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCGTTCAGCATTCATTACGTCTCACGGTACGGCGCTATCATGTACTCATG
1OS-1-Oligo-110Biotin-C6-AGATTCTATAAACTGTGCGGTCCTTNNNNNNGCTGAGGAAGCCCAATGTTCAGTACGGTACGGCGCTATCATGTACTCATG

TABLE 9
Listing of the 110 combinations of peptide-HLA Binding Molecules and
the respective Labels, 1OS and 2OS DNA-barcodes, that were used to encode
the given specificity of all Detection Molecules applied in experiments 3-6.
In the table, HLA code ′A1′ refers to HLA-A0101. HLA code ′A2′ refers to HLA-A0201.
HLA code ′A3′ refers to HLA-A0301. HLA code ′A11′ refers to HLA-A1101. HLA code
′A24′ refers to HLA-A2402. HLA code ′B7′ refers to HLA-B0702.
HLA
1OS Barcode2OS BarcodecodePeptideSequence
1A1B1A1CMV pp65 YSEYSEHPTFTSQY
2A1B2A1CMV pp50 VTEVTEHDTLLY
3A1B3A1FLU BP-VSDVSDGGPNLY
4A1B4A11EBV-EBNA4AVFDRKSDAK
5A1B5A11HCMV pp65GPISGHVLK
6A1B6A11VP1DLQGLVLDY
7A1B7A11VP1VLGRKMTPK
8A1B8A11VP1VTLRKRWVK
9A1B9A11VP1LVLDYQTEY
10A1B10A11VP1GQEKTVYPK
11A2B1A11VP1VTFQSNQQDK
12A2B2A11VP1LKGPQKASQK
13A2B3A11VP1NVASVPKLLVK
14A2B4A11VP1TSNWYTYTY
15A2B5A11VP1LVLDYQTEYPK
16A2B6A11VP1TLRKRWVKNPY
17A2B7A11VP1AVTFQSNQQDK
18A2B8A11VP1PLKGPQKASQK
19A2B9A2VP1RIYEGSEQL
20A2B10A11VP1SLFSNLMPK
21A3B1A2VP1KLLVKGGVEV
22A3B2A11VP1SLINVHYWDMK
23A3B3A2HPV E6 29-38TIHDIILECV
24A3B4A2FLU MP 58-66 GILGILGFVFTL
25A3B5A2EBV LMP2 CLGCLGGLLTMV
26A3B6A2EBV BMF1 GLCGLCTLVAML
27A3B7A2EBV LMP2 FLYFLYALALLL
28A3B8A2CMV pp65 NLVNLVPMVATV
29A3B9A2EBV BRLF1 YVLYVLDHLIVV
30A3B10A2HPV E7 11-20YMLDLQPETT
31A4B1A2CMVIE1 VLEVLEETSVML
32A4B2A2VP1GCCPNVASV
33A4B3A2VP1SITQIELYL
34A4B4A2VP1LQMWEAISV
35A4B5A2VP1AISVKTEVV
36A4B6A2VP1KMTPKNQGL
37A4B7A2VP1TVLQFSNTL
38A4B8A2VP1GLFISCADI
39A4B9A2VP1LLVKGGVEVL
40A4B10A2VP1ELYLNPRMGV
41A5B1A2VP1NLPAYSVARV
42A5B2A2VP1TLQMWEAISV
43A5B3A2VP1QMWEAISVKT
44A5B4A2VP1VVGISSLINV
45A5B5A2VP1SLINVHYWDM
46A5B6A2VP1HMFAIGGEPL
47A5B7A2VP1FAIGGEPLDL
48A5B8A2VP1NLINSLFSNL
49A5B9A2VP1FLFKTSGKMAL
50A5B10A2VP1ALHGLPRYFNV
51A6B1A2VP1NLINSLFSNLM
52A6B2A2VP1FLDKFGQEKTV
53A6B3A2VP1VKGGVEVLSV
54A6B4A24HCMV 248-256AYAQKIFKIL
55A6B5A24EBV LMP2IYVLVMLVL
56A6B6A24EBV BRLF1TYPVLEEMF
57A6B7A24EBV BMLF1DYNFVKQLF
58A6B8A3CMV pp150 TTVTTVYPPSSTAK
59A6B9A3FLU NP 265-273 ILRILRGSVAHK
60A6B10A3EBV EBNA 3a RLRRLRAEAQVK
61A1B11A3CMV pp150 TVYTVYPPSSTAK
62A1B12A3EBV BRLF1 148-56 RVRRVRAYTYSK
63A1B13A3VP1ASVPKLLVK
64A1B14A3VP1CCPNVASVPK
65A1B15A3VP1ITIETVLGR
66A1B16A3VP1NTLTTVLLD
67A1B17A3VP1ALHGLPRYF
68A1B18A3VP1VASVPKLLVK
69A1B19A3VP1VSGQPMEGK
70A1B20A3VP1KASSTCKTPK
71A2B11A3VP1KTPKRQCIPK
72A2B12A3VP1YTYTYDLQPK
73A2B13A3VP1PITIETVLGR
74A2B14B7VP1SVARVSLPM
75A2B15A3VP1NSLFSNLMPK
76A2B16A3VP1KVSGQPMEGK
77A2B17A3VP1TVYPKPSVAP
78A2B18A3VP1SLINVHYWDMK
79A2B19A3VP1GVEVLSVVT
80A2B20A3VP1PLDLQGLVL
81A3B11A3VP1GLDPQAKAK
82A3B12A3VP1EVWCPDPSK
83A3B13A3VP1ADIVGFLFK
84A3B14A3VP1KTSGKMALH
85A3B15A3VP1KMALHGLPR
86A3B16A3VP1RYFNVTLRK
87A3B17A3VP1TLRKRWVKN
88A3B18B7CMV pp65 TPRTPRVTGGGAM
89A3B19B7CMV pp65 RPH-LRPHERNGFTV
90A3B20B7EBV EBNA RPPRPPIFIRLL
91A4B11B7VP1KPGCCPNVA
92A4B12B7VP1QPIKENLPA
93A4B13B7VP1LPRYFNVTL
94A4B14B7VP1MPKVSGQPM
95A4B15B7VP1YPKPSVAPA
96A4B16B7VP1KPSVAPAAV
97A4B17B7VP1APLKGPQKA
98A4B18B7VP1APKRKASSTC
99A4B19B7VP1SVARVSLPML
100A4B20B7VP1YPKTTNGGPI
101A5B11B7VP1YPKPSVAPAA
102A5B12B7VP1KPGCCPNVASV
103A5B13B7VP1NPRMGVNSPDL
104A5B14B7VP1LPAYSVARVSL
105A5B15B7VP1TPTVLQFSNTL
106A5B16B7VP1LPRYFNVTLRK
107A5B17B7VP1YPVVNLINSLF
108A5B18B7VP1YPKPSVAPAAV
109A5B19B7VP1KPSVAPAAVTF
110A5B20B7VP1APKRKASST

TABLE 10
The PCR Master mix applied prior to sequencing of DNA-
barcodes associated with sorted cells. The forward and
reverse primer included adaptors for the sequencing
reaction (A-key and P1-key respectively). Moreover the
forward primer carried a sample-identification barcode
(table 12 and 13). The template was drawn from the residual
fluid (10-19 ul) containing the sorted cells. Nuclease free
H2O was added to a final volume of 50 ul per PCR
ComponentVolume per sample (μl)
Master mix25
Forward primer (5 μM)3 (300 nM)
Reverse primer (5 μM)3 (300 nM)
Template10-19
Nuclease free H2O0-9
Total50

TABLE 11
The thermal profile applied for amplification of DNA-barcodes
associated with sorted cells. 36 cycles were applied if >1,000
cells were sorted while 38 cycles were applied if <1,000 cells
were sorted.
Temperature (° C.)TimeNo. of cycles
9510 min1
9530 s
6045 s36-38
7230 s
72 4 min1
4

TABLE 12
Forward and reverse primers applied for amplification of enriched
Detection Molecules prior to sequencing. Primers carry Ion Torrent adaptors, A-
key and P1-key. Moreover the forward primer encodes a unique sample-ID
barcode (6xN). Compatible with the 1OS Label system (F1 = forward primer,
R1 = reverse primer)
Primer nameForward primer region6xNIon Torrent region (A Key)
A-Key 1OS-F1-1GATTCTATAAACTGTGCGGTCCGAAGATCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-2GATTCTATAAACTGTGCGGTCCTCCTGACCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-3GATTCTATAAACTGTGCGGTCCTGTGGACCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-4GATTCTATAAACTGTGCGGTCCCATTTACCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-5GATTCTATAAACTGTGCGGTCCTTACCCCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-6GATTCTATAAACTGTGCGGTCCATTCTCCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-7GATTCTATAAACTGTGCGGTCCAGACCCCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-8GATTCTATAAACTGTGCGGTCCCGCATGCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-9GATTCTATAAACTGTGCGGTCCTCCTCGCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-10GATTCTATAAACTGTGCGGTCCATTCCTCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-11GATTCTATAAACTGTGCGGTCCCGTCGACCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-12GATTCTATAAACTGTGCGGTCCGCCAATCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-13GATTCTATAAACTGTGCGGTCCATACGGCCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-14GATTCTATAAACTGTGCGGTCCGTCAGACCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 1OS-F1-15GATTCTATAAACTGTGCGGTCCCGAGTTCCATCTCATCCCTGCGTGTCTCCGACTCAG
Primer nameIon Torrent (P1 Key)Reverse primer region
P1-key 1OS-R1CCTCTCTATGGGCAGTCGGTGATGAGTACATGATAGCGCCGTAC

TABLE 13
Forward and reverse primers applied for amplification of enriched
Detection Molecules prior to sequencing. Primers carry Ion Torrent adaptors, A-
key and P1-key. Moreover the forward primer encodes a unique sample-ID
barcode. A-key 2OS-F1-1-15 have a 6xN sample-ID barcode region (applied in
experiment 4 and 6) while A-key 2OS-F1-16-40 have a 8xN sample-ID and a
2xN spacer (used in all other experiments applying the 2OS system).
Compatible with the 2OS Label system (F1 = forward primer, R1 = reverse primer)
6N2N
Primer nameForward primer regionregion8N regionspacer
A-Key 2OS-F1-1GAAGTTCCAGCCAGCGTCCTGGGG
A-Key 2OS-F1-2GAAGTTCCAGCCAGCGTCCTCCAC
A-Key 2OS-F1-3GAAGTTCCAGCCAGCGTCCTTACC
A-Key 2OS-F1-4GAAGTTCCAGCCAGCGTCTGGCAG
A-Key 2OS-F1-5GAAGTTCCAGCCAGCGTCTGAGTA
A-Key 2OS-F1-6GAAGTTCCAGCCAGCGTCATTCAG
A-Key 2OS-F1-7GAAGTTCCAGCCAGCGTCTGAGCT
A-Key 2OS-F1-8GAAGTTCCAGCCAGCGTCGGCGTG
A-Key 2OS-F1-9GAAGTTCCAGCCAGCGTCAAATTG
A-Key 2OS-F1-10GAAGTTCCAGCCAGCGTCGCTGAC
A-Key 2OS-F1-11GAAGTTCCAGCCAGCGTCTTCTTA
A-Key 2OS-F1-12GAAGTTCCAGCCAGCGTCTGGTGG
A-Key 2OS-F1-13GAAGTTCCAGCCAGCGTCGCAGTC
A-Key 2OS-F1-14GAAGTTCCAGCCAGCGTCTCGTGA
A-Key 2OS-F1-15GAAGTTCCAGCCAGCGTCTACAGT
A-Key 2OS-F1-16GAAGTTCCAGCCAGCGTCTTGCGTTATG
A-Key 2OS-F1-17GAAGTTCCAGCCAGCGTCCGAGCGAGTG
A-Key 2OS-F1-18GAAGTTCCAGCCAGCGTCCGACTCTGTG
A-Key 2OS-F1-19GAAGTTCCAGCCAGCGTCATCCGTCCTG
A-Key 2OS-F1-20GAAGTTCCAGCCAGCGTCTTAAACGATG
A-Key 2OS-F1-21GAAGTTCCAGCCAGCGTCTAGCTTTTTG
A-Key 2OS-F1-22GAAGTTCCAGCCAGCGTCCACATGTATG
A-Key 2OS-F1-23GAAGTTCCAGCCAGCGTCGATAGCCATG
A-Key 2OS-F1-24GAAGTTCCAGCCAGCGTCACCTGTTATG
A-Key 2OS-F1-25GAAGTTCCAGCCAGCGTCTGCGAATTTG
A-Key 2OS-F1-26GAAGTTCCAGCCAGCGTCGTACATTTTG
A-Key 2OS-F1-27GAAGTTCCAGCCAGCGTCCTATTGCATG
A-Key 2OS-F1-28GAAGTTCCAGCCAGCGTCACGATACATG
A-Key 2OS-F1-29GAAGTTCCAGCCAGCGTCCTTAGCGCTG
A-Key 2OS-F1-30GAAGTTCCAGCCAGCGTCCGGAAACCTG
A-Key 2OS-F1-31GAAGTTCCAGCCAGCGTCGATGTTGGTG
A-Key 2OS-F1-32GAAGTTCCAGCCAGCGTCATCGGCGTTG
A-Key 2OS-F1-33GAAGTTCCAGCCAGCGTCTAGTACGATG
A-Key 2OS-F1-34GAAGTTCCAGCCAGCGTCGACGTGATTG
A-Key 2OS-F1-35GAAGTTCCAGCCAGCGTCTGAGCCAATG
A-Key 2OS-F1-36GAAGTTCCAGCCAGCGTCCCTCGCAGTG
A-Key 2OS-F1-37GAAGTTCCAGCCAGCGTCAGATCCAGTG
A-Key 2OS-F1-38GAAGTTCCAGCCAGCGTCTTGGCTGATG
A-Key 2OS-F1-39GAAGTTCCAGCCAGCGTCGACCGCTATG
A-Key 2OS-F1-40GAAGTTCCAGCCAGCGTCGAGCTTAATG
A-Key 2OS-F1-41GAAGTTCCAGCCAGCGTCGGACTGGTTG
A-Key 2OS-F1-42GAAGTTCCAGCCAGCGTCTGGGAGTCTG
A-Key 2OS-F1-43GAAGTTCCAGCCAGCGTCGCGATGGCTG
A-Key 2OS-F1-44GAAGTTCCAGCCAGCGTCACTTGGTTTG
A-Key 2OS-F1-45GAAGTTCCAGCCAGCGTCATACTCATTG
A-Key 2OS-F1-46GAAGTTCCAGCCAGCGTCTAGTGTCCTG
A-Key 2OS-F1-47GAAGTTCCAGCCAGCGTCGCATATAATG
A-Key 2OS-F1-48GAAGTTCCAGCCAGCGTCGGCGATTGTG
A-Key 2OS-F1-49GAAGTTCCAGCCAGCGTCGGGCTGTATG
A-Key 2OS-F1-50GAAGTTCCAGCCAGCGTCCGCTATTTTG
A-Key 2OS-F1-51GAAGTTCCAGCCAGCGTCGTACTGCATG
A-Key 2OS-F1-52GAAGTTCCAGCCAGCGTCTGTCTATGTG
A-Key 2OS-F1-53GAAGTTCCAGCCAGCGTCCGAATCACTG
A-Key 2OS-F1-54GAAGTTCCAGCCAGCGTCCGTCCTAATG
A-Key 2OS-F1-55GAAGTTCCAGCCAGCGTCACAAATGGTG
A-Key 2OS-F1-56GAAGTTCCAGCCAGCGTCTCTACTTTTG
A-Key 2OS-F1-57GAAGTTCCAGCCAGCGTCAATTCGAGTG
A-Key 2OS-F1-58GAAGTTCCAGCCAGCGTCGAACTCGGTG
A-Key 2OS-F1-59GAAGTTCCAGCCAGCGTCGCGGACGCTG
A-Key 2OS-F1-60GAAGTTCCAGCCAGCGTCCTTGTCCATG
A-Key 2OS-F1-61GAAGTTCCAGCCAGCGTCGTCGCGGTTG
A-Key 2OS-F1-62GAAGTTCCAGCCAGCGTCCAGGTCGTTG
A-Key 2OS-F1-63GAAGTTCCAGCCAGCGTCTCTCATCCTG
A-Key 2OS-F1-64GAAGTTCCAGCCAGCGTCGCTTCGTGTG
A-Key 2OS-F1-65GAAGTTCCAGCCAGCGTCCGTGATAATG
A-Key 2OS-F1-66GAAGTTCCAGCCAGCGTCTTGCTCACTG
A-Key 2OS-F1-67GAAGTTCCAGCCAGCGTCCGCTCTCCTG
A-Key 2OS-F1-68GAAGTTCCAGCCAGCGTCATTCTACTTG
A-Key 2OS-F1-69GAAGTTCCAGCCAGCGTCAAGGCGTTTG
A-Key 2OS-F1-70GAAGTTCCAGCCAGCGTCGCGGGATTTG
Primer nameIon Torrent region (A Key)
A-Key 2OS-F1-1CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-2CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-3CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-4CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-5CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-6CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-7CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-8CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-9CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-10CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-11CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-12CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-13CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-14CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-15CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-16CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-17CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-18CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-19CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-20CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-21CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-22CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-23CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-24CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-25CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-26CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-27CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-28CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-29CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-30CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-31CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-32CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-33CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-34CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-35CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-36CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-37CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-38CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-39CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-40CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-41CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-42CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-43CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-44CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-45CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-46CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-47CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-48CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-49CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-50CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-51CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-52CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-53CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-54CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-55CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-56CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-57CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-58CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-59CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-60CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-61CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-62CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-63CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-64CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-65CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-66CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-67CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-68CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-69CCATCTCATCCCTGCGTGTCTCCGACTCAG
A-Key 2OS-F1-70CCATCTCATCCCTGCGTGTCTCCGACTCAG

TABLE 14
Listing of the 175 combinations of peptide-HLA Binding Molecules
and the respective 2OS DNA-barcodes that were used to encode the given
specificity of all Detection Molecules applied in experiments 7.
168 of the peptide-ligands are associated with melanoma while the remaining 7
are different virus specific peptides. All are HLA-A0201 peptide antigens.
BarcodeHLA
2OSallelePeptideSequence
A7B1HLA-A0201707-APRVAALARDAP
A7B2HLA-A0201ATIC (AICRT)RLDFNLIRV
A7B3HLA-A0201ATIC (AICRT)MVYDLYKTL
A7B4HLA-A0201BA46 (MFGE8)NLFETPVEA
A7B5HLA-A0201BA46 (MFGE8)GLQHWVPEL
A7B6HLA-A0201Bcl-2PLFDFSWLSL
A7B7HLA-A0201Bcl-2WLSLKTLLSL
A7B8HLA-A0201Bcl-xLYLNDHLEPWI
A7B9HLA-A0201BING-4CQWGRLWQL
A7B10HLA-A0201B-RAFLATEKSRWSG
A7B11HLA-A0201cyclophilin B (Cyp-B)VLEGMEVV
A7B12HLA-A0201Cadherin 3/P-cadherinFILPVLGAV
A7B13HLA-A0201Cadherin 3/P-cadherinFIIENLKAA
A7B14HLA-A0201CDCA1/NUF2YMMPVNSEV
A7B15HLA-A0201CDCA1/NUF2KLATAQFKI
A7B16HLA-A0201CDK4ACDPHSGHFV
A7B17HLA-A0201CML28 (EXOSC5)ALVDAGVPM
A7B18HLA-A0201COA-1 (UBXN11)FMTRKLWDL
A7B19HLA-A0201COA-1 (UBXN11)RLLASLQDL
A7B20HLA-A0201CPSFKVHPVIWSL
A7B21HLA-A0201CPSFLMLQNALTTM
A7B22HLA-A0201Cyclin 1LLDRFLATV
A7B23HLA-A0201cyclin B1AGYLMELCC
A7B24HLA-A0201cyclin B1AKYLMELTM
A8B1HLA-A0201B-RAFLATEKSRWS
A8B2HLA-A0201cyclophilin B (Cyp-B)KLKHYGPGWV
A8B3HLA-A0201DAM-6, -10 (MAGE-B1,FLWGPRAYA
-B2)
A8B4HLA-A0201EphA2IMNDMPIYM
A8B5HLA-A0201EphA2VLAGVGFFI
A8B6HLA-A0201EphA2VLLLVLAGV
A8B7HLA-A0201EphA2TLADFDPRV
A8B8HLA-A0201EZH2FINDEIFVEL
A8B9HLA-A0201EZH2FMVEDETVL
A8B10HLA-A0201GnTVVLPDVFIRCV
A8B11HLA-A0201gp100/Pme117YLEPGPVTA
A8B12HLA-A0201gp100/Pme117LLDGTATLRL
A8B13HLA-A0201gp100/Pme117ITDQVPFSV
A8B14HLA-A0201gp100/Pme117VLYRYGSFSV
A8B15HLA-A0201gp100/Pme117RLMKQDFSV
A8B16HLA-A0201gp100/Pme117RLPRIFCSC
A8B17HLA-A0201gp100/Pme117AMLGTHTMEV
A8B18HLA-A0201gp100/Pme117SLADTNSLAV
A8B19HLA-A0201gp100/Pme117KTWGQYWQV
A8B20HLA-A0201HERV-K-MELMLAVISCAV
A8B21HLA-A0201hsp70LLLLDVAPL
A8B22HLA-A0201IDO1ALLEIASCL
A8B23HLA-A0201LAGE-1MLMAQEALAFL
A8B24HLA-A0201Livin (ML-IAP)RLASFYDWLP
A9B1HLA-A0201Livin (ML-IAP)SLGSPVLGL
A9B2HLA-A0201Livin (ML-IAP)QLCPICRAPV
A9B3HLA-A0201M2BPRIDITLSSV
A9B4HLA-A0201MAGE-A1KVLEYVIKV
A9B5HLA-A0201GnTVVLPDVFIRC
A9B6HLA-A0201gp100/Pme117IMDQVPFSV
A9B7HLA-A0201gp100/Pme117MLGTHTMEV
A9B8HLA-A0201hsp70LLDVAPLSL
A9B9HLA-A0201MAGE-A10GLYDGMEHL
A9B10HLA-A0201MAGE-A2LVHFLLLKY
A9B11HLA-A0201MAGE-A2LVQENYLEY
A9B12HLA-A0201MAGE-A2YLQLVFGIEV
A9B13HLA-A0201MAGE-A2KMVELVHFL
A9B14HLA-A0201MAGE-A3LVFGIELMEV
A9B15HLA-A0201MAGE-A4GVYDGREHTV
A9B16HLA-A0201MAGE-A1YLEYRQVPV
A9B17HLA-A0201MAGE-A8GLMDVQIPT
A9B18HLA-A0201MAGE-A8KVAELVRFL
A9B19HLA-A0201MAGE-A9ALSVMGVYV
A9B20HLA-A0201MAGE-C2ALKDVEERV
A9B21HLA-A0201MAGE-C2LLFGLALIEV
A9B22HLA-A0201MAGE-C2VIWEVLNAV
A9B23HLA-A0201MAGE-C2TLDEKVAELV
A9B24HLA-A0201MAGE-C2KVLEFLAKL
A10B1HLA-A0201MAGE-A3KVAELVHFL
A10B2HLA-A0201MC1RTILLGIFFL
A10B3HLA-A0201Melan-A/MART-1ELAGIGILTV
A10B4HLA-A0201Melan-A/MART-1ILTVILGVL
A10B5HLA-A0201Meloe-1TLNDECWPA
A10B6HLA-A0201MG50CMHLLLEAV
A10B7HLA-A0201MG50VLSVNVPDV
A10B8HLA-A0201NY-ESO-1/LAGE-2QLSLLMWIT
A10B9HLA-A0201NY-ESO-1/LAGE-2SLLMWITQCFL
A10B10HLA-A0201P PolypeptideIMLCLIAAV
A10B11HLA-A0201p53VVPCEPPEV
A10B12HLA-A0201p53KTCPVQLWV
A10B13HLA-A0201p53RMPEAAPPV
A10B14HLA-A0201p53LLPENNVLSPV
A10B15HLA-A0201p53KLCPVQLWV
A10B16HLA-A0201p53SMPPPGTRV
A10B17HLA-A0201p53LLGRNSFEV
A10B18HLA-A0201p53GLAPPQHLIRV
A10B19HLA-A0201p53SLPPPGTRV
A10B20HLA-A0201p53YLGSYGFRL
A10B21HLA-A0201PGK1IIGGGMAFT
A10B22HLA-A0201PRAMEALYVDSLFFL
A10B23HLA-A0201PRAMESLLQHLIGL
A10B24HLA-A0201SOX10SAWISKPPGV
A11B1HLA-A0201PRAMESLYSFPEPEA
A11B2HLA-A0201PRAMEVLDGLDVLL
A11B3HLA-A0201PRDX5LLLDDLLVSI
A11B4HLA-A0201NY-ESO-1/LAGE-2SLLMWITQC
A11B5HLA-A0201RAB38/NY-MEL-1VLHWDPETV
A11B6HLA-A0201RAGE-1LKLSGVVRL
A11B7HLA-A0201RAGE-1PLPPARNGGL
A11B8HLA-A0201Replication protein AYLMDTSGKV
A11B9HLA-A0201SART-3LLQAEAPRL
A11B10HLA-A0201SART-3RLAEYQAYI
A11B11HLA-A0201secernin 1KMDAEHPEL
A11B12HLA-A0201SOX10AWISKPPGV
A11B13HLA-A0201SSX-2RLQGISPKI
A11B14HLA-A0201SSX-2KASEKIFYV
A11B15HLA-A0201STAT1-alpha/11KLQELNYNL
A11B16HLA-A0201STEAP1FLYTLLREV
A11B17HLA-A0201STEAP1LLLGTIHAL
A11B18HLA-A0201STEAP1MIAVFLPIV
A11B19HLA-A0201SurvivinLMLGEFLKL
A11B20HLA-A0201SurvivinELTLGEFLKL
A11B21HLA-A0201SurvivinTLPPAWQPFL
A11B22HLA-A0201TAG-1SLGWLFLLL
A11B23HLA-A0201TelomeraseRLFFYRKSV
A11B24HLA-A0201TRP-2VYDFFW/LHY
A12B1HLA-A0201NY-ESO-1/LAGE-2SLLMWITQA
A12B2HLA-A0201TelomeraseRLVDDFLLV
A12B3HLA-A0201TelomeraseILAKFLHWL
A12B4HLA-A0201Topoisomerase IIFLYDDNQRV
A12B5HLA-A0201TRAG-3ILLRDAGLV
A12B6HLA-A0201TRP-2FW/LHYYSV
A12B7HLA-A0201TRP-2SLDDYNHLV
A12B8HLA-A0201TRP-2TLDSQVMSL
A12B9HLA-A0201TRP-2SVYDFFVWL
A12B10HLA-A0201TRP2-6bATTNILEHY
A12B11HLA-A0201tyrosinaseCLLWSFQTSA
A12B12HLA-A0201tyrosinaseMLLAVLYCL
A12B13HLA-A0201tyrosinaseYMDGTMSQV
A12B14HLA-A0201XBP-1LLSGQPASA
A12B15HLA-A0201MG50LLLEAVPAV
A12B16HLA-A0201MG50TLKCDCEIL
A12B17HLA-A0201MG50WLPKILGEV
A12B18HLA-A0201MG50RLGPTLMCL
A12B19HLA-A0201Meloe-2RCPPKPPLA
A12B20HLA-A0201PRDX5AMAPIKVRL
A12B21HLA-A0201cyclin B1ILIDWLVQV
A12B22HLA-A0201Melan-A/MART-1EAAGIGILTV
A12B23HLA-A0201adipophilinSVASTITGV
A12B24HLA-A0201alpha-actinin-4FIASNGVKLV
A13B1HLA-A0201Meloe-2RLPPKPPLA
A13B2HLA-A0201CDKN1ALMAGCIQEA
A13B3HLA-A0201CDKN1AGLGLPKLYL
A13B4HLA-A0201CDKN1AFAWERVRGL
A13B5HLA-A0201CLP (coactosin-like protein)NLVRDDGSAV
A13B6HLA-A0201CLP (coactosin-like protein)RLFAFVRFT
A13B7HLA-A0201CLP (coactosin-like protein)VVQNFAKEFV
A13B8HLA-A0201c-METYVDPVITSI
A13B9HLA-A0201CYP1B1WLQYFPNPV
A13B10HLA-A0201IMP-3NLSSAEVVV
A13B11HLA-A0201IMP-3RLLVPTQFV
A13B12HLA-A0201KIF20ALLSDDDVVV
A13B13HLA-A0201KIF20ACIAEQYHTV
A13B14HLA-A0201KIF20AAQPDTAPLPV
A13B15HLA-A0201MAGE-A10SLLKFLAKV
A13B16HLA-A0201MAGE-A12FLWGPRALV
A13B17HLA-A0201MAGE-C2FLAKLNNTV
A13B18HLA-A0201Melan-A/MART-1AAGIGILTV
A13B19HLA-A0201SurvivinQMFFCFKEL
A13B20HLA-A0201TelomeraseLLTSRLRFI
A13B21HLA-A0201TYMSLMALPPCHAL
A13B22HLA-A0201FLU MP 58-66 GILGILGFVFTL
A13B23HLA-A0201EBV LMP2 CLGCLGGLLTMV
A13B24HLA-A0201EBV BMF GLCGLCTLVAML
A14B1HLA-A0201cyclin D1LLGATCMFV
A14B2HLA-A0201HIV polTPRVTGGGAM
A14B3HLA-A0201EBV LMP2 FLYFLYALALLL
A14B4HLA-A0201CMV pp65 NLVNLVPMVATV
A14B5HLA-A0201EBV BRLF1 YVLYVLDHLIVV
A14B6HLA-A0201BAP31KLDVGNAEV
A14B7HLA-A0201CMV 1E1 VLEVLEETSVML

Example 20

This is an example where the Sample is a whole blood sample. Instead of being a mix of two PBMC donor materials as described in e.g. example 3 it is a mix of two whole blood samples. Except for the sample preparation the example is performed as example 3

Thus, the Linker is a dextrane conjugate with streptavidin and fluorocrome (PE Dextramer backbone from Immudex ApS).

The Binding Molecules are peptide-MHC (pMHC) complexes. In this example a panel of 110 labeled pMHC-multimers, constituting a library of Detection Molecules, are tested. Apart from 26 virus epitopes that are commonly found in healthy donors (derived from EBV, CMV, FLU and HPV), is included a number of polyomavirus capsid protein (VP1)-derived epitopes that has previously led to detection of T cells in healthy donors.

110 different Labels are generated as example 3. Detection Molecules are synthetized as in example 3.

Sample, whole blood, was incubated with an amount of a library of detection molecules as described in example 3.

The cell-bound detection molecules are separated from the non-cell bound detection molecules as described in example 3.

FACS isolated cells were subjected to PCR amplification of the oligonucleotide label associated with the detection molecules bound to cells. Subsequent extensive sequencing of PCR products revealed the identity of Detection Molecules that bound to the T cells present in the sample.

    • 1. Sample preparation. The cell samples used in this experiment are obtained by mixing whole blood from two different donors to obtain a cell sample where a number of T-cell specificities are known prior to the experiment. Thus, the sensitivity of the method as well as the relevance of the results obtained in the experiment can be evaluated at the end of the experiment, by comparison with data obtained previously, using other methods but cell samples prepared from the same donors.
      • a. Acquiring sample: Blood is obtained from the Danish Blood Bank.
      • b. Modifying sample:
        • i. Blood is drawn into BD Vacutainer® Plus Plastic K2EDTA Tubes according to manufacturer's protocol.
        • ii. Anti-coagulated blood samples are diluted 1:1 in RPMI (RPMI 1640, GlutaMAX, 25 mM Hepes; gibco-Life technologies).
        • iii. The samples used in the experiment are obtained by mixing blood from a donor (e.g. BC260) with 5% of the T cells specific for HLA-B7/CMV pp65 TPR into blood from a donor (e.g. BC262) without HLA-B7/CMV pp65 TPR specific T cells in fivefold dilutions, creating seven samples (with 5%, 1%, 0.2%, 0.04%, 0.008%, 0.0016% and 0.00032% HLA-B7/CMV pp65 TPR-specific T cells). The CMV pp65 TPR negative sample preparation instead has a population of HLA-A11/EBV-EBNA4 specific T cells.
        • iv. The remaining of the example is as for example 3.
    • 2. Linker preparation: The linker used in this example is dextrane conjugate with streptavidin and fluorocrome (PE Dextramer backbone from Immudex) as described in 1.
    • 3. Binding molecules preparation: The 110 different Binding Molecules are prepared as in example 3.
    • 4. Label preparation: The 110 different DNA oligo Labels are prepared as in example 3.
    • 5. Detection molecules preparation: 110 different Detection Molecules are prepared as in example 3.
    • 6. Incubation of sample and detection molecules:
      • a. Amount of sample: 0.2 mL anti-coagulated blood diluted 1:1 in RPMI.
      • b. Amount of detection molecule: As for e.g. example 3.
      • c. Conditions: Sample is incubated with dasatinib (50 nM final concentration), 30 min, 37° C. Subsequently, sample and Detection Molecule is mixed and incubated as e.g. example 3.
    • 7. Enrichment of detection molecules with desired characteristics: Detection Molecules are isolated with their associated cells by FACS as described in example 3.
    • 8. Identification of enriched detection molecule: By identifying the Label (in this Example, the oligonucleotide label), the pMHCs that bound separated cells can be identified. Therefore, the oligonucleotide labels that were comprised within the Detection Molecule that were recovered with the cells, were sequenced. This allowed the identification of pMHCs that bound cells of the cell sample. Labels associated with FACS isolated cells are sequenced as described in example 3.

Example 21

This is an example where the Sample is mononuclear cells derived from bone marrow (BM MNCs). Instead of being a mix of two PBMC donor materials as described in example 3 it is a mix of two whole blood samples. Except for the sample preparation the example is performed as example 3.

Thus, the Linker is a dextrane conjugate with streptavidin and fluorocrome (PE Dextramer backbone from Immudex).

The Binding Molecules are peptide-MHC (pMHC) complexes. In this example a panel of 110 labeled pMHC-multimers, constituting a library of Detection Molecules, are tested. Apart from 26 virus epitopes that are commonly found in healthy donors (derived from EBV, CMV, FLU and HPV), is included a number of polyomavirus capsid protein (VP1)-derived epitopes that has previously led to detection of T cells in healthy donors.

110 different Labels are generated as in example 3. Detection Molecules are synthetized as in example 3.

Sample, whole blood, was incubated with an amount of a library of detection molecules as described in example 3.

The cell-bound detection molecules are separated from the non-cell bound detection molecules as described in example 3.

FACS isolated cells were subjected to PCR amplification of the oligonucleotide label associated with the detection molecules bound to cells. Subsequent extensive sequencing of PCR products revealed the identity of Detection Molecules that bound to the T cells present in the sample.

    • 1. Sample preparation. The cell samples used in this experiment was obtained by mixing bone marrow from two different donors to obtain a cell sample where a number of T-cell specificities were known prior to the experiment. Thus, the sensitivity of the method as well as the relevance of the results obtained in the experiment could be evaluated at the end of the experiment, by comparison with data obtained previously, using other methods but cell samples prepared from the same donors.
      • a. Acquiring sample: Collect bone marrow from the upper iliac crest or the sternum by using an aspiration needle.
      • b. Modifying sample:
        • i. Dilute aspirated human bone marrow at a ratio of 7:1 with buffer (phosphate buffered saline (PBS), pH 7.2, and 2 mM EDTA), e.g., dilute 30 mL of bone marrow with 5 mL of buffer to a final volume of 35 mL.
        • ii. Pass cells through a 100 μm filter to remove bone fragments and cell clumps.
        • iii. Carefully layer 35 mL of diluted cell suspension over 15 mL of Ficoll-Paque in a 50 mL conical tube. Centrifuge at 445×g for 35 minutes at 20° C. in a swinging bucket rotor without brake. Aspirate the upper layer leaving the mononuclear cell layer undisturbed at the interphase. Carefully transfer the BM MNCs at the interphase to a new 50 mL conical tube. Wash cells by adding up to 40 mL of buffer, mix gently and centrifuge at 300×g for 10 minutes at 20° C. Carefully remove supernatant completely. For removal of platelets, resuspend the cell pellet in 50 mL of buffer and centrifuge at 200×g for 10-15 minutes at 20° C. Carefully remove the supernatant completely. Resuspend cell pellet in 5 mL buffer for downstream applications.
        • iv. The samples used in the experiment is obtained by mixing BM MNCs from a donor (e.g. BC260) with 5% of the T cells specific for HLA-B7/CMV pp65 TPR into BM MNCs from a donor (e.g. BC262) without HLA-B7/CMV pp65 TPR specific T cells in fivefold dilutions, creating seven samples (with 5%, 1%, 0.2%, 0.04%, 0.008%, 0.0016% and 0.00032% HLA-B7/CMV pp65 TPR-specific T cells). The CMV pp65 TPR negative BM MNC preparation instead has a population of HLA-A11/EBV-EBNA4 specific T cells.
        • v. The remaining of the example is as for example 3.
    • 2. Linker preparation: The linker used in this example is dextrane conjugate with streptavidin and fluorocrome (PE Dextramer backbone from Immudex) as described in 1.
    • 3. Binding molecules preparation: The 110 different Binding Molecules are prepared as in example 3.
    • 4. Label preparation: The 110 different DNA oligo Labels are prepared as in example 3.
    • 5. Detection molecules preparation: 110 different Detection Molecules are prepared as in example 3.
    • 6. Incubation of sample and detection molecules:
      • a. Amount of sample: 0.2 mL anti-coagulated blood diluted 1:1 in RPMI.
      • b. Amount of detection molecule: As for example 3.
      • c. Conditions: Sample is incubated with dasatinib (50 nM final concentration), 30 min, 37° C. Subsequently, sample and Detection Molecule is mixed and incubated as e.g. example 3.
    • 7. Enrichment of detection molecules with desired characteristics: Detection Molecules are isolated with their associated cells by FACS as described in example 3.
    • 8. Identification of enriched detection molecule: By identifying the Label (in this Example, the oligonucleotide label), the pMHCs that bound separated cells can be identified. Therefore, the oligonucleotide labels that were comprised within the Detection Molecule that were recovered with the cells, were sequenced. This allowed the identification of pMHCs that bound cells of the cell sample. Labels associated with FACS isolated cells are sequenced as described in example 3.

Example 60

This is an example where the binding molecules (BM) are MHC-like antigen-presenting molecules such as e.g. CD1a, CD1b, CD1c and CD1d.

The Linker used in this example is dextran conjugate with streptavidin and fluorocrome (PE Dextramer backbone from Immudex), the label used is a DNA oligonucleotide and the Sample is PBMC's.

Isolation and identification of detection molecules capable of binding to cells of the cell sample is done by FACS of cells with PE labeled dextran conjugate, and the identity and amount of associated labels are determined by DNA sequencing.

    • 1. Sample preparation.
    • The sample used in this example can be any type of cell sample, for example one PBMC sample from a human being, as described in Example 1.
    • 2. Linker preparation:
    • The linker used in this experiment is a dextran molecule, prepared as described in Example 1.
    • 3. Binding molecules preparation
      • a. Synthesis: CD1a, CD1b, CD1c and CD1d is produced as described by Khurana A, et. al, J Vis Exp. 2007; (10): 556. and as performed by a person skilled in the art.
      • b. Modification: CD1a, CD1b, CD1c and CD1d is loaded with 15 potential lipid antigens (GMM, glucose monomycolate; Sulfolipid, diacylated sulfoglycolipid; PIM's, phosphatidylinositol mannosides; Man-LAM, mannosylated lipoarabinomannan; MPM, mannosyl-b1-phosphomycoketide; MPP, mannosyl-b1-phosphoheptaprenol; DDM, dideoxymycobactin; GSL-1, a-glucoronsylceramide; GaIDAG, a-galactosyldiacylglycerol; LPG, lipophosphoglycan; PI, phosphatidylinositol; PG, phosphatidylglycerol; PE, phosphatidylethanolamine; iGb3, isoglobotrihexosylceramide; Alpha-GC, a-galactosylceramide). Lipids are dissolved to 2 mg/mL in DMSO, heated to 50° C. for 2 min and diluted further 1:10 in PBS+0.1% Tween20. Lipids are mixed individually with the four different CD1 molecules giving rise to 4×15=60 combinations. Briefly 10 uL 1 ug/uL CD1 protein is mixed with 2 uL 0.2 mg mL lipid in PBS+0.1% Tween20+10% DMSO. Load lipids into CD1 4 h at 30° C.
      • c. Purification: The CD1a-lipid, CD1b-lipid, CD1c-lipid and CD1d-lipid complexes are not purified further.
    • 4. Label preparation: Labels as described in example 3 are used.
      • a. Synthesis: The first 60 Labels in table 8 are used.
      • b. Modification: no further
      • c. Purification: no further
    • 5. Detection molecules preparation
      • a. Synthesis: The detection molecules are prepared as in example 3 except that the 60 different CD1 and lipid combinations are mixed with 60 individual DNA oligonucleotide labels (labels 1-60 from table 8).
      • b. Modification: no further modifications
      • c. Purification: no further
    • 6. Incubation of sample and detection molecules
      • The sample and detection molecules of step 1 and 5, respectively, are mixed and incubated as described in Example 3.
    • 7. Enrichment of detection molecules with desired characteristic: Cells positive for PE fluorochrome on the Linker is isolated by FACS as described in example 3.
    • 8. Identification of enriched detection molecule: The detection molecules recovered in step 7 are identified by sequencing, as described in Example 3.

Example 61

This is an example where the binding molecules (BM) are MHC class II proteins. The Linker used in this example is dextran conjugate with streptavidin and fluorocrome (PE Dextramer backbone from Immudex), the label used is a DNA oligonucleotide and the Sample is PBMC's.

Isolation and identification of detection molecules capable of binding to cells of the cell sample is done by FACS of cells with PE labeled dextran conjugate, and the identity and amount of associated labels are determined by DNA sequencing.

    • 1. Sample preparation: The sample used in this example can be any type of cell sample, for example one PBMC sample from a human being, as described in Example 1.
    • 2. Linker preparation: The linker used in this experiment is a dextran molecule, prepared as described in Example 1.
    • 3. Binding molecules preparation
      • a. Synthesis: MHC class II protein in the form of biotinylated monomers are obtained from The NIH Tetramer Core Facility, at Emory University, US. The following eight MHC Class II monomers are used (DPB1*04:01 C. tetani TT 948-968 FNNFTVSFWLRVPKVSASHLE, DPB1*04:01 human MAGE3 243-258 KKLLTQHFVQENYLEY, DPB1*04:01 human oxytocinase 272-284 KKYFAATQFEPLA, DPB1*04:01 human CLIP 87-101 PVSKMRMATPLLMQA, DPB1*04:01 human CTAG1 157-170 SLLMWITQCFLPVF, DPB1*04:01 HIV env 31-45 TEKLVVVTVYYGVPVW, DQB1*03:02 human CLIP 87-101 PVSKMRMATPLLMQA, DQB1*03:02 human FcR2 104-119 QDLELSWNLNGLQADL
      • b. Modification: Monomers are obtained ready folded and biotinylated from The NIH Tetramer Core Facility.
      • c. Purification: No further modification
    • 4. Label preparation
      • a. Synthesis: The first 8 labels from Table 8 are used
      • b. Modification: No further
      • c. Purification: No further
    • 5. Detection molecules preparation
      • a. Synthesis: The detection molecules are prepared as in example 3 except that the 8 different MHC class II molecules are combined with 8 different DNA oligonucleotide labels as described in example 3.
      • b. Modification: No further
      • c. Purification: No further
    • 6. Incubation of sample and detection molecules: The sample and detection molecules of step 1 and 5, respectively, are mixed and incubated as described in Example 3.
    • 7. Enrichment of detection molecules with desired characteristics: Cells positive for PE fluorochrome on the Linker is isolated by FACS as described in example 5.
    • 8. Identification of enriched detection molecule: The detection molecules recovered in step 7 are identified by sequencing, as described in Example 3.

Example 80

In this example, the labels used are DNA oligonucleotides of different length. The identity of the individual label is based on its mass, by either mass spectrometry or differential migration in a gel electrophoresis analysis.

    • 1. Sample preparation.

Blood comprising different kinds of blood cells is drawn from a person, and used directly in the incubation, step E below. Alternatively, any type of sample described in the examples above can be used.

    • 3. Binding molecules preparation

The binding molecules used in this study are 10 different antibodies, recognizing 10 different cell differentiation markers (CDs) respectively, namely CD3, CD4, CD8, CD16, CD56, . . . **. Each of these are commercially available.

    • 4. Label preparation

10 DNA oligonucleotides of length 50 nt, 100 nt, 150 nt, 200 nt, 250 nt, 300 nt, 350 nt, 400 nt, 450 nt, and 500 nt, respectively, with a terminal N-succinimidyl ester moiety are prepared by standard means.

    • 5. Detection molecules preparation

Each of the 10 antibodies mentioned under (B) are incubated in appropriate buffer (e.g. PBS pH 8) together with one of the 10 oligonucleotides mentioned in step (C) above, allowing the N-succinimidyl ester to react with free amines on the surface of the antibody, to form a covalent link between the antibody and the oligonucleotide. In this way, 10 detection molecules are generated, each of which comprise one specific antibody (e.g. anti-CD3 antibody) and one specific oligonucleotide (e.g. 50 nt DNA oligo).

    • 6. Incubation of sample and detection molecules

The sample of step (A) is incubated with the 10 detection molecules under appropriate conditions (e.g. appropriate buffer, e.g. Tris pH 7.5 is added, or the oligonucleotides from step (D) in e.g. PBS pH 8 is simply mixed with sample without further addition of buffer. Incubation can be at 0, 4, 10, 20, 30, or 40 degrees celcius.

    • 7. Isolating bound and unbound detection molecules

The incubation mixture of step (E) is centrifuged, to recover all cells of the sample. Supernatant is removed, the cells resuspended and buffer added. Centrifugation is repeated 1-3 times, and the cells finally resuspended. The suspension of cells will contain detection molecules that are capable of binding to one or more cells. Thus, the detection molecules of the suspension are representative of the distribution of receptors on the cell surface of the cells of the original sample.

    • 8. Determining the identity and amount of the recovered detection molecules.

1 μL of the mixture of cells and bound detection molecules are transferred to a 100 μL PCR reaction comprising forward and reverse primers that can anneal to each of the 10 oligonucleotide labels described in step (C) above, and comprising all other components for an efficient PCR reaction. The PCR reaction is performed under standard conditions, and aliquots are taken out after 25, 30 and 35 cycles are performed.

The PCR product is applied to a gel capable of resolving the individual oligonucleotide fragments (e.g. a 2.5% agarose gel), and electrophoresis is performed. Once the fastest moving product (corresponding to the 50 nt oligonucleotide) has migrated about ¾ of the gel length, electrophoresis is terminated. The position of a band in the gel reflects its size and therefore identifies the corresponding oligonucleotide label; the uppermost band represents the largest fragment (500 nt), and the lowermost band represents the smallest fragment (50 nt). The intensity of each of the 10 bands, corresponding to each of the 10 different oligonucleotide fragments, is indicative of the relative amount recovered of each oligonucleotide label and therefore, of each detection molecule.

Example 81

This example is as example 80, except that the labels used here are PNA fragments rather than DNA oligonucleotides, and the identity of the labels of the recovered detection molecules are determined by mass spectrometry analysis rather than gel electrophoresis.

Sample and binding molecules are as described in example 80.

Labels are prepared as follows. 10 different PNAs are prepared, of different size (e.g. comprising 5, 7, 9, 11, 13, 15, 17, 19, 21, and 23 bases). During synthesis, N-succinimidyl ester is introduced at one of the PNA fragment. Further, also during its synthesis, a disulfide bond (S—S) is introduced between the ester and the rest of the PNA fragment. Thus, in the label a disulfide bond links the N-succinimidyl ester with the remainder of the PNA fragment.

Detection molecules are prepared as in example 80, except that the PNA labels are used instead of the DNA oligonucleotide labels of example 80. Each of the resulting 10 detection molecules therefore consists of a specific binding molecule (a specific antibody, e.g. CD3) and a specific PNA label (e.g. PNA comprising 5 bases). Isolation of detection molecules capable of binding to cells of the sample is done by centrifugation as described in example 80, and the final cell suspension thus contains cells plus detection molecules capable of binding to cells of the sample.

Finally, the identity and amount of the recovered detection molecules is determined in the following way: First, the PNA labels are cleaved off from the detection molecules (and therefore, released from the cells that the detection molecule is bound to) by addition of DTT, which cleaves the disulfide bond. The cells are spun down by centrifugation, and the supernatant (comprising the released PNA labels but not the cells) is then subjected to a mass spectrometry analysis.

The mass spectrometry analysis will reveal the relative amount of each of the 10 PNA labels (corresponding to the relative amount of each of the 10 detection molecules recovered by centrifugation of the cells).

Example 82

This example is as example 81, except that the labels used here are peptide fragments rather than PNA fragments

Labels are prepared as follows. 10 different peptides are prepared, of different size (e.g. comprising 5, 7, 9, 11, 13, 15, 17, 19, 21, and 23 amino acids). During synthesis, N-succinimidyl ester is introduced at one of the peptide fragment. Further, also during its synthesis, a disulfide bond (S—S) is introduced between the ester and the rest of the peptide fragment. Thus, in the label a disulfide bond links the N-succinimidyl ester with the remainder of the peptide fragment.

Detection molecules are prepared as in example 81, except that the peptide labels are used instead of the PNA labels of example 81. Each of the resulting 10 detection molecules therefore consist of a specific binding molecule (a specific antibody, e.g. CD3) and a specific peptide label (e.g. peptide comprising 5 amino acids).

The mass spectrometry analysis will reveal the relative amount of each of the 10 peptide labels (corresponding to the relative amount of each of the 10 detection molecules recovered by centrifugation of the cells).

Example 120

This is an example where the samples are three buffy coats (BC's), the Linker is PE and streptavidin conjugated dextran, and the Binding Molecules are biotinylated peptide-MHC (pMHC) complexes. The Labels are synthetic oligonucleotides modified with a terminal biotin capture-tag. The labels are combined oligonucleotide label arising by annealing an A oligonucleotide (modified with biotin) to a partially complimentary B oligonucleotide label followed by enzymatic DNA polymerase extension of Oligo A and Oligo B to create a fully double stranded label.

The Detection Molecules are created by combining biotinylated pMHC and labels in the form of biotin-modified oligonucleotide onto a streptavidin-modified dextran linker. The detection molecule further contained a fluorochrome (PE). 30 different Detection Molecules are generated wherein the individual detection molecules containing different pMHC are encoded by corresponding individual oligonucleotide labels. Two versions (Set 1 and Set 2, see table x) of each of the 30 different Detection Molecules are created in the way that all Detection molecules are generated in two forms with different labels. Three samples, PBMC's, were incubated with an amount of mixed Detection Molecules.

In this example CD8+ T cell-bound Detection Molecules are separated from the non-cell bound Detection Molecules by capture of CD8+ T cells using anti CD8-coated magnetic Dynabeads. Magnetic bead-isolated CD8-positive T cells are subjected to PCR amplification of associated Label and the PCR product is analyzed as a read of the oligonucleotide label associated with the detection molecules bound to the isolated cells thus revealing the identity of detection molecules bound to the T cells present in the sample.

    • 1. Sample preparation.
      • a. Acquiring sample: The cell sample used in this experiment is obtained by preparing PBMC's from blood drawn from three donors (D131, D149 and D158) that, by conventional MHC-multimer staining, are characterized for their specific T cells towards a number of virus antigens.
      • b. Modifying sample: The PBMC's are prepared as described in example 1.
    • 2. Linker preparation: The Linker is PE and streptavidin conjugated dextran (Dextramer from Immudex) which is prepared as in example 1.
    • 3. Binding molecules preparation: Binding Molecules are biotinylated pMHC complexes with peptide/HLA combination as described in table below

LabelsLabels
set 1set 2 HLAAntigenPeptide
2OS-A1-B22OS-A1-B12A2CMV pp65NLVPMVATV
2OS-A1-B32OS-A1-B13A2EBV-E3BLLDFVRFMGV
2OS-Al-B42OS-A1-B14A2HPV E6TIHDIILECV
2OS-Ai-B52OS-A1-B15A2HSV-1gBRMLGDVMAV
2OS-Al-B62OS-A1-B16A2Neg ControlALIAPVHAV
2OS-A2-B22OS-A2-B12A2CMV-IE1VLEETSVML
2OS-A2-B32OS-A2-B13A2EBV-BALF4FLDKGTYTL
2OS-A2-B42OS-A2-B14A2FLUGILGFVFTL
2OS-A2-B52OS-A2-B15A2MART-1ELAGIGILTV
2OS-A2-B62OS-A2-B16A3Cancer-gp100ALLAVGATK
2OS-A3-B22OS-A3-B12A2EBV-EBMAC3CLGGLLTMV
2OS-A3-B32OS-A3-B13A2EBV-BMRF1TLDYKPLSV
2OS-A3-B42OS-A3-B14A2FLU-BNPKLGEFYNQMM
2OS-A3-B52OS-A3-B15A2HIV-gagSLYNTVATL
2OS-A3-B62OS-A3-B16B07Neg ControlGPAESAAGL
2OS-A4-B22OS-A4-B12A2EBV-BMLF1GLCTLVAML
2OS-A4-B32OS-A4-B13A2EBV-BRLF1YVLDHLIVV
2OS-A4-B42OS-A4-B14A2HSV1gBGIFEDRAPV
2OS-A4-B52OS-A4-B15A2HIVILKEPVHGV
2OS-A4-B62OS-A4-B16B08Neg ControlAAKGRGAAL
2OS-A5-B22OS-A5-B12A2EBV-LMP1YLLEMLWRL
2OS-A5-B32OS-A5-B13A2HPV-E7YMLDLQPETT
2OS-A5-B42OS-A5-B14A2Hsv1gBYLANGGFLI
2OS-A5-B52OS-A5-B15A2HIVFLGKIWPS
2OS-A5-B62OS-A5-B16H2KbNeg ControlAYAGSAGSI
2OS-A6-B22OS-A6-B12A2EBV-LMP1YLQQNVWVTL
2OS-A6-B32OS-A6-B13A2HPV-E7LLMGTLGIVC
2OS-A6-B42OS-A6-B14A2HSV-U125FLWEDQTLL
2OS-A6-B52OS-A6-B15A1Neg ControlSTEGGGLAY
2OS-A6-B62OS-A6-B16Neg Controlno peptide
    • 4. Label preparation: Label is generated as described in example 4 except that in this example only 2×60 labels are used (See table above)
    • 5. Detection molecules preparation
      • a. Synthesis
        • i. According to table below transfer the required volume Dextran conjugate to an Eppendorf tube. Centrifuge (10.000 g, 10 min, 4° C.).
        • ii. Transfer the supernatant containing dextrane conjugate without precipitates to 2*30 wells on two plates.
        • iii. In a 96 well format add the 2×30 Label oligos in 2:1 molar concentration to PE-Dextramer conjugate.
        • iv. Mix well and incubate 30 min, 4° C.
        • v. Thaw pMHC binding molecules on ice, dilute to 100 μg/mL in PBS and centrifuge (2000 g, 5 minutes, 18° C.).
        • vi. Transfer the pMHC monomers to wells of the 96 well plate according to the above table to allow the combination of pMHC with labeled linker.
        • vii. Mix well and incubate 30 minutes at r.t. and PBS subsequently.

Detection molecule preparationConcentration
Dextran conjugate16*10{circumflex over ( )}-8M
Oligo lable54.25*10{circumflex over ( )}-8M
Detection molecules/samples
Number of Detection molecules30
Number of samples20
Assembling Label and LinkerVolumes needed
1: Transfer Dextran to Eppendorf630μl
tube and centrifuge 10000 g, 5 min, 4 C.
2: Transfer the supernatant to wells20μl/well
on a plate according to setup
3: Add oligo labels to wells according11.8μl/well
to setup, Mix and incubate 30 min, 4 C.
Preparation of binding molecules:
1: Thaw pMHC monomers on ice
2: Dilute monomers in PBS
to 100 μg/ml = 2 μM
Vtotal needed per pMHC per plate40μl/pMHC
Preparation of Detection Molecules
1: Centrifuge pMHC monomers
2000 g, 10 min 18 C.
2: Transfer supernatant to the 96well26.4μl/well
plate
(To the Dextran-MHC control is an
equal volume of PBS added)
3: Mix well and incubate 30 min at RT
4: Add PBS1.8μl/well
5: Store at 4 C.
Vtotal in each well60.0
      • a. Modification: No further
      • b. Purification: No further
    • 6. Incubation of sample and detection molecules
      • a. Amount of sample: 2 million PBMC's per sample
      • b. Amount of detection molecule: According to table below. Each donor sample is incubated with 4 different amounts of Detection Molecule
      • c. Conditions:
        • i. PBMC's are thawed in 10 ml RPM1-10% FCS and washed twice in 2 mL RPMI-10% FCS. Cells are then washed in 2 mL PBS w. 0.5% BSA, 100 μg/ml Herring DNA, 2 mM EDTA. (All washing of cells in this experiment refers to centrifuge for 5 min at 800 g to collect cells followed by discarding of wash buffer)
        • ii. Resuspend cells in 500 μl PBS w. 0.5% BSA, 100 μg/ml Herring DNA, 2 mM EDTA to a concentration 20 million/ml.
        • iii. Add 25 μl 1 μM Dasatinib and incubate 30 min at 37° C. (Final Dasatinib conc. 50 nM).
        • iv. Centrifuge Detection Molecules (2000 g, 5 minutes, 18° C.)
        • v. Add 166.5 μl 10 μM biotin to an Eppendorf tube.
        • vi. Pool 25 μl of each Detection Molecule from each of the two sets into the Eppendorf tube (Detection Molecule library) Vtot=1666, 5 μl which is enough for 3 donors in 4 concentrations.
        • vii. Centrifuge the library of Detection Molecules (10000 g, 10 min) and transfer 1550 μl to a new tube avoiding aggregates.
        • viii. Transfer from new tube to 5 ml tubes according to setup/volumes below.
        • ix. To all twelve tubes add 100 μl cells (2 million cells) of the respective donors, to a VTot in each sample of 300 μl. Incubate 15 minutes at 37° C.
        • x. Wash cells twice in 2 ml PBS w. 0.5% BSA, 100 μg/ml Herring DNA, 2 mM EDTA (Centrifuge 5 min at 800 g)

3 μl1.5 μl0.75 μl0.375 μl
Sample/Detect
Mol
D131S1: 200 μlS2: 100 μlS3: 50 μlS4: 25 μl
D149S5: 200 μlS6: 100 μlS7: 50 μlS8: 25 μl
D158S9: 200 μlS10: 100 μlS11: 50 μlS12: 25 μl
PBS w.
0.5% BSA,
100 μg/ml
Herring DNA,
2 mM EDTA
D131S1: 0 μlS2: 100 μlS3: 150 μlS4: 175 μl
D149S5: 0 μlS6: 100 μlS7: 150 μlS8: 175 μl
D158S9: 0 μlS10: 100 μlS11: 150 μlS12: 175 μl
    • 7. Enrichment of detection molecules with desired characteristics
      • a. Apply: In this example Detection Molecules are isolated by capturing CD8+ cells using anti CD8 antibody coated magnetic beads (DYNABEADS CD8, #11147D, Life Technologies). Detection molecules associated with captured CD8+ cells are isolated. Dynabeads used according to manufacturer's protocol.
        • i. Briefly; Dynabeads are vortex >30 sec and transfer 50 μl/sample=600 μl to a 5 ml tube and add 1 ml PBS+0.1% BSA+2 mM EDTA. Re-suspended dynabeads are placed in magnet 1 min, supernatant is discarded and dynabeads are re-suspended in 600 μl PBS+0.1% BSA+2 mM EDTA.
        • ii. To all samples add 50 μl washed beads+950 μl Isolation buffer=>Vtot=1 ml. Incubate 20 min 4 C on a tilting plate.
      • b. Wash:
        • i. Place tubes in magnet (Dynal, Life Technologies) for 2 min and carefully remove supernatant
        • ii. Wash twice: Add 2 ml PBS+0.1% BSA+2 mM EDTA, vortex, place in magnet 2 min, remove supernatant.
        • iii. Re-suspend beads in 500 μl PBS w. 0.5% BSA, 100 μg/ml Herring DNA, 2 mM EDTA, transfer to Eppendorf tubes.
        • iv. Centrifuge tubes 5 min at 5000 g, remove supernatant final volume app. 20 μl→Store at −80 C or store on ice at 4 C O.N for PCR
      • c. Separate: Is done during washing.
    • 8. Identification of enriched detection molecule: Separated Detection Molecules are analyzed by PCR amplification of the attached labels followed by sequencing of the PCR products to reveal the identity of isolated detection molecules and thus the identity of those antigens for which specific T cells were present in the sample. The PCR amplification and sequencing of PCR product is done as for example 3 using the same sets of primers and the same sequencing service and sequencing de-convolution service.

Example 121

This is an example where the samples are PBMC's prepared as three buffy coats (BC's), the Linker is PE and streptavidin conjugated dextran, and the Binding Molecules are biotinylated peptide-MHC (pMHC) complexes. The Labels are synthetic oligonucleotides modified with a terminal biotin capture-tag.

In this example CD8+ T cell-bound Detection Molecules are separated from non-cell bound Detection Molecules by capture of CD8+ T cells by magnetic labeling of CD8+ cells with CD8 MicroBeads (CD8 MicroBeads, human, #130-045-201, Miltenyi, Germany). Magnetic bead-isolated CD8-positive T cells are subjected to PCR amplification of associated Label (as described in example 3) and the PCR product is analyzed as a read of the oligonucleotide label associated with the detection molecules bound to the isolated cells thus revealing the identity of detection molecules bound to the T cells present in the sample (as example 3).

    • 1. Sample preparation. The samples are PBMC's from three donor materials prepared as described in example 3.
    • 2. Linker preparation: The Linker is PE and streptavidin conjugated dextran (Dextramer from Immudex) which is prepared as in example 1.
    • 3. Binding molecules preparation: Binding Molecules are biotinylated pMHC complexes with peptide/HLA combination as described in example 3.
    • 4. Label preparation: Labels are biotin modified DNA oligonucleotides as described in example 3.
    • 5. Detection molecules preparation: Detection Molecules are prepared by combining 110 aliquots of Linker with individual Labels followed by adding pMHC Binding Molecules as described in Example 3.
    • 6. Incubation of sample and detection molecules: PBMC's are mixed with the 110 member library of Detection Molecules as described in example 3.
    • 7. Enrichment of detection molecules with desired characteristics:
      • a. Apply: In this example Detection Molecules are isolated by capturing CD8+ cells using anti CD8 antibody coated magnetic beads (CD8 MicroBeads, human, #130-045-201, Miltenyi, Germany). Detection molecules associated with captured CD8+ cells are isolated. CD8 MicroBeads are used according to manufacturer's protocol. Briefly;
        • i. Centrifuge cell suspensions at 300×g for 10 minutes. Aspirate supernatant completely.
        • ii. Resuspend cell pellets in 80 μL of buffer (phosphate-buffered saline (PBS), pH 7.2, 0.5% bovine serum albumin (BSA), and 2 mM EDTA).
        • iii. Add 20 μL of CD8 MicroBeads. Mix well and incubate for 15 minutes in the refrigerator (2-8)
        • iv. Wash cells by adding 1-2 mL of buffer and centrifuge at 300×g for 10 minutes. Aspirate supernatant completely. Resuspend cells in 500 μL of buffer.
        • v. Place an MS column (Miltenyi, Germany) in the magnetic field of a OctoMACS column stand Separator. Prepare column by rinsing with 500 μL of buffer:
        • vi. Apply cell suspension onto the column. Collect unlabeled cells that pass through and wash column with 500 μL of buffer. Collect total effluent; this is the unlabeled cell fraction. Perform washing steps by adding buffer three times. Only add new buffer when the column reservoir is empty.
        • vii. Remove column from the separator and place it on a suitable collection tube.
        • viii. Pipette 1 mL of buffer onto the column.
        • ix. Immediately flush out the magnetically labeled cells by firmly pushing the plunger into the column.
        • x. Centrifuge tubes 5 min at 500 g, remove supernatant. The final volume of app. 20 μl containing collected CD8+ cells and their associated Detection Molecules are store at −80 C for later analysis.
    • 8. Identification of enriched detection molecule: Separated cells and their associated Detection Molecules are analyzed by PCR amplification of the attached labels followed by sequencing of the PCR products to reveal the identity of isolated Detection Molecules and thus the identity of those antigens for which specific T cells were present in the sample. The identification is done as for example 3.

Example 122

This is an example where the samples are PBMC's prepared as three buffy coats (BC's), the Linker is PE and streptavidin conjugated dextran, and the Binding Molecules are biotinylated peptide-MHC (pMHC) complexes. The Labels are synthetic oligonucleotides modified with a terminal biotin capture-tag.

In this example, though, all cells and their bound Detection Molecules are first separated, by centrifugation, from the supernatant containing Detection Molecules not bound to cells. Secondly, remaining Detection Molecules are captured by their PE modification on the Linker using anti PE MicroBeads (anti PE MicroBeads, #130-048-801, Miltenyi, Germany). Magnetic bead-isolated cells are subjected to PCR amplification of Detection-Molecule associated Labels (as described in example 3) and the PCR product is analyzed as a read of the oligonucleotide label associated with the detection molecules bound to the isolated cells thus revealing the identity of detection molecules bound to the T cells present in the sample as in example 3.

    • 1. Sample preparation. The samples are PBMC's from three donor materials prepared as described in example 3.
    • 2. Linker preparation: The Linker is PE and streptavidin conjugated dextran (Dextramer from Immudex) which is prepared as in example 1.
    • 3. Binding molecules preparation: Binding Molecules are biotinylated pMHC complexes with peptide/HLA combination as described in example 3.
    • 4. Label preparation: Labels are biotin modified DNA oligonucleotides as described in example 3.
    • 5. Detection molecules preparation: Detection Molecules are prepared by combining 110 aliquots of Linker with individual Labels followed by adding pMHC Binding Molecules as described in Example 3.
    • 6. Incubation of sample and detection molecules: PBMC's are mixed with the 110 member library of Detection Molecules as described in example 3.
    • 7. Enrichment of detection molecules with desired characteristics
      • a. Apply: In this example Detection Molecules are isolated by centrifugation separation of cells and their bound Detection Molecules from the supernatant containing Detection Molecules not bound to cells followed by magnetic capture of cells with bound PE-labeled Detection Molecules thereby separating from cells without bound PE-labeled Detection Molecules using anti PE MicroBeads (anti PE MicroBeads, #130-048-801, Miltenyi, Germany). MicroBeads are used according to manufacturer's protocol. Briefly;
        • i. Centrifuge cell suspensions at 300×g for 10 minutes. Aspirate supernatant completely.
        • ii. Wash cells twice by adding 2 mL of buffer (phosphate-buffered saline (PBS), pH 7.2, 0.5% bovine serum albumin (BSA), and 2 mM EDTA) and centrifuge at 300×g for 10 minutes. Aspirate supernatant completely.
        • iii. Resuspend cell pellets in 80 μL of buffer.
        • iv. Add 20 μL of anti PE MicroBeads. Mix well and incubate for 15 minutes in the refrigerator (2-8)
        • v. Wash cells by adding 1-2 mL of buffer and centrifuge at 300×g for 10 minutes. Aspirate supernatant completely. Resuspend cells in 500 μL of buffer.
        • vi. Place an MS column (Miltenyi, Germany) in the magnetic field of a OctoMACS column stand Separator. Prepare column by rinsing with 500 μL of buffer:
        • vii. Apply cell suspension onto the column. Collect unlabeled cells that pass through and wash column with 500 μL of buffer. Collect total effluent; this is the unlabeled cell fraction. Perform washing steps by adding buffer three times. Only add new buffer when the column reservoir is empty.
        • viii. Remove column from the separator and place it on a suitable collection tube.
        • ix. Pipette 1 mL of buffer onto the column.
        • x. Immediately flush out the magnetically labeled cells by firmly pushing the plunger into the column.
        • xi. Centrifuge tubes 5 min at 500 g, remove supernatant. The final volume of app. 20 μl containing collected CD8+ cells and their associated Detection Molecules are store at −80 C for later analysis.
    • 8. Identification of enriched detection molecule: Separated cells and their associated Detection Molecules are analyzed by PCR amplification of the attached labels followed by sequencing of the PCR products to reveal the identity of isolated Detection Molecules and thus the identity of those antigens for which specific T cells were present in the sample. The identification is done as for e.g. example 6.

Example 123

This is an example where the samples are PBMC's prepared as three buffy coats (BC's), the Linker is PE and streptavidin conjugated dextran, and the Binding Molecules are biotinylated peptide-MHC (pMHC) complexes. The Labels are synthetic oligonucleotides modified with a terminal biotin capture-tag.

In this example, cells and their bound Detection Molecules are separated, by centrifugation, from the supernatant containing Detection Molecules not bound to cells. The DNA oligonucleotide Labels on Detection Molecules associated with captured cells are subjected to PCR amplification (as example 3) and the PCR product is analyzed as a read of the oligonucleotide label associated with the detection molecules bound to the isolated cells thus revealing the identity of detection molecules bound to the T cells present in the sample (as example 3).

    • 1. Sample preparation. The samples are PBMC's from three donor materials prepared as described in example 3.
    • 2. Linker preparation: The Linker is PE and streptavidin conjugated dextran (Dextramer from Immudex) which is prepared as in example 1.
    • 3. Binding molecules preparation: Binding Molecules are biotinylated pMHC complexes with peptide/HLA combination as described in example 3.
    • 4. Label preparation: Labels are biotin modified DNA oligonucleotides as described in example 3.
    • 5. Detection molecules preparation: Detection Molecules are prepared by combining 110 aliquots of Linker with individual Labels followed by adding pMHC Binding Molecules as described in Example 3.
    • 6. Incubation of sample and detection molecules: PBMC's are mixed with the 110 member library of Detection Molecules as described in example 3.
    • 7. Enrichment of detection molecules with desired characteristics
      • b. Apply: Detection Molecules are isolated by centrifugation-separation of cells and their bound Detection Molecules from the supernatant containing Detection Molecules not bound to cells. Briefly;
        • i. Centrifuge cell suspensions at 300×g for 10 minutes. Aspirate supernatant completely.
        • ii. Wash cells twice by adding 2 mL of buffer (phosphate-buffered saline (PBS), pH 7.2, 0.5% bovine serum albumin (BSA), and 2 mM EDTA) and centrifuge at 300×g for 10 minutes.
        • iii. Aspirate supernatant completely. The final volume of app. 20 μl containing collected cells and their associated Detection Molecules are store at −80C for later analysis.
    • 8. Identification of enriched Detection Molecule: Separated cells and their associated Detection Molecules are analyzed by PCR amplification of the attached labels followed by sequencing of the PCR products to reveal the identity of isolated Detection Molecules and thus the identity of those antigens for which specific T cells were present in the sample. The identification is done as for example 3.

Example 124

This is an example where the samples are PBMC's prepared as three buffy coats (BC's), the Linker is PE and streptavidin conjugated dextran, and the Binding Molecules are biotinylated peptide-MHC (pMHC) complexes. The Labels are synthetic oligonucleotides modified with a terminal biotin capture-tag.

In this example, INFγ producing cells and their bound Detection Molecules are separated from Detection Molecules not bound to INFγ producing cells by magnetic labeling and capture of INFγ producing cells using the MicroBead based INFγ Secretion Assay (INFγ Secretion Assay, #130-054-201, Miltenyi, Germany). Magnetic bead-isolated INFg producing cells are subjected to PCR amplification of associated DNA oligonucleotide Label (as described in example 3) and the PCR product is analyzed as a read of the oligonucleotide label associated with the detection molecules bound to the isolated cells thus revealing the identity of detection molecules bound to the INFγ producing cells present in the sample (as example 3).

    • 1. Sample preparation. The samples are PBMC's from three donor materials prepared as described in example 3.
    • 2. Linker preparation: The Linker is PE and streptavidin conjugated dextran (Dextramer from Immudex) which is prepared as in example 1.
    • 3. Binding molecules preparation: Binding Molecules are biotinylated pMHC complexes with peptide/HLA combination as described in example 3.
    • 4. Label preparation: Labels are biotin modified DNA oligonucleotides as described in example 3.
    • 5. Detection molecules preparation: Detection Molecules are prepared by combining 110 aliquots of Linker with individual Labels followed by adding pMHC Binding Molecules as described in Example 3.
    • 6. Incubation of sample and detection molecules: PBMC's are mixed with the 110 member library of Detection Molecules as described in example 3.
    • 7. Enrichment of detection molecules with desired characteristics
      • i. Apply: In this example Detection Molecules are isolated by capturing INF γ producing cells using anti INF γ catch antibody in combination with anti INF γ detection antibody. All reagents and procedures as described by Miltenyi (INF γ Secretion Assay, #130-054-201, Miltenyi, Germany). Finally INFγ producing cells are captured using anti PE Microbeads.
      • ii. Centrifuge tubes 5 min at 500 g, remove supernatant. The final volume of app. 20 μl containing collected INFγ producing cells and their associated Detection Molecules are store at −80 C for later analysis.
    • 8. Identification of enriched detection molecule: Separated cells and their associated Detection Molecules are analyzed by PCR amplification of the attached labels followed by sequencing of the PCR products to reveal the identity of isolated Detection Molecules and thus the identity of those antigens for which specific T cells were present in the sample. The identification is done as for example 3.

Items Set #1

  • 1. A method comprising the following steps:
    • a. Combining at least one cell with at least one detection molecule, where the detection molecule comprises a binding molecule (BM), a linker (Li), and a label (La);
    • b. Allowing the detection molecules to recognize and bind cells through their binding molecule entity;
    • c. Detecting or isolating pairs of cell-detection molecule complexes formed in step (b);
    • d. Identifying detection molecules capable of binding to a cell in step (b).
  • 2. The method of item 1 where the binding molecule (BM) is a peptide-MHC complex, an antibody or an oligonucleotide.
  • 3. The method of item 1 or 2 where the label (La) is an antibody, a nucleic acid, a particle comprising an electronic or electromagnetic signal, or a particle comprising a radio signal.
  • 4. The method of any of items 1-3 where the linker (Li) is a streptavidin, polysaccharide, dextran, peptide, or carbon-based polymer.
  • 5. The method of any of items 1-4 where step c is carried out by immobilization of the cell-detection molecule pairs
  • 6. The method of item 5 where said immobilization is by precipitating cells by centrifugation, immunoprecipitation of the cells optionally involving centrifugation, or any other means that precipitates the cells, leading to co-precipitation of detection molecules bound to cells.
  • 7. The method of item 6 where said immobilization is by binding one or more cells to a bead, particle or surface, or any other means that immobilizes said one or more cells, leading to co-immobilization of the detection molecules bound to said one or more cells.
  • 8. The method of any of items 5-7 where said immobilization involves the use of an antibody or other molecule, capable of specifically binding a subset of cells
  • 9. The method of item 8 where said antibody or other molecule specifically recognizes the T cell receptor (TCR) or the CD4 or CD8 proteins of T cells.
  • 10. The method of any of the preceding items where a label uniquely identifies individual detection molecules or identifies specific subsets of detection molecules
  • 11. The method of any of the preceding items where the Label is an oligonucleotide.
  • 12. The method of any of the preceding items where the binding molecule is a pMHC complex, an antibody or an oligonucleotide aptamer.
  • 13. The method of any of the preceding items where the linker is a dextran molecule, polysaccharide, oligonucleotide, streptavidin, peptide, carbon-based molecule, carbohydrate or an organic molecule.
  • 14. A multimeric major histocompatibility complex (MHC) comprising
    • a. two or more MHC's linked by a backbone molecule; and
    • b. at least one nucleic acid molecule linked to said backbone, wherein said nucleic acid molecule comprises a central stretch of nucleic acids (barcode region) designed to be amplified by e.g. PCR.
  • 15. The multimeric major histocompatibility complex according to item 14, wherein the backbone molecule is selected from the group consisting of polysaccharides, such as glucans such as dextran, a streptavidin or a streptavidin multimer.
  • 16. The multimeric major histocompatibility complex according to item 14 or 15, wherein the MHC's are coupled to the backbone through a streptavidin-biotin binding, streptavidin-avidin.
  • 17. The multimeric major histocompatibility complex according to any of the preceding items, wherein the MHC's are linked to the backbone via the MHC heavy chain.
  • 18. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the MHC is artificially assembled.
  • 19. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, composed of at least four MHC's, such as at least eight, such as at least ten, 2-30, 2-20, such as 2-10 or such as 4-10 MHC's.
  • 20. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the at least one nucleic acid molecule is composed of at least a 5′ first primer region, a central region (barcode region), and a 3′ second primer region.
  • 21. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the at least one nucleic acid molecule has a length in the range 20-100 nucleotides, such as 30-100, such as 30-80, such as 30-50 nucleotides.
  • 22. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the at least one nucleic acid molecule is linked to said backbone via a streptavidin-biotin binding and/or streptavidin-avidin binding.
  • 23. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the at least one nucleic acid molecule comprises or consists of DNA, RNA, and/or artificial nucleotides such as PLA or LNA.
  • 24. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the MHC is selected from the group consisting of class I MHC, a class II MHC, a CD1, or a MHC-like molecule.
  • 25. The multimeric major histocompatibility complex (MHC) according to any of the preceding items, wherein the backbone further comprises one or more linked fluorescent labels.
  • 26. A composition comprising a subset of multimeric major histocompatibility complexes (MHC's) according to any of items 14-25, wherein each set of MHC's has a different peptide decisive for T cell recognition and a unique “barcode” region in the DNA molecule.
  • 27. The composition according to item 26, wherein the primer regions in the DNA molecule are identical for each set of MHC's.
  • 28. A) The composition according to item 26 or 27, comprising at least 10 different sets of MHC's such as at least 100, such as at least 500, at least 1000, at least 5000, such as in the range 10-50000, such as 10-1000 or such as 50-500 sets of MHC's.
  • 28. B) A kit of parts comprising
    • a. a composition according to any of items 26 to 28; and
    • b. one or more sets of primers for amplifying the nucleic acid molecules.
  • 29. A method for detecting antigen responsive cells in a sample comprising:
  • i) providing one or more multimeric major histocompatibility complexes (MHC's) according to any of items 1-12 or a composition according to any of items 14-16; ii) contacting said multimeric MHC's with said sample; and detecting binding of the multimeric MHC's to said antigen responsive cells, thereby detecting cells responsive to an antigen present in a set of MHC's, wherein said binding is detected by amplifying the barcode region of said nucleic acid molecule linked to the one or more MHC's.
  • 30. The method according to item 29, wherein unbound MHC's are removed before amplification, e.g. by washing and/or spinning.
  • 31. The method according to item 29 or 30, wherein the sample is a blood sample, such as an peripheral blood sample, a blood derived sample, a tissue biopsy or another body fluid, such as spinal fluid, or saliva.
  • 32. The method according to any of items 29-31, wherein said sample has been obtained from a mammal, such as a human, mouse, pigs, and/or horses.
  • 33. The method according to any of item 30-32, wherein the method further comprises cell sorting by e.g. flow cytometry such as FACS.
  • 34. The method according to any of items 29-33, wherein said binding detection includes comparing measured values to a reference level, e.g. a negative control and/or total level of response.
  • 35. The method according to any of item 29-34, wherein said amplification is PCR such as QPCR.
  • 36. The method according to any of items 29-35, wherein the detection of barcode regions includes sequencing of said region such as deep sequencing or next generation sequencing.
  • 37. Use of a multimeric major histocompatibility complex (MHC) according to any of items 14-25 or a composition according to any of items 26-29 for the detecting of antigen responsive cells in a sample.
  • 38. Use of a multimeric major histocompatibility complex (MHC) according to any of items 14-25 or a composition according to any of items 26-29 in the diagnosis of diseases or conditions, preferably cancer and/or infectious diseases.
  • 39. Use of a multimeric major histocompatibility complex (MHC) according to any of items 14-25 or a composition according to any of items 26-29 in the development of immune-therapeutics.
  • 40. Use of a multimeric major histocompatibility complex (MHC) according to any of items 14-25 or a composition according to any of items 26-29 in the development of vaccines.
  • 41. Use of a multimeric major histocompatibility complex (MHC) according to any of items 14-25 or a composition according to any of items 26-29 for the identification of epitopes.
    References
  • 1. Altman J D, Moss P A, Goulder P J, Barouch D H, McHeyzer-Williams M G, Bell J I, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996; 274:94-6.
  • 2. Davis M M, Bjorkman P J. T-cell antigen receptor genes and T-cell recognition. Nature. 1988; 334:395-402.
  • 3. Robins H S, Campregher P V, Srivastava S K, Wacher A, Turtle C J, Kahsai O, et al. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood. 2009; 114:4099-107.
  • 4. Hadrup S R, Bakker A H, Shu C J, Andersen R S, van V J, Hombrink P, et al. Parallel detection of antigen-specific T-cell responses by multidimensional encoding of MHC multimers. Nature Methods. 2009; 6:520-6.
  • 5. Andersen R S, Kvistborg P, March T F, Pedersen N W, Lyngaa R, Bakker A H, et al. Parallel detection of antigen-specific T-cell responses by combinatorial encoding of MHC multimers. NatProtoc. 2012
  • 6. Newell E W, Sigal N, Nair N, Kidd B a, Greenberg H B, Davis M M. Combinatorial tetramer staining and mass cytometry analysis facilitate T-cell epitope mapping and characterization. Nat Biotechnol. 2013; 1-9.
  • 7. Soen Y, Chen D S, Kraft D L, Davis M M, Brown P O. Detection and characterization of cellular immune responses using peptide-MHC microarrays. PLoSBiol. 2003; 1:429-38.
  • 8. Stone J D, Demkowicz Jr. W E, Stern L J. HLA-restricted epitope identification and detection of functional T cell responses by using MHC-peptide and costimulatory microarrays. Proc Natl Acad Sci USA. 2005; 102:3744-9.
  • 9. Newell E W, Davis M M. Beyond model antigens: high-dimensional methods for the analysis of antigen-specific T cells. Nat Biotechnol. 2014; 32.
  • 10. Dössinger G, Bunse M, Bet J, Albrecht J, Paszkiewicz P J, Weiβbrich B, et al. MHC multimer-guided and cell culture-independent isolation of functional T cell receptors from single cells facilitates TCR identification for immunotherapy. PLoS One. 2013; 8:e61384.
  • 11. Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, et al. Improved Survival with T Cell Clonotype Stability After Anti-CTLA-4 Treatment in Cancer Patients. Sci Transl Med. 2014; 6:238ra70.
  • 12. Robert L, Tsoi J, Wang X, Emerson R O, Homet B, Chodon T, et al. CTLA4 blockade broadens the peripheral T cell receptor repertoire. Clin Cancer Res. 2014
  • 13. Morgan R A, Dudley M E, Wunderlich J R, Hughes M S, Yang J C, Sherry R M, et al. Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes. Science. 2006.
  • 14. Pannetier C, Even J, Kourilsky P. T-cell repertoire diversity and clonal expansions in normal and clinical samples. ImmunolToday. 1995; 16:176-81.
  • 15. Cameron B J, Gerry A B, Dukes J, Harper J V, Kannan V, Bianchi F C, et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013; 5:197ra103.
  • 16. Linette G P, Stadtmauer E a, Maus M V, Rapoport A P, Levine B L, Emery L, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013; 122:863-71.

Items Set #2

  • 1. A detection molecule comprising
    • a. at least one binding molecule (BM),
    • b. at least one linker (Li), and
    • c. at least one label (La).
  • 2. A cell-detection molecule complex comprising
    • a. At least one detection molecule comprising a binding molecule (BM), a linker (Li) and a label (La), and
    • b. at least one cell.
  • 3. A composition comprising two or more different detection molecules, or two or more sets of different detection molecules, each detection molecule comprising at least one binding molecule (BM), at least one linker (Li) and at least one label (La),
    • wherein each of the two or more detection molecules, or two or more sets of detection molecules, comprises a label which is unique to and specific for the binding molecule of each of said two or more different detection molecules.
  • 4. The composition according to item 3, said composition comprising 2 to 1,000,000 different detection molecules, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40 different detection molecules or sets of different detection molecules; for example 1-3, 3-5, 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-110, 110-120, 120-130, 130-140, 140-150, 150-175, 175-200, 200-250, 250-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 900-1000, 1000-1500, 1500-2000, 2000-3000, 3000-4000, 4000-5000, 5000-7500, 7500-10,000, 10,000-20,000, 20,000-50,000, 50,000-100,000, 100,000-200,000, 200,000-500,000, 500,000-1,000,000 different detection molecules, or sets of different detection molecules.
  • 5. The detection molecule according to any of the preceding items, wherein said binding molecule is capable of specifically associating with, recognizing and/or binding to a structure belonging to or associated with an entity in a sample, such as a target structure.
  • 6. The detection molecule according to any of the preceding items, wherein said binding molecule associates with, recognizes and/or binds to a marker molecule specific for a given cell or cell type.
  • 7. The detection molecule according to any of the preceding items, wherein said binding molecule is capable of specifically associating with, recognizing and/or binding to a specific cell type, such as selected from the group consisting of immune cells, lymphocytes, monocytes, dendritic cells, T-cells, B-cells, NK cells, CD4+ T cells, CD8+ T cells, αβ T cells, invariant γδ T cells, antigen-specific T-cells, cells comprising TCRs, cells comprising BCRs, a specific cancer cell
  • 8. The detection molecule according to any of the preceding items, wherein said binding molecule is capable of specifically associating with, recognizing and/or binding to a target specifically associated with an organ selected from the group consisting of lymph nodes, kidney, liver, skin, brain, heart, muscles, bone marrow, skin, skeleton, lungs, the respiratory tract, spleen, thymus, pancreas, exocrine glands, bladder, endocrine glands, reproduction organs including the phallopian tubes, eye, ear, vascular system, the gastroinstestinal tract including small intestines, colon, rectum, canalis analis and prostate gland.
  • 9. The detection molecule according to any of the preceding items, wherein said binding molecule is peptide-based or protein-based.
  • 10. The detection molecule according to any of the preceding items, wherein said binding molecule is an anti-target molecule capable of specifically associating with, recognizing and/or binding to a predetermined target structure.
  • 11. The detection molecule according to any of the preceding items, wherein said binding molecule is an anti-target-molecule.
  • 12. The detection molecule according to any of the preceding items, wherein said binding molecule is selected from the group consisting of an antibody, an antibody mimetic, an antibody-like molecule, a peptide, an oligonucleotide, a peptide aptamer, a nucleic acid aptamer, a DNA aptamer, an RNA aptamer, an XNA aptamer, a ligand, a natural ligand, a variant or fragment of a natural ligands, a synthetic ligand and a small organic molecule.
  • 13. The detection molecule according to any of the preceding items, wherein said antibody mimetic is selected from the group consisting of affibody molecules, affilinns, affimers, affitins, alphabodies, anticalins, avimers, DARPins, fynomers, Kunitz domain peptides and monobodies.
  • 14. The detection molecule according to any of the preceding items, wherein said binding molecule is an antibody selected from the group consisting of a monoclonal antibody, a polyclonal antibody, a bi-specific antibody, an antibody-like molecule, a Fc-molecule, a KIR-molecule, a ScFv and a Fab.
  • 15. The detection molecule according to any of the preceding items, wherein said binding molecule is a peptide of 1-100 amino acid residues without tertiary structure.
  • 16. The detection molecule according to any of the preceding items, wherein said binding molecule is a MHC molecule or MHC complex.
  • 17. The detection molecule according to any of the preceding items, wherein said binding molecule is a MHC class I complex.
  • 18. The detection molecule according to any of the preceding items, wherein said binding molecule is a MHC class II complex.
  • 19. The detection molecule according to any of the preceding items, wherein said binding molecule is a MHC-like molecule.
  • 20. The detection molecule according to any of the preceding items, wherein said binding molecule is CD1, wherein said CD1 is selected from the group consisting of CD1 CD1a, CD1b, CD1c, CD1d and CD1e.
  • 21. The detection molecule according to any of the preceding items, wherein said binding molecule is a MHC Class I-like proteins; including MIC A, MIC B, CD1d, HLA E, HLA F, HLA G, HLA H, ULBP-1, ULBP-2, and ULBP-3.
  • 22. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a cell-surface target.
  • 23. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is an intracellular target.
  • 24. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a receptor, such as a cell-surface receptor, an intracellular receptor, a soluble receptor or an extracellular receptor.
  • 25. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a T-cell receptor.
  • 26. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a B-cell receptor.
  • 27. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is CD4 of T cells.
  • 28. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is CD8 of T cells.
  • 29. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is CD20 of B cells.
  • 30. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is selected from the group consisting of cancer cell markers, developmental markers, cell cycle markers, proliferation markers, activation markers, hormones, hormone receptors, intracellular markers, cluster of differentiation (CD), cell surface markers, cytokines and cytokine receptors.
  • 31. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is selected from the group consisting of CD2, CD3, CD4, CD5, CD8, CD9, CD27, CD28, CD30, CD69, CD134 (OX40), CD137 (4-1BB), CD147, CDw150 (SLAM), CD152 (CTLA-4), CD153 (CD30L), CD40L (CD154), NKG2D, ICOS, HVEM, HLA Class II, PD-1, Fas (CD95), FasL, CD40, CD48, CD58, CD70, CD72, B7.1 (CD80), B7.2 (CD86), B7RP-1, B7-H3, PD-L1, PD-L2, CD134L, CD137L, ICOSL, LIGHT CD16, NKp30, NKp44, NKp46, NKp80, 2B4, KIR, LIR, CD94/NKG2A, CD94/NKG2C, LFA-1, CD11a/18, CD54 (ICAM-1), CD106 (VCAM), CD49a,b,c,d,e,f/CD29 (VLA-4), CD11a, CD14, CD15, CD19, CD25, CD30, CD37, CD49a, CD49e, CD56, CD27, CD28, CD45, CD45RA, CD45RO, CD45RB, CCR7, CCRS, CD62L, CD75, CD94, CD99, CD107b, CD109, CD152, CD153, CD154, CD160, CD161, CD178, CDw197, CDw217, Cd229, CD245, CD247 and Foxp3.
  • 32. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a cytokine selected from the group consisting of TNFα, TNFβ, TNF, IFNα, IFNβ, IFNγ, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-10-20, IL-20-30, IL-30, IL-31, IL-32, IL-33, IL-34, IL-35, IL-36, IL-37, IL-38, IL-39, IL-40, NFκB, chemokines including CC chemokines (CCL1 to CCL-28), CXC chemokines (CXCL1 to CXCL17) C chemokines (XCL-1 and -2) and CX3X chemokines (CX3CL1).
  • 33. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a cancer cell marker selected from the group consisting of HER2, CA125, Tyrosinase, Melanoma-associated antigen (MAGE), abnormal products of Ras or p53, Carcinoembryonic antigen, Muc-1, Epithelial tumor antigen, Carbonic Anhydrase, VEGFR, EGFR, TRAIL and RAN KL.
  • 34. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a stem cell marker selected from the group consisting of Stro-1, CD146, CD105, CD44, c-kit, Oct4, Sox-2, Klf4, EphB, Nestin and TWIST-1.
  • 35. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a developmental marker selected from the group consisting of Nanog, Oct4, Sox2, TEKT-1, NANOS, c-kit, Sox9, Notch, Msx1, Msx2 and Col1.
  • 36. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a proliferation marker selected from the group consisting of CyclinA, CyclinB, PCNA, PC10, p53, Mdm2, Cyclin D, Cyclin E, Rb, ARF and HDM2.
  • 37. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is an activation marker selected from the group consisting of CD28, Tbet, Eomes, Blimp, Bcl-6, CD27, MHC-II, TNF, IFN, Fizz1, ARG1 and CCL22R.
  • 38. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is an hormone selected from the group consisting of estrogen, PTH, ADH, T3, ANP, Epinephrine, Norepinephrine, Cortisol, Corticosterone, Aldosterone, Progestin, EPO, Leptin, Insulin, Glucagon, T4, ACTH, FSH, oxytocin and Calcitriol.
  • 39. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is an hormone receptor selected from the group consisting of EstrogenR (ER), GLP-1R, Thyroid receptor, Leptin receptor, Epinephrine receptor, Insulin receptor and Glucagon receptor.
  • 40. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is a cluster of differentiation (CD) molecule selected from the group consisting of CD1-10, CD10-20, CD20-30, CD30-40, CD40-50, CD50-100, CD100-200, CD200-300 and CD300-364.
  • 41. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is an intracellular marker selected from the group consisting of Cyclins, Cytokines and organelle markers (for example Apg12, Syntaxin, PAF-46, Histones, Early endosome antigen, clathrin, tubulins, PAF49, FTCD).
  • 42. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is selected from the group consisting of CD1, CD1a, CD1b, CD1c, CD1d, and MR1.
  • 43. The detection molecule according to any of the preceding items, wherein the target of the binding molecule is selected from the group consisting of targets included in Table 4 of the examples.
  • 44. The detection molecule according to any of the preceding items, wherein said binding molecule is a surface-molecule receptor to a cytokine receptor selected from the group consisting of interleukins and TNF-like molecules.
  • 45. The detection molecule according to any of the preceding items, wherein said label is any molecule, atom or signal the identity of which can be determined.
  • 46. The detection molecule according to any of the preceding items, wherein said label is unique to and/or specifies the binding molecule or a group of binding molecules.
  • 47. The detection molecule according to any of the preceding items, wherein said detection molecule comprises one label.
  • 48. The detection molecule according to any of the preceding items, wherein said detection molecule comprises more than one label.
  • 49. The detection molecule according to any of the preceding items, wherein said detection molecule comprises two or more labels, such as 3 labels, 4 labels, 5 labels, 6 labels, 7 labels, 8 labels, 9 labels, 10 labels, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 20-25, 25-30, 30-40, 40-50, 50-75, 75-100 labels.
  • 50. The detection molecule according to any of the preceding items, wherein said detection molecule comprises two or more labels and each of said labels are identical to each other.
  • 51. The detection molecule according to any of the preceding items, wherein said detection molecule comprises two or more labels and at least two of said labels are different.
  • 52. The detection molecule according to any of the preceding items, wherein said label is attached to the linker of the detection molecule.
  • 53. The detection molecule according to any of the preceding items, wherein said label is attached to the binding molecule of the detection molecule.
  • 54. The detection molecule according to any of the preceding items, wherein said label comprises a connector molecule (attachment molecule) for attachment to the detection molecule.
  • 55. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label.
  • 56. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label selected from the group consisting of a DNA label, an RNA label, and an artificial nucleic acid label.
  • 57. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising one or more nucleotides individually derived from one or more of DNA, RNA, and an artificial nucleic acid.
  • 58. The detection molecule according to any of the preceding items, wherein said artificial nucleic acid is selected from the group consisting of XNA, LNA, PNA, GNA, TNA, HNA, CeNA, and morpholino-nucleic acids.
  • 59. The detection molecule according to any of the preceding items, wherein said label is a DNA label.
  • 60. The detection molecule according to any of the preceding items, wherein said nucleic acid label comprises 1 to 1,000,000 nucleic acids, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40 nucleic acids; for example 1-3, 3-5, 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-110, 110-120, 120-130, 130-140, 140-150, 150-175, 175-200, 200-250, 250-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 900-1000, 1000-1500, 1500-2000, 2000-3000, 3000-4000, 4000-5000, 5000-7500, 7500-10,000, 10,000-100,000, 100,000-1,000,000 nucleic acids.
  • 61. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising one or more of
    • a. barcode region,
    • b. 5′ first primer region (forward)
    • c. 3′ second primer region (reverse),
    • d. random nucleotide region,
    • e. connector molecule
    • f. stability-increasing components
    • g. short nucleotide linkers in between any of the above-mentioned components
    • h. adaptors for sequencing
    • i. annealing region
  • 62. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising at least a barcode region.
  • 63. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising at least a barcode region, wherein said barcode region comprises a sequence of consecutive nucleic acids.
  • 64. The detection molecule according to any of the preceding items, wherein the barcode region of said nucleic acid comprises 1-3, 3-5, 5-10, 10-15, 15-20, 20-25, 25-30, 30-35, 35-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-110, 110-120, 120-130, 130-140, 140-150, 150-175, 175-200, 200-250, 250-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 900-1000 nucleic acids.
  • 65. The detection molecule according to any of the preceding items, wherein said barcode region comprises or consists of 5-10, 10-15, 15-20, 20-25, 25-30, 30-40, 40-45, 45-50 nucleic acids.
  • 66. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising at least a 3′ primer region, a barcode region, and a 5′ primer region.
  • 67. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising at least a 3′ primer region, a barcode region, and a 5′ primer region, wherein said barcode region is designed to be amplified by e.g. PCR and identified by e.g. sequencing.
  • 68. The detection molecule according to any of the preceding items, wherein the primer regions of said nucleic acid label are identical for subsets of detection molecules comprising different labels.
  • 69. The detection molecule according to any of the preceding items, wherein said label is a nucleic acid label comprising a connector molecule which is able to interact with a component on the linker and/or binding molecule of the detection molecule.
  • 70. The detection molecule according to any of the preceding items, wherein said nucleic acid label comprises a connector molecule which is biotin.
  • 71. The detection molecule according to any of the preceding items, wherein said nucleic acid label comprises a random nucleotide region comprising 3-20 nucleotides, such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 nt.
  • 72. The detection molecule according to any of the preceding items, wherein said nucleic acid label comprises one or more stability-increasing components, such as HEG or TEG.
  • 73. The detection molecule according to any of the preceding items, wherein a sample identifying sequence is attached to the nucleic acid label such as by attachment to one of the primers capable of binding to the primer regions of the nucleic acid label.
  • 74. The detection molecule according to any of the preceding items, wherein said label is a peptide label.
  • 75. The detection molecule according to any of the preceding items, wherein said peptide label comprises a stretch of consecutive amino acid residues (coding region).
  • 76. The detection molecule according to any of the preceding items, wherein said peptide label comprises a stretch of consecutive amino acid residues (coding region) and a protease cleavage site.
  • 77. The detection molecule according to any of the preceding items, wherein said peptide label comprises a stretch of consecutive amino acid residues (coding region) and a protease cleavage site.
  • 78. The detection molecule according to any of the preceding items, wherein said protease cleavage site in said peptide label is located proximal to the linker that connects the label to the binding molecule.
  • 79. The detection molecule according to any of the preceding items, wherein said peptide label comprising a protease cleavage site allows for cleavage of the stretch of consecutive amino acid residues (coding region) and release thereof from the detection molecule.
  • 80. The detection molecule according to any of the preceding items, wherein said label is a peptide label comprising 2 or more consecutive amino acids, such as 2-3, 3-4, 4-5, 5-6, 6-7, 7-8, 8-9, 9-10, 10-11, 11-12, 12-13, 13-14, 14-15, 15-16, 16-17, 17-18, 18-19, 19-20, 20-21, 21-22, 22-23, 23-24, 24-25, 25-26, 26-27, 27-28, 28-29, 29-30, 30-31, 31-32, 32-33, 33-34, 34-35, 35-36, 36-37, 37-38, 38-39, 39-40, 40-45, 45-50, 50-55, 55-60, 60-65, 65-70, 70-75, 75-80, 80-85, 85-90, 90-95, 95-100, 100-110, 110-120, 120-130, 130-140, 140-150, 150-160, 160-170,170-180, 180-190, 190-200, 200-225, 225-250, 250-275, 275-300, 300-350, 350-400, 400-450, 450-500, 500-600, 600-700, 700-800, 800-900, 900-1000, 1000-1500, 1500-2000, or more than 2000, consecutive amino acids.
  • 81. The detection molecule according to any of the preceding items, wherein said peptide label comprises proteinogenic and/or non-proteinogenic amino acids.
  • 82. The detection molecule according to any of the preceding items, wherein said label is a fluorescent label (fluorophore label).
  • 83. The detection molecule according to any of the preceding items, wherein said label is a fluorescent label selected from the group consisting of fluorescein isothiocyanate (FITC), fluorescein (Flu) derivates, rhodamine, tetramethylrhodamine, phycoerythrin, R-phycoerythrin (RPE), allophycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde, fluorescamine; 2-(4′-maleimidylanilino)naphthalene-6-sulfonic acid; 5-((((2-iodoacetyl)amino)ethyl)amino) naphthalene-1-sulfonic acid; Pyrene-1-butanoic acid; AlexaFluor 350 (7-amino-6-sulfonic acid-4-methyl coumarin-3-acetic acid); AMCA (7-amino-4-methyl coumarin-3-acetic acid); 7-hydroxy-4-methyl coumarin-3-acetic acid; Marina Blue (6,8-difluoro-7-hydroxy-4-methyl coumarin-3-acetic acid); 7-dimethylamino-coumarin-4-acetic acid; Fluorescamin-N-butyl amine adduct; 7-hydroxy-coumarine-3-carboxylic acid; CascadeBlue (pyrene-trisulphonic acid acetyl azide); Cascade Yellow; Pacific Blue (6,8 difluoro-7-hydroxy coumarin-3-carboxylic acid); 7-diethylamino-coumarin-3-carboxylic acid; N-(((4-azidobenzoyl)amino)ethyl)-4-amino-3,6-disulfo-1,8-naphthalimide; Alexa Fluor 430; 3-perylenedodecanoic acid; 8-hydroxypyrene-1,3,6-trisulfonic acid; 12-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)dodecanoic acid; N,N′-dimethyl-N-(iodoacetyl)-N′-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine; Oregon Green 488 (difluoro carboxy fluorescein); 5-iodoacetamidofluorescein; propidium iodide-DNA adduct; Carboxy fluorescein; 5- or 6-carboxyfluorescein; 6-(fluorescein)-5-(and 6)-carboxamido hexanoic acid; Texas Red, Princeton Red, Green fluorescent protein (GFP) and analogues thereof; PerCP; AlexaFluor® (AF), AF405, AF488,AF500, AF514, AF532, AF546, AF555, AF568, AF594, AF610, AF633, AF635, AF647, AF680, AF700, AF710, AF750, AF800; Quantum Dot based dyes, Qdot®525, Qdot®565, Qdot®585, Qdot®605, Qdot®655, Qdot®705, Qdot®800; DyLight™ Dyes (Pierce) (DL); DL549, DL649, DL680, DL800; Cy-Dyes, Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7; Fluorescent Proteins, RPE, PerCp, APC, Green fluorescent proteins; GFP and GFP derivated mutant proteins; BFP, CFP, YFP, DsRed, T1, Dimer2, mRFP1,MBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry; Tandem dyes, RPE-Cy5, RPE-Cy5.5, RPE-Cy7, RPE-AlexaFluor® tandem conjugates; RPE-Alexa610, RPE-TxRed, APC-Aleca600, APC-Alexa610, APC-Alexa750, APC-Cy5 and APC-Cy5.5.
  • 84. The detection molecule according to any of the preceding items, wherein said label is a phosphorescence label.
  • 85. The detection molecule according to any of the preceding items, wherein said label is a bioluminescence label or chemoluminescence label.
  • 86. The detection molecule according to any of the preceding items, wherein said chemoluminescence label is selected from luminol, isoluminol, acridinium esters, acridinium salt, theromatic acridinium ester, 1,2-dioxetanes, oxalate ester, imidazole and pyridopyridazines.
  • 87. The detection molecule according to any of the preceding items, wherein said bioluminescence label is selected from the group luciferin, luciferase and aequorin.
  • 88. The detection molecule according to any of the preceding items, wherein said label is an enzymatic label.
  • 89. The detection molecule according to any of the preceding items, wherein said label is an enzymatic label, wherein the enzyme catalyze a reaction between chemicals in the near environment of the labeling molecules, which results in one or more of producing a light signal (chemi-luminescence) and precipitation of chromophor dyes.
  • 90. The detection molecule according to any of the preceding items, wherein said label is an enzymatic label selected from peroxidases, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • 91. The detection molecule according to any of the preceding items, wherein said label is an enzymatic label selected from horse radish peroxidase (HRP), alkaline phosphatase (AP), beta-galactosidase (GAL), glucose-6-phosphate dehydrogenase, beta-N-acetylglucosaminidase, β-glucuronidase, invertase, Xanthine Oxidase, firefly luciferase and glucose oxidase (GO).
  • 92. The detection molecule according to any of the preceding items, wherein said label is capable of reflection of light, such as gold, plastic, glass, polystyrene and pollen.
  • 93. The detection molecule according to any of the preceding items, wherein said label is capable of capable of absorption of light, such as a chromophore or a dye.
  • 94. The detection molecule according to any of the preceding items, wherein said label is capable of emission of light after excitation, such as a fluorochrome.
  • 95. The detection molecule according to any of the preceding items, wherein said label is a nanoparticle label.
  • 96. The detection molecule according to any of the preceding items, wherein said label is an element.
  • 97. The detection molecule according to any of the preceding items, wherein said label is selected from the group consisting of heavy metal labels, isotope labels, radiolabels, radionuclide, stable isotopes, chains of isotopes and single atoms.
  • 98. The detection molecule according to any of the preceding items, wherein said label is a single atom selected from the group consisting of zinc (Zn), iron (Fe), magnesium (Mg), any of the lanthanides (Ln) including La, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm, Yb and Lu; scandium (Sc) and yttrium (Y).
  • 99. The detection molecule according to any of the preceding items, wherein said radioactivity labels comprises incorporated isotopes of iodide, cobalt, selenium, tritium and/or phosphor.
  • 100. The detection molecule according to any of the preceding items, wherein said label is a DNA fluorescing stain, such as Propidium iodide, Hoechst stain, DAPI, DraQ5 or Acridine orange.
  • 101. The detection molecule according to any of the preceding items, wherein said label comprises a nucleic acid label and at least a second label.
  • 102. The detection molecule according to any of the preceding items, wherein said label comprises a nucleic acid label and at least a second label according to any of the preceding items.
  • 103. The detection molecule according to any of the preceding items, wherein said label comprises a nucleic acid label and a fluorophore label.
  • 104. The detection molecule according to any of the preceding items, wherein said label comprises one type of label.
  • 105. The detection molecule according to any of the preceding items, wherein said label comprises more than one type of label, such as comprising 2 types of labels, for example comprising 3 types of labels, such as comprising 4 types of labels, for example comprising 5 types of labels.
  • 106. The detection molecule according to any of the preceding items, wherein said linker is a molecular entity and/or bond that connects the binding molecule and the label.
  • 107. The detection molecule according to any of the preceding items, wherein one or more of the binding molecules are covalently associated with the one or more linkers.
  • 108. The detection molecule according to any of the preceding items, wherein one or more of the binding molecules are non-covalently associated with the one or more linkers.
  • 109. The detection molecule according to any of the preceding items, wherein one or more labels are covalently associated with the one or more linkers.
  • 110. The detection molecule according to any of the preceding items, wherein one or more of labels are non-covalently associated with the one or more linkers.
  • 111. The detection molecule according to any of the preceding items, wherein the one or more labels and/or one or more binding molecules are associated with a molecule in the one or more linkers, such as a connector molecule, a sugar residue, a protein, an antibody, a DNA, an aptamer, reactive groups, nucleophilic group, electrophilic groups, radicals, or conjugated double bonds.
  • 112. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more multimerization domains.
  • 113. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more scaffolds.
  • 114. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more connectors.
  • 115. The detection molecule according to any of the preceding items, wherein the one or more multimerization domains comprises at least one scaffold and at least one connector.
  • 116. The detection molecule according to any of the preceding items, wherein the binding molecule and/or the label is attached to the linker via a streptavidin-biotin linkage.
  • 117. The detection molecule according to any of the preceding items, wherein the one or more linkers comprise one or more optionally substituted organic molecules.
  • 118. The detection molecule according to any of the preceding items, wherein the optionally substituted organic molecule comprises one or more functionalized cyclic structures.
  • 119. The detection molecule according to any of the preceding items, wherein the one or more functionalized cyclic structures comprises one or more benzene rings.
  • 120. The detection molecule according to any of the preceding items, wherein the optionally substituted organic molecule comprises a scaffold molecule comprising at least three reactive groups, or at least three sites suitable for non-covalent attachment.
  • 121. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more biological cells and/or cell-like structures, such as antigen presenting cells or dendritic cells.
  • 122. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more membranes.
  • 123. The detection molecule according to any of the preceding items, wherein the one or more membranes comprises liposomes or micelles.
  • 124. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more polymers such as one or more synthetic polymers.
  • 125. The detection molecule according to any of the preceding items, wherein reactive groups involved in forming an association between the multimerization domain and the binding molecule are located on glutamate or aspartate residues, or on a vinyl sulfone activated dextran.
  • 126. The detection molecule according to any of the preceding items, wherein the one or more linker polymers are selected from the group consisting of polysaccharides.
  • 127. The detection molecule according to any of the preceding items, wherein the linker comprises one or more dextran moieties.
  • 128. The detection molecule according to any of the preceding items, wherein the one or more dextran moieties are covalently attached to one or more binding molecules.
  • 129. The detection molecule according to any of the preceding items, wherein the one or more dextran moieties are non-covalently attached to one or more binding molecules.
  • 130. The detection molecule according to any of the preceding items, wherein the one or more dextran moieties are covalently attached to one or more labels.
  • 131. The detection molecule according to any of the preceding items, wherein the one or more dextran moieties are non-covalently attached to one or more labels.
  • 132. The detection molecule according to any of the preceding items, wherein reactive groups of the multimerization domains include hydroxyls of polysaccharides such as dextrans
  • 133. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties are modified.
  • 134. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties are activated.
  • 135. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties are activated by reaction of the dextran hydroxyls with divinyl sulfon.
  • 136. The detection molecule according to any of the preceding items, wherein dextran is activated by a multistep reaction that results in the decoration of the dextran with maleimide groups.
  • 137. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties comprises one or more amino-dextrans.
  • 138. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties comprises one or more amino-dextrans modified with divinyl sulfone.
  • 139. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties comprises one or more dextrans with a molecular weight of from 1,000 to 50,000, such as from 1,000 to 5,000, for example 5,000 to 10,000, such as from 10,000 to 15,000, for example 15,000 to 20,000, such as from 20,000 to 25,000, for example 25,000 to 30,000, such as from 30,000 to 35,000, for example 35,000 to 40,000, such as from 40,000 to 45,000, for example 45,000 to 50,000.
  • 140. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties comprises one or more dextrans with a molecular weight of from 50,000 to 150,000, such as from 50,000 to 60,000, for example 60,000 to 70,000, such as from 70,000 to 80,000, for example 80,000 to 90,000, such as from 90,000 to 100,000, for example 100,000 to 110,000, such as from 110,000 to 120,000, for example 120,000 to 130,000, such as from 130,000 to 140,000, for example 140,000 to 150,000.
  • 141. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties comprises one or more dextrans with a molecular weight of from 150,000-270,000 such as from 150,000 to 160,000, for example 160,000 to 170,000, such as from 170,000 to 180,000, for example 180,000 to 190,000, such as from 190,000 to 200,000, for example 200,000 to 210,000, such as from 210,000 to 220,000, for example 220,000 to 230,000, such as from 230,000 to 240,000, for example 240,000 to 250,000, such as from 250,000 to 260,000, for example 260,000 to 270,000, such as from 270,000 to 280,000, for example 280,000 to 290,000, such as from 290,000 to 300,000, for example 300,000 to 310,000 such as from 310,000 to 320,000, for example 320,000 to 330,000 such as from 330,000 to 340,000, for example 340,000 to 350,000 such as from 350,000 to 360,000, for example 360,000 to 370,000 such as from 370,000 to 380,000, for example 380,000 to 390,000, such as from 390,000 to 400,000.
  • 142. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties are linear.
  • 143. The detection molecule according to any of the preceding items, wherein the one or more linker dextran moieties are branched.
  • 144. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a carboxy methyl dextran and/or a dextran polyaldehyde and/or a carboxymethyl dextran lactone and/or a cyclodextrin.
  • 145. The detection molecule according to any of the preceding items, wherein the one or more linker synthetic polymers are selected from the group consisting of PNA, polyimide and PEG.
  • 146. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises one or more entities selected from the group consisting of an IgG domain, a coiled-coil polypeptide structure, a DNA duplex, a nucleic acid duplex, PNA-PNA, PNA-DNA, DNA-RNA.
  • 147. The detection molecule according to any of the preceding items, wherein the one or more linkers, such as one or more multimerization domains, comprises one or more avidins, such as one or more streptavidins.
  • 148. The detection molecule according to any of the preceding items, wherein the one or more streptavidins comprises one or more tetrameric streptavidin variants.
  • 149. The detection molecule according to any of the preceding items, wherein the one or more streptavidins comprises one or more monomeric streptavidin variants.
  • 150. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises an antibody.
  • 151. The detection molecule according to any of the preceding items, wherein the linker antibody is selected from the group consisting of polyclonal antibody, monoclonal antibody, IgA, IgG, IgM, IgD, IgE, IgG1, IgG2, IgG3, IgG4, IgA1, IgA2, IgM1, IgM2, humanized antibody, humanized monoclonal antibody, chimeric antibody, mouse antibody, rat antibody, rabbit antibody, human antibody, camel antibody, sheep antibody, engineered human antibody, epitope-focused antibody, agonist antibody, antagonist antibody, neutralizing antibody, naturally-occurring antibody, isolated antibody, monovalent antibody, bispecific antibody, trispecific antibody, multispecific antibody, heteroconjugate antibody, immunoconjugates, immunoliposomes, labeled antibody, antibody fragment, domain antibody, nanobody, minibody, maxibody, diabody, fusion antibody.
  • 152. The detection molecule according to any of the preceding items, wherein the detection molecule comprises one or more organic molecules selected from the group consisting of small organic scaffold molecules, small organic molecules, steroids, peptides, aromatic organic molecules, monocyclic structures, functionalized or substituted benzene rings, dicyclic structures, polycyclic structures, aliphatic molecules, monocyclic molecules, dicyclic molecules, polycyclic molecules.
  • 153. The detection molecule according to any of the preceding items, wherein the detection molecule comprises one or more monomeric molecules able to polymerize; one or more biological polymers such as one or more proteins; one or more small molecule scaffolds; one or more supramolecular structure(s) such as one or more nanoclusters; and/or one or more protein complexes.
  • 154. The detection molecule according to any of the preceding items, wherein the linker of the detection molecule comprises one or more beads.
  • 155. The detection molecule according to any of the preceding items, wherein the linker is a bead coated with streptavidin, such as streptavidin monomers or tetramers, and the one or more binding molecules are biotinylated.
  • 156. The detection molecule according to any of the preceding items, wherein the linker is a bead coated with polysaccharide, such as a polysaccharide comprising dextran moieties.
  • 157. The detection molecule according to any of the preceding items, wherein the one or more beads are selected from the groups consisting of beads that carry electrophilic groups e.g. divinyl sulfone activated polysaccharide, polystyrene beads that have been functionalized with tosyl-activated esters, magnetic polystyrene beads functionalized with tosyl-activated esters, and beads where binding molecules have been covalently immobilized to these by reaction of nucleophiles comprised within the binding molecules with the electrophiles of the beads.
  • 158. The detection molecule according to any of the preceding items, wherein the one or more beads is selected from the groups consisting of sepharose beads, sephacryl beads, polystyrene beads, agarose beads, polysaccharide beads, polycarbamate beads and any other kind of beads that can be suspended in an aqueous buffer.
  • 159. The detection molecule according to any of the preceding items, wherein the linker comprises one or more compounds selected from the group consisting of agarose, sepharose, resin beads, glass beads, pore-glass beads, glass particles coated with a hydrophobic polymer, chitosan-coated beads, SH beads, latex beads, spherical latex beads, allele-type beads, SPA bead, PEG-based resins, PEG-coated bead, PEG-encapsulated bead, polystyrene beads, magnetic polystyrene beads, glutathione agarose beads, magnetic bead, paramagnetic beads, protein A and/or protein G sepharose beads, activated carboxylic acid bead, macroscopic beads, microscopic beads, insoluble resin beads, silica-based resins, cellulosic resins, cross-linked agarose beads, polystyrene beads, cross-linked polyacrylamide resins, beads with iron cores, metal beads, dynabeads, Polymethylmethacrylate beads activated with NHS, streptavidin-agarose beads, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, nitrocellulose, polyacrylamides, gabbros, magnetite, polymers, oligomers, non-repeating moieties, polyethylene glycol (PEG), monomethoxy-PEG, mono-(C1-C10)alkoxy-PEG, aryloxy-PEG, poly-(N-vinyl pyrrolidone)PEG, tresyl monomethoxy PEG, PEG propionaldehyde, bis-succinimidyl carbonate PEG, polystyrene bead crosslinked with divinylbenzene, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, dextran, aminodextran, carbohydrate-based polymers, cross-linked dextran beads, polysaccharide beads, polycarbamate beads, divinyl sulfone activated polysaccharide, polystyrene beads that have been functionalized with tosyl-activated esters, magnetic polystyrene beads functionalized with tosyl-activated esters, streptavidin beads, streptavidin-monomer coated beads, streptavdin-tetramer coated beads, Streptavidin Coated Compel Magnetic beads, avidin coated beads, dextramer coated beads, divinyl sulfone-activated dextran, Carboxylate-modified bead, amine-modified beads, antibody coated beads, cellulose beads, grafted co-poly beads, poly-acrylamide beads, dimethylacrylamide beads optionally crosslinked with N—N′-bis-acryloylethylenediamine, hollow fiber membranes, fluorescent beads, collagen-agarose beads, gelatin beads, collagen-gelatin beads, collagen-fibronectin-gelatin beads, collagen beads, chitosan beads, collagen-chitosan beads, protein-based beads, hydrogel beads, hemicellulose, alkyl cellulose, hydroxyalkyl cellulose, carboxymethylcellulose, sulfoethylcellulose, starch, xylan, amylopectine, chondroitin, hyarulonate, heparin, guar, xanthan, mannan, galactomannan, chitin and chitosan.
  • 160. The detection molecule according to any of the preceding items, wherein the multimerization domain comprises one or more beads further comprises a linker moiety.
  • 161. The detection molecule according to any of the preceding items, wherein the multimerization domain comprises one or more beads comprising a linker moiety which is a flexible linker, a rigid linker, a water-soluble linker or a cleavable linker.
  • 162. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a dimerization domain.
  • 163. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a trimerization domain.
  • 164. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a tetramerization domain.
  • 165. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a pentamerization domain.
  • 166. The detection molecule according to any of the preceding items, wherein the pentamerization domain comprises a coiled-coil polypeptide structure.
  • 167. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a hexamerization domain, such as a hexamerization domain comprises three IgG domains.
  • 168. The detection molecule according to any of the preceding items, wherein the one or more linkers comprises a polyamide and/or a polyethylene glycol and/or a polysaccharide and/or a sepharose.
  • 169. The detection molecule according to any of the preceding items, wherein the one or more linkers have a molecular weight of less than 1,000 Da.
  • 170. The detection molecule according to any of the preceding items, wherein the one or more linkers have a molecular weight of from 1,000 Da to preferably less than 10,000 Da.
  • 171. The detection molecule according to any of the preceding items, wherein the one or more linkers have a molecular weight of from 10,000 Da to preferably less than 100,000 Da.
  • 172. The detection molecule according to any of the preceding items, wherein the one or more linkers have a molecular weight of from 100,000 Da to preferably less than 1,000,000 Da.
  • 173. The detection molecule according to any of the preceding items, wherein the one or more linkers have a molecular weight of more than 1,000,000 Da.
  • 174. The detection molecule according to any of the preceding items further comprising one or more scaffolds, carriers, connectors and/or linkers selected from the group consisting of streptavidin (SA) and avidin and derivatives thereof, biotin, immunoglobulins, antibodies (monoclonal, polyclonal, and recombinant), antibody fragments and derivatives thereof, leucine zipper domain of AP-1 (jun and fos), hexa-his (metal chelate moiety), hexa-hat GST (glutathione S-tranferase) glutathione affinity, Calmodulin-binding peptide (CBP), Strep-tag, Cellulose Binding Domain, Maltose Binding Protein, S-Peptide Tag, Chitin Binding Tag, Immuno-reactive Epitopes, Epitope Tags, E2Tag, HA Epitope Tag, Myc Epitope, FLAG Epitope, AU1 and AU5 Epitopes, Glu-Glu Epitope, KT3 Epitope, IRS Epitope, Btag Epitope, Protein Kinase-C Epitope, VSV Epitope, lectins that mediate binding to a diversity of compounds, including carbohydrates, lipids and proteins, e.g. Con A (Canavalia ensiformis) or WGA (wheat germ agglutinin) and tetranectin or Protein A or G (antibody affinity).
  • 175. The detection molecule according to any of the preceding items, wherein the binding molecule and/or the label comprises a connector molecule, such as biotin, and the linker comprises a connector such as streptavidin or another avidin connector.
  • 176. The detection molecule according to any of the preceding items, wherein the binding molecule is linked to at least one of the one or more multimerization domains by a non-covalent linker moiety, such as natural dimerization or protein-protein interactions.
  • 177. The detection molecule according to any of the preceding items, wherein the binding molecule is linked to at least one of the one or more multimerization domains by a protein-protein interaction selected from the group consisting of Fos/Jun interactions, Acid/Base coiled coil structure based interactions, antibody/antigen interactions, polynucleotide-polynucleotide interactions, synthetic molecule-synthetic molecule interactions and protein-small molecule interactions.
  • 178. The detection molecule according to any of the preceding items, wherein the binding molecule is linked to at least one of the one or more multimerization domains by natural dimerization selected from the group consisting of antigen-antibody pairs, DNA-DNA interactions, natural interactions, biotin and streptavidin.
  • 179. The detection molecule according to any of the preceding items, wherein the detection molecule further comprises an enzyme capable of catalysing the transfer of a cell surface moiety (e.g. a peptide fragment or ‘peptide tag’) from a cell surface protein to the binding molecule of the detection molecule, when said surface moiety binds to the binding molecule.
  • 180. The detection molecule according to any of the preceding items, comprising
    • a. a monomeric or a multimeric major histocompatibility complex (MHC) molecule, such as a monomeric or multimeric peptide MHC complex,
    • b. a linker comprising a multimerization domain and optionally one or more connectors, and
    • c. a nucleic acid label.
  • 181. The detection molecule according to any of the preceding items, comprising
    • a. a monomeric or a multimeric major histocompatibility complex (MHC) molecule, such as a monomeric or multimeric peptide MHC complex,
    • b. a linker comprising a multimerization domain and optionally one or more connectors, and
    • c. a peptide label.
  • 182. The detection molecule according to any of the preceding items, comprising
    • a. CD1, wherein said CD1 is selected from the group consisting of CD1 CD1a, CD1b, CD1c, CD1d and CD1e,
    • b. a linker comprising a multimerization domain and optionally one or more connectors, and
    • c. a nucleic acid label.
  • 183. The detection molecule according to any of the preceding items, comprising
    • a. an anti-target molecule capable of associating with, recognizing and/or binding to a predetermined marker molecule (or target) on a cell type, wherein said marker molecule is specific for a certain cell type
    • b. a linker (Li) comprising a multimerization domain and optionally one or more connectors, and
    • c. a nucleic acid label.
  • 184. The detection molecule according to any of the preceding items, wherein said linker comprising a multimerization domain and optionally one or more connectors is a dextran optionally comprising streptavidin or avidin, and said binding molecule and/or label optionally comprises biotin.
  • 185. A kit of parts comprising
    • a. One or more detection molecules according to any of the preceding items, and
    • b. one or more additional components.
  • 186. The kit of parts according to any of the preceding items, wherein said one or more additional components comprise reagents for detecting and/or amplifying the label of the detection molecule.
  • 187. The kit of parts according to any of the preceding items, wherein said one or more additional components comprise reagents for detecting and/or amplifying the nucleic acid label of the detection molecule.
  • 188. The kit of parts according to any of the preceding items, wherein said reagents for detecting the nucleic acid label of the detection molecule comprises one or more primer sets capable of amplifying the nucleic acid label.
  • 189. A detection method comprising the steps of
    • a. Combining a sample with at least one detection molecule; wherein the detection molecule comprises a binding molecule (BM), a linker (Li), and a label (La) according to any of the preceding items; and wherein said sample comprises at least one cell and/or entity,
    • b. Incubating the at least one detection molecule and the sample;
    • c. Isolating and/or detecting the at least one detection molecule of step b), and
    • d. Optionally determining the identity of the at least one detection molecule of step c).
  • 190. The detection method according to any of the preceding items, wherein in step b) the one or more detection molecules are allowed to associate with, recognize, and/or bind to said at least one cell and/or entity through their binding molecule.
  • 191. The detection method according to any of the preceding items, wherein in step c) and d) said detection molecule is comprised in a cell-detection molecule complex or an entity-detection molecule complex.
  • 192. The detection method according to any of the preceding items, wherein in step c) and d) said detection molecule is not comprised in a cell-detection molecule complex or an entity-detection molecule complex.
  • 193. The detection method according to any of the preceding items, wherein in step c) and d) said detection molecule is no longer comprised in a cell-detection molecule complex or an entity-detection molecule complex, wherein said detection molecule has previously interacted with a cell-detection molecule complex or an entity-detection molecule complex.
  • 194. The detection method according to any of the preceding items, wherein said cell-detection molecule complexes comprises a cell, such as an immune cell, associated with or bound to a detection molecule having a binding molecule specific for the immune cell.
  • 195. The detection method according to any of the preceding items, wherein step c) comprises isolating and detecting the at least one detection molecule.
  • 196. The detection method according to any of the preceding items, wherein step c) comprises first isolating and then detecting the at least one detection molecule.
  • 197. The detection method according to any of the preceding items, wherein step c) comprises detecting the at least one detection molecule.
  • 198. The detection method according to any of the preceding items, wherein step c) comprises isolating the at least one detection molecule.
  • 199. The detection method according to any of the preceding items, wherein step c) comprises first detecting and then isolating the at least one detection molecule.
  • 200. The detection method according to any of the preceding items, wherein in step c) isolating comprises flow cytometry and/or FACS sorting
  • 201. The detection method according to any of the preceding items, wherein in step c) isolating comprises one or more steps of washing, centrifugation and/or precipitation.
  • 202. The detection method according to any of the preceding items, wherein in step c) isolating comprises one or more steps of filtration.
  • 203. The detection method according to any of the preceding items, wherein in step c) isolating comprises one or more steps of application on an affinity column.
  • 204. The detection method according to any of the preceding items, wherein in step c) isolating comprises sorting of cell populations based on the functional response to a stimuli (responsive or non-responsive population), such as cytokine secretion, phosphorylation, calcium release.
  • 205. The detection method according to any of the preceding items, wherein in step c) isolating comprises sorting of cell populations based on phenotype, such as by linking a certain set of phenotypic characteristics to the antigen-responsiveness.
  • 206. The detection method according to any of the preceding items, wherein in step c) isolating comprises immobilization of the detection molecule and/or cell-detection molecule complexes.
  • 207. The detection method according to any of the preceding items, wherein said immobilization of the cell-detection molecule complexes comprises precipitating cells, such as by centrifugation, by immunoprecipitation, or any other means that precipitates the cells.
  • 208. The detection method according to any of the preceding items, wherein said immobilization of the cell-detection molecule complexes comprises binding the cell-detection molecule complexes to a bead, a particle, another surface, an antibody or an MHC complex.
  • 209. The detection method according to any of the preceding items, wherein said immobilization of the detection molecule and/or cell-detection molecule complexes comprises hybridization onto an array.
  • 210. The detection method according to any of the preceding items, wherein said immobilization of the detection molecule and/or cell-detection molecule complexes by hybridization onto an array comprises a nucleic acid/nucleic acid-interaction between the nucleic acid label of the detection molecule and an antisense nucleic acid sequence in the array.
  • 211. The detection method according to any of the preceding items, wherein said immobilization of the detection molecule and/or cell-detection molecule complexes by hybridization onto an array comprises a DNA/DNA-interaction between the DNA label of the detection molecule and an antisense DNA in the array.
  • 212. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises one or more steps of adding primary antibodies that bind to the immobilized detection molecule and/or cell-detection molecule complexes and detecting said primary antibodies directly wherein the primary antibody is labelled, or indirectly by adding labelled secondary antibodies.
  • 213. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises one or more steps of detecting the immobilized detection molecule and/or cell-detection molecule complexes by monitoring read-out from a second label such as a fluorophore.
  • 214. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises one or more steps of determining the identity of said label.
  • 215. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises interaction between ‘coating DNA’ on the cell surface and the DNA label of the detection molecule.
  • 216. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises protease cleavage of the peptide label of the detection molecule.
  • 217. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises transfer of a cell surface moiety to the detection molecule (e.g. a ‘peptide tag’).
  • 218. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises detection of the label based on the physical characteristics of the label, including mass, sequence, charge, volume, size, dimensions, fluorescence, absorption, emission, NMR spectra and others.
  • 219. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises amplification of the label.
  • 220. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises sequencing of the label (e.g. DNA sequencing, peptide sequencing).
  • 221. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises amplification of the barcode sequence of a nucleic acid label by PCR and/or sequencing of the barcode sequence.
  • 222. The detection method according to any of the preceding items, wherein said sequencing comprises deep sequencing or next generation sequencing.
  • 223. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises mass spectrometry.
  • 224. The detection method according to any of the preceding items, wherein said detecting in step c) and/or determining the identity in step d) comprises one or more of gel electrophoresis, gel filtration, PAGE, column fractionation, PCR and QPCR.
  • 225. The detection method according to any of the preceding items, said method further comprising one or more steps of providing a sample, preferably a sample comprising at least one entity and/or at least one cell.
  • 226. The detection method according to any of the preceding items, said method further comprising one or more steps of pre-treatment of the sample, and/or pre-treatment of cells of the sample.
  • 227. The detection method according to any of the preceding items, said method further comprising one or more steps of separating unbound detection molecules from cell- or entity-detection molecule complexes.
  • 228. The detection method according to any of the preceding items, said method further comprising one or more steps of removing unbound detection molecules by washing and/or centrifuging.
  • 229. The detection method according to any of the preceding items, said method further comprising one or more steps of single-cell sorting and sequencing.
  • 230. The detection method according to any of the preceding items, said method further comprising one or more steps of single-cell T cell sorting of and single-cell TCR sequencing.
  • 231. The detection method according to any of the preceding items, wherein said sample comprises one or more cells.
  • 232. The detection method according to any of the preceding items, wherein said sample comprises at least one cell and/or entity to which the binding molecule of the detection molecule is able to associate with, recognize and/or bind.
  • 233. The detection method according to any of the preceding items, wherein said sample is selected from the group consisting of a solid sample, a fluid sample, a semifluid sample, a liquid sample, a solubilised sample and a sample comprising dissociated cells of a solid sample.
  • 234. The detection method according to any of the preceding items, wherein said sample is selected from the group consisting of a biofilm, a biopsy, a surgical sample, a tissue sample, a microarray-fixed sample, a section such as a fresh section, a frozen section and a FFPE section.
  • 235. The detection method according to any of the previous items, wherein said sample is selected from the group consisting of blood, whole blood, plasma, serum, Peripheral blood mononuclear cells (PBMC), human PBMN (HPBMC), buffy coat, synovial fluid, bone marrow, cerebrospinal fluid, saliva, lymph fluid, seminal fluid, urine, stool, exudate, transdermal exudates, pharyngeal exudates, nasal secretions, sputum, sweat, bronchoalveolar lavage, tracheal aspirations, fluid from joints, vitreous fluid, vaginal or urethral secretions or semen.
  • 236. The detection method according to any of the previous items, wherein said sample comprises cell populations isolated from a fluid, a semifluid sample or a solid sample.
  • 237. The detection method according to any of the previous items, wherein said cell is selected from the group consisting of immune cells, lymphocytes, monocytes, dendritic cells, T-cells, B-cells and NK cells
  • 238. The detection method according to any of the previous items, wherein said cell is a T-cell, such as a T cell selected from the group consisting of CD4+ T cells, CD8+ T cells, αβ T cells and invariant γδ T cells.
  • 239. The detection method according to any of the previous items, wherein said cell is an antigen-specific T-cell or antigen-responsive T cell.
  • 240. The detection method according to any of the previous items, wherein said cell comprises T-cell receptors.
  • 241. The detection method according to any of the previous items, wherein said cell is a cancer cell.
  • 242. The detection method according to any of the previous items, wherein said sample is derived from an organ selected from the group consisting of lymph nodes, kidney, liver, skin, brain, heart, muscles, bone marrow, skin, skeleton, lungs, the respiratory tract, spleen, thymus, pancreas, exocrine glands, bladder, endocrine glands, reproduction organs including the phallopian tubes, eye, ear, vascular system, the gastroinstestinal tract including small intestines, colon, rectum, canalis analis and prostate gland.
  • 243. The detection method according to any of the previous items, wherein the surface of sample cells is coated with proteases capable of cleaving a peptide label, for example by adding antibody-protease conjugates where the antibody recognizes a particular cell surface structure.
  • 244. The detection method according to any of the previous items, wherein the surface of sample cells is coated with DNA oligonucletides (“coating DNA”), for example by adding antibody-DNA conjugates where the antibody recognizes a particular cell surface structure.
  • 245. A method for detecting antigen-specific T cells in a sample, said method comprising providing a detection molecule and a detection method according to any of the preceding items.
  • 246. A method for detecting specific cells in a sample, said method comprising providing a detection molecule and a detection method according to any of the preceding items.
  • 247. A method for diagnosing a disease, said method comprising providing a detection molecule and a detection method according to any of the preceding items.
  • 248. A method for diagnosing a disease according to the previous items, wherein said disease is selected from the group consisting of cancer, Cancerous diseases, infectious diseases, Infectious diseases caused by virus, Infectious diseases caused by bacteria, Infectious diseases caused by fungus, Parasitic diseases, Allergic diseases, Transplantation-related diseases and Autoimmune and inflammatory diseases.
  • 249. A method for detecting the presence and/or abundance of a certain cell or cell type in a sample, said method comprising providing a detection molecule and a detection method according to any of the preceding items, wherein said detection molecule comprises a binding molecule capable of associating specifically with the cell or cell type in said sample
  • 250. A method for investigating the binding characteristics of a certain cell or cell type in a sample, said method comprising providing a detection molecule and a detection method according to any of the preceding items, wherein said detection molecule comprises a binding molecule capable of associating specifically with a known target.
  • 251. A method for the identification of epitopes comprising providing a detection molecule and a detection method according to any of the preceding items
  • 252. A method for vaccine development comprising providing a detection molecule and a detection method according to any of the preceding items.
  • 253. A method for measuring immune reactivity after vaccination comprising providing a detection molecule and a detection method according to any of the preceding items.
  • 254. A method for development of immune-therapeutics comprising providing a detection molecule and a detection method according to any of the preceding items.

Full text: Click here
Patent 2023
In four-week-old mice, calyceal innervation levels were determined by sparsely labeling calyces of Held using dye electroporation injections as previously described9 (link),14 (link),15 . Mice were transcardially perfused with artificial cerebrospinal fluid (aCSF; 130 mM NaCl, 3 mM KCl, 1.2 mM KH2PO4, 20 mM NaHCO3, 3 mM HEPES, 10 mM glucose, 2 mM CaCl2, 1.3 mM MgSO4 infused with 95% O2 and 5% CO2). Brains were quickly dissected and placed in a chamber with oxygenated aCSF. Brains remained in oxygenated aCSF for about 30 min and were then transferred to an aCSF-containing petri dish. A pulled glass micropipette was filled with rhodamine dextran amine (RDA; MW 3000, Invitrogen; in solution of 6.35% RDA with 0.4% Triton-X100 in PBS). RDA was electrophoresed at the rate of 5 pulses/second at 55 V for 50 ms using an Electro Square Porator (ECM830; BTX); multiple pulses were delivered at the midline to target the ventral acoustic stria (VAS). These pulses resulted in sparse dye-labeling of GBC axons and their calyceal terminals in MNTB on both sides. Brains were placed back into the aCSF chamber for approximately 2 h under continuous oxygenation to allow for dye transport. The tissue was then transferred to 4% PFA solution overnight followed by incubation in 30% sucrose solution in 0.1 M PBS. Brainstems were dissected and coronally cryosectioned at 18 µm in a series of 5 alternating slides. Tissue sections containing RDA-labeled calyces were immunolabeled for vesicular glutamate transporter 1/2 (VGLUT1/2) and counterstained with fluorescent Nissl.
Full text: Click here
Publication 2022
Acoustics Amines ARID1A protein, human Axon Bicarbonate, Sodium Brain Brain Stem Cell Respiration Cerebrospinal Fluid Electroporation Therapy Glucose HEPES Hyperostosis, Diffuse Idiopathic Skeletal Kidney Calices Mice, Laboratory Pulse Rate Pulses rhodamine dextran Sodium Chloride Striae Distensae Sucrose Sulfate, Magnesium Tissues Triton X-100 Vesicular Glutamate Transport Protein 1

Example 1

Cyclic heptapeptide cyclo(FΦRRRRQ) (cFΦR4, where Φ is L-2-naphthylalanine) was found to be efficiently internalized by mammalian cells. In this study, its mechanism of internalization was investigated by perturbing various endocytic events through the introduction of pharmacologic agents and genetic mutations. The results show that cFΦR4 can bind directly to membrane phospholipids, can be internalized into human cancer cells through endocytosis, and can escape from early endosomes into the cytoplasm. Its cargo capacity was examined with a wide variety of molecules including small-molecule dyes, linear and cyclic peptides of various charged states, and proteins. Depending on the nature of the cargos, they may be delivered by endocyclic (insertion of cargo into the cFΦR4 ring), exocyclic (attachment of cargo to the Gln side chain), or bicyclic approaches (fusion of cFΦR4 and cyclic cargo rings). The overall delivery efficiency (i.e., delivery of cargo into the cytoplasm and nucleus) of cFΦR4 was 4-12-fold higher than those of nonaarginine (R9), HIV Tat derived peptide (Tat), or penetratin (Antp). The higher delivery efficiency, coupled with superior serum stability, minimal toxicity, and synthetic accessibility, renders cFΦR4 a useful transporter for intracellular cargo delivery and a suitable system for investigating the mechanism of endosomal escape.

Introduction

The plasma membrane presents a major challenge in drug discovery, especially for biologics such as peptides, proteins and nucleic acids. One potential strategy to subvert the membrane barrier and deliver the biologics into cells is to attach them to “cell-penetrating peptides (CPPs)”. Since the initial observation that HIV trans-activator of transcription, Tat, internalizes into mammalian cells and activates viral replication in the late 1980s (Frankel, A D and Pabo, C O. Cell, 1988, 55, 1189-1193; Green, M and Loewenstein, P M. Cell, 1988, 55, 1179-1188) a large number of CPPs consisting of 6-20 residues have been reported (Langel, Ü. Cell-penetrating peptides: methods and protocols, Humana Press, New York, 2011, p xv; Schmidt, N et al. FEBS Lett., 2010, 584, 1806-1813; Futaki, S. Adv. Drug Delivery Rev., 2005, 57, 547-558; Stewart, K M et al. Org. Biomol. Chem., 2008, 6, 2242-2255; Deshayes, S et al. Cell. Mol. Life Sci., 2005, 62, 1839-1849; Goun, E A et al. ChemBioChem, 2005, 7, 1497-1515). CPPs have been used to deliver small-molecule drugs (Rothbard, J B et al. Nat. Med., 2000, 6, 1253-1257; Nori, A et al. Bioconjugate Chem., 2003, 14, 44-50), DNA (Hoyer, J and Neundorf, I. Acc. Chem. Res., 2012, 45, 1048-1056; Eguchi, A et al. J. Biol. Chem., 2001, 276, 26204-26210), RNA (Nakase, I et al. Acc. Chem. Res., 2012, 45, 1132-1139; Andaloussi, S E et al. Nucleic Acids Res., 2011, 39, 3972-3987; Jeong, J H et al. Bioconjugate Chem., 2009, 20, 5-14; Muratovska, A and Eccles, M R. FEBS Lett., 2004, 558, 63-68), proteins (Wadia, J S and Dowdy, S F. Adv. Drug Delivery Rev., 2005, 57, 579-596; Pooga, M et al. FASEB J., 2001, 15, 1451-1453; Schwarze, S R et al. Science, 1999, 285, 1569-1572), and nanoparticles (Josephson, L et al. Bioconjugate Chem., 1999, 10, 186-191; Gupta, B et al. Adv. Drug Delivery Rev., 2005, 57, 637-651; Liu, J et al. Biomacromolecules, 2001, 2, 362-8), into mammalian cells and tissues through either covalent attachment or electrostatic association. Many CPPs display minimal toxicity and immunogenicity at physiologically relevant concentrations (Saar, K et al. Anal. Biochem., 2005, 345, 55-65; Suhorutsenko, J et al. Bioconjugate Chem., 2011, 22, 2255-2262) and the incorporation of specific unnatural amino acids (Rueping, M et al. ChemBioChem, 2002, 3, 257-259) and other chemical moieties (Cooley, C B et al. J. Am. Chem. Soc., 2009, 131, 16401-16403; Pham, W et al. Chembiochem, 2004, 5, 1148-1151) have been found to increase stability and cytosolic delivery.

Despite three decades of investigation, the fundamental basis for CPP activity remains elusive. Two distinct and non-mutually exclusive mechanisms have been proposed for the CPPs whose primary sequences are characterized by having multiple arginine residues. In the first mechanism (direct membrane translocation), the arginine guanidinium groups interact with phospholipids of the plasma membrane to generate neutral ion pairs that passively diffuse across the membrane (Herce, H D and Garcia, A E. Proc. Natl. Acad. Sci. U.S.A., 2007, 104, 20805-20810; Hirose, H et al. Mol. Ther., 2012, 20, 984-993) or promote the formation of transient pores that permit the CPPs to traverse the lipid bilayer (Herce, H D et al. Biophys. J., 2009, 97, 1917-1925; Palm-Apergi, C et al. FASEB J., 2009, 23, 214-223). In the second mechanism, CPPs associate with cell surface glycoproteins and membrane phospholipids, internalize into cells through endocytosis (Richard, J P et al. J. Biol. Chem., 2005, 280, 15300-15306; Ferrari, A et al. Mol. Ther., 2003, 8, 284-294; Fittipaldi, A et al. J. Biol. Chem., 2003, 278, 34141-34149; Kaplan, I M et al. J. Controlled Release, 2005, 102, 247-253; Nakase, I et al. Biochemistry, 2007, 46, 492-501) and subsequently exit from endosomes into the cytoplasm. Taken together, the majority of data show that at low CPP concentrations, cellular uptake occurs mostly through endocytosis, whereas direct membrane translocation becomes prevalent at concentrations above 10 μM (Duchardt, F et al. Traffic, 2007, 8, 848-866). However, the mechanism(s) of entry and the efficiency of uptake may vary with the CPP identity, cargo, cell type, and other factors (Mueller, J et al. Bioconjugate Chem., 2008, 19, 2363-2374; Maiolo, J R et al. Biochim. Biophys. Acta., 2005, 1712, 161-172).

CPPs that enter cells via endocytosis must exit from endocytic vesicles in order to reach the cytosol. Unfortunately, the endosomal membrane has proven to be a significant barrier towards cytoplasmic delivery by these CPPs; often a negligible fraction of the peptides escapes into the cell interior (El-Sayed, A et al. AAPS J., 2009, 11, 13-22; Varkouhi, A K et al. J. Controlled Release, 2011, 151, 220-228; Appelbaum, J S et al. Chem. Biol., 2012, 19, 819-830). For example, even in the presence of the fusogenic hemagglutinin peptide HA2, which has been demonstrated to enhance endosomal cargo release, >99% of a Tat-Cre fusion protein remains entrapped in macropinosomes 24 h after initial uptake (Kaplan, I M et al. J. Controlled Release, 2005, 102, 247-253). Recently, two new types of CPPs with improved endosomal escape efficiencies have been discovered. Appelbaum et al. showed that folded miniature proteins containing a discrete penta-arginine motif were able to effectively overcome endosomal entrapment and reach the cytosol of mammalian cells (Appelbaum, J S et al. Chem. Biol., 2012, 19, 819-830). This motif consists of five arginines across three turns of an α-helix, and proteins containing this motif were released from early (Rab5+) endosomes into the cell interior. It has also been found that cyclization of certain arginine-rich CPPs enhances their cellular uptake (Qian, Z et al. ACS Chem. Biol., 2013, 8, 423-431; Lattig-Tunnemann, G et al. Nat. Commun., 2011, 2, 453; Mandal, D et al. Angew. Chem. Int. Ed., 2011, 50, 9633-9637; Zhao, K et al. Soft Matter, 2012, 8, 6430-6433). Small amphipathic cyclic peptides such as cyclo(FΦRRRRQ) (cFΦR4, where Φ is L-2-naphthylalanine) are internalized by mammalian cells in an energy-dependent manner, and enter the cytoplasm and nucleus with efficiencies 2-5-fold higher than that of nonaarginine (R9) (Qian, Z et al. ACS Chem. Biol., 2013, 8, 423-431). Moreover, membrane impermeable cargos such as phosphopeptides can be inserted into the cFΦR4 ring resulting in their delivery into the cytoplasm of target cells. However, insertion of a cargo into the cyclic peptide ring, which is referred to herein as the “endocyclic” delivery method (FIG. 1A), is limited to relatively short peptides (≤7 amino acids), as large rings display poor internalization efficiency (Qian, Z et al. ACS Chem. Biol., 2013, 8, 423-431).

To gain insight into the cFΦR4 mechanism of action and potentially design cyclic CPPs of still higher efficiency, herein the internalization mechanism of cFΦR4 was investigated through the use of artificial membranes and pharmacologic agents as well as genetic mutations that perturb various endocytic events. The data show that cFΦR4 can bind directly to the plasma membrane phospholipids and can enter cells through endocytosis. Like the miniature proteins displaying the penta-arginine motif (Appelbaum, J S et al. Chem. Biol., 2012, 19, 819-830), cFΦR4 can escape from the early endosomes into the cytosol. The ability of cFΦR4 to deliver a wide range of cargo molecules, including linear peptides of varying charges, cyclic peptides, and large proteins, into the cytoplasm of mammalian cells by exocyclic (attachment of cargo to the Gln side chain; FIG. 1B) or bicyclic delivery methods (fusion of the cFΦR4 and cyclic cargo rings; FIG. 1C) was also examined. It was found that cFΦR4 is tolerant to the size and nature of cargos and efficiently transported all of the cargos tested into the cytoplasm and nucleus of mammalian cells. In addition, cFΦR4 exhibits superior stability against proteolysis over linear CPPs but minimal cytotoxicity. cFΦR4 therefore provides a practically useful transporter for cytosolic cargo delivery as well as a system for investigating the mechanism of early endosomal cargo release.

Materials.

Reagents for peptide synthesis were purchased from Advanced ChemTech (Louisville, Ky.), NovaBiochem (La Jolla, Calif.), or Anaspec (San Jose, Calif.). 2,2′-Dipyridyl disulfide, Lissamine rhodamine B sulfonyl chloride, fluorescein isothiocyanate (FITC), dexamethasone (Dex), coenzyme A trilithium salt, FITC-labeled dextran (dextranFITC) and human serum were purchased from Sigma-Aldrich (St. Louis, Mo.). Cell culture media, fetal bovine serum (FBS), penicillin-streptomycin, 0.25% trypsin-EDTA, Hoescht 33342, Alexa488-labeled detran (dextranAlexa488), Dulbecco's phosphate-buffered saline (DPBS) (2.67 mM potassium chloride, 1.47 mM potassium phosphate monobasic, 137 mM sodium chloride, 8.06 mM sodium phosphate dibasic), and Lipofectamine 2000 were purchased from Invitrogen (Carlsbad, Calif.). PD-10 desalting columns were purchased from GE-Healthcare (Piscataway, N.J.). Nuclear staining dye DRAQ5™ was purchased from Thermo Scientific (Rockford, Ill.), while cell proliferation kit (MTT) was purchased from Roche (Indianapolis, Ind.). Anti-phosphotyrosine (pY) antibody (clone 4G10) was purchased from Millipore (Temecula, Calif.).

Rink resin LS (100-200 mesh, 0.2 mmol/g) was purchased from Advanced ChemTech. LC-SMCC (succinimidyl-4-[N-maleimidomethyl] cyclohexane-1-carboxy-[6-amidocaproate]) was purchased from Thermo Scientific (Rockford, Ill.), while 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho(1′-rac-glycerol) (sodium salt) (POPG), 1-palmitoyl-2-oleoyl-sn-glycero-3-phophoethanolamine (POPE), sphingomyelin (Brain, Porcine), and cholesterol were purchased from Avanti Polar Lipids (Alabaster, Ala.). Heparan sulfate (HO-03103, Lot #HO-10697) was obtained from Celcus Laboratories (Cincinnati, Ohio).

Peptide Synthesis and Labeling.

Peptides were synthesized on Rink Resin LS (0.2 mmol/g) using standard Fmoc chemistry. The typical coupling reaction contained 5 equiv of Fmoc-amino acid, 5 equiv of 2-(7-aza-H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU) and 10 equiv of diisopropylethylamine (DIPEA) and was allowed to proceed with mixing for 75 min. After the addition of the last (N-terminal) residue, the allyl group on the C-terminal Glu residue was removed by treatment with Pd(PPh3)4 and phenylsilane (0.1 and 10 equiv, respectively) in anhydrous DCM (3×15 min). The N-terminal Fmoc group was removed by treatment with 20% piperidine in DMF and the peptide was cyclized by treatment with benzotriazole-1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBOP)/HOBt/DIPEA (5, 5, and 10 equiv) in DMF for 3 h. The peptides were deprotected and released from the resin by treatment with 82.5:5:5:5:2.5 (v/v) TFA/thioanisole/water/phenol/ethanedithiol for 2 h. The peptides were triturated with cold ethyl ether (3×) and purified by reversed-phase HPLC on a C18 column. The authenticity of each peptide was confirmed by MALDI-TOF mass spectrometry.

Peptide labeling with FITC was performed by dissolving the purified peptide (˜1 mg) in 300 μL of 1:1:1 (vol/vol) DMSO/DMF/150 mM sodium bicarbonate (pH 8.5) and mixing with 10 μL of FITC in DMSO (100 mg/mL). After 20 min at room temperature, the reaction mixture was subjected to reversed-phase HPLC on a C18 column to isolate the FITC-labeled peptide. To generate rhodamine- and Dex-labeled peptides (FIG. 2), an Nε-4-methoxytrityl-L-lysine was added to the C-terminus. After the solid phase peptide synthesis, the lysine side chain was selectively deprotected using 1% (v/v) trifluoroacetic acid in CH2Cl2. The resin was incubated with Lissamine rhodamine B sulfonyl chloride/DIPEA (5 equiv each) in DMF overnight. The peptides were fully deprotected, triturated with diethyl ether, and purified by HPLC. The Dex-labeled peptide was produced by incubating the resin with dexamethasone-21-thiopropionic acid/HBTU/DIPEA (5, 5, and 10 equiv) in DMF for 3 h (Appelbaum, J S et al. Chem. Biol., 2012, 19, 819-830). The peptide was then deprotected, triturated, and purified by HPLC. Bicyclic peptides, phosphocoumaryl aminopropionic acid (pCAP), and pCAP-containing peptides (PCPs) were synthesized as previously described (Lian, W et al. J. Am. Chem. Soc., 2013, 135, 11990-11995; Mitra, S and Barrios, A M. Bioorg. Med. Chem. Lett., 2005, 15, 5124-5145; Stanford, S M et al. Proc. Natl. Acad. Sci. U.S.A., 2012, 109, 13972-13977). The authenticity of each peptide was confirmed by MALDI-TOF mass spectrometry.

Preparation of cFΦR4-Protein Conjugates.

The gene coding for the catalytic domain of PTP1B (amino acids 1-321) was amplified by the polymerase chain reaction using PTP1B cDNA as template and oligonucleotides 5′-ggaattccatatggagatggaaaaggagttcgagcag-3′ and 5′-gggatccgtcgacattgtgtggctccaggattcgtttgg-3′ as primers. The resulting DNA fragment was digested with endonucleases Nde I and Sal I and inserted into prokaryotic vector pET-22b(+)-ybbR (Yin, J et al. Proc. Natl. Acad. Sci. U.S.A., 2005, 102, 15815-15820). This cloning procedure resulted in the addition of a ybbR tag (VLDSLEFIASKL) to the N-terminus of PTP1B. Expression and purification of the ybbR tagged PTP1B were carried out as previously described (Ren, L et al. Biochemistry, 2011, 50, 2339-2356).

Peptide cFΦR4 containing a C-terminal cysteine (cFΦR4-SH, ˜10 μmol; FIG. 3) was dissolved in 1 mL of degassed DPBS and mixed with 2,2′-dipyridyl disulfide (5 equiv) dissolved in acetone (0.5 mL). After 2 h at room temperature, the reaction product cFΦR4-SS-Py was purified by reversed-phase HPLC. The product was incubated with coenzyme A (2 equiv) in DPBS for 2 h. The resulting cFΦR4-SS-CoA adduct was purified again by reversed-phase HPLC. Green fluorescent protein (GFP) containing an N-terminal ybbR tag (VLDSLEFIASKL) and a C-terminal six-histidine tag was expressed in Escherichia coli and purified as previously described (Yin, J et al. Proc. Natl. Acad. Sci. U.S.A., 2005, 102, 15815-15820). Next, ybbR-GFP (30 μM), cFΦR4-SS-CoA (30 μM), and phosphopantetheinyl transferase Sfp (0.5 μM) were mixed in 50 mM HEPES (pH 7.4), 10 mM MgCl2 (total volume 1.5 mL) and incubated at 37° C. for 15 min. The labeled protein, cFΦR4-S-S-GFP (FIG. 3), was separated from unreacted cFΦR4-SS-CoA by passing the reaction mixture through a PD-10 desalting column. GFP conjugated to Tat (Tat-S-S-GFP) and cFΦR4-conjugated PTP1B (cFΦR4-PTP1B) were prepared in a similar fashion (FIG. 4).

Peptide containing a C-terminal lysine (cFΦR4-Lys, ˜10 μmol; FIG. 4) was synthesized on the solid phase, deprotected and released from the support, dissolved in degassed DPBS (pH 7.4, 1 mL), and mixed with bifunctional linker LC-SMCC (5 equiv) dissolved in DMSO (0.2 mL). After incubation at room temperature for 2 h, the reaction product cFΦR4-SMCC (FIG. 4) was purified by reversed-phase HPLC equipped with a C18 column. The product was then mixed with coenzyme A (2 equiv) in DPBS and incubated for 2 h. The resulting cFΦR4-SMCC-CoA adduct was purified again by reversed-phase HPLC. Next, ybbR-tagged PTP1B (30 μM), cFΦR4-SMCC-CoA (30 μM), and phosphopantetheinyl transferase Sfp (0.5 μM) were mixed in 50 mM HEPES (pH 7.4), 10 mM MgCl2 (total volume of 1.5 mL) and incubated at 37° C. for 15 min. The labeled protein (cFΦR4-PTP1B; FIG. 4) was separated from unreacted cFΦR4-SMCC-CoA by passing the reaction mixture through a PD-10 desalting column eluted with DPBS.

Cell Culture and Transfection.

HEK293, HeLa, MCF-7, NIH 3T3 and A549 cells were maintained in medium consisting of DMEM, 10% FBS and 1% penicillin/streptomycin. Jurkat, H1650, and H1299 cells were grown in RPMI-1640 supplemented with 10% FBS and 1% penicillin/streptomycin. Cells were cultured in a humidified incubator at 37° C. with 5% CO2. For HeLa cells transfection, cells were seeded onto 96-well plate at a density of 10,000 cells/well. Following attachment, cells were transfected with plasmids encoding Rab5-green fluorescent protein fusion (Rab5-GFP), Rab7-GFP (Addgene plasmid #28047), glucocorticoid receptor (C638G)-GFP fusion (GR-GFP) (Holub, J M et al. Biochemistry, 2013, 50, 9036-6046), DsRed-Rab5 WT (Addgene plasmid #13050) or DsRed-Rab5Q79L (Addgene plasmid #29688) following Lipofectamine 2000 manufacturer protocols.

Preparation of Small Unilamellar Vesicles (SUVs).

SUVs were prepared by modifying a previously reported procedure (Magzoub, M et al. Biochim. Biophys. Acta, 2002, 1563, 53-63). A proper lipid mixture was dissolved in chloroform in a test tube. The lipid mixture was dried gently by blowing argon over the solution, and kept in a desiccator overnight. The dried lipids were rehydrated in DPBS to final total lipid concentration of 10 mM. The suspension was rigorously mixed by vortexing and sonication on ice until it became clear. A typical preparation yields a homogeneous solution containing vesicles with average diameter of ˜80 nm and polydispersity (PdI) index of <0.15 as determined by dynamic light scattering measurements using Zeta Sizer Nano Series (Malvern, Brookhaven, Conn.). The SUV solution was stored at 4° C. and used for FP experiments on the same day.

Fluorescence Polarization.

A typical experiment was performed by incubating 100 nM FITC-labeled peptide with varying concentrations of heparan sulfate (0-5,000 nM) in DPBS for 2 h at room temperature. The FP values were measured on a Molecular Devices Spectramax M5 spectrofluorimeter, with excitation and emission wavelengths at 485 and 525 nm, respectively. EC50 were determined by plotting the FP values as a function of heparan sulfate concentrations and fitted to a four-parameter logistic curve with GraphPad PRISM ver.6 software.

To obtain the EC50 value of CPP with lipid membranes, the FP experiment was similarly conducted using 100 nM FITC-labeled peptide with increasing concentrations of SUV solutions (0-10 mM) in DPBS. The FP values were similarly measured, plotted, and analyzed.

Image Analysis.

Raw images were uniformly modified using imageJ. Pearson's correlation coefficient (R) was obtained from endosomal regions using Just Another Colocalization Plugin (JACoP) (Bolte, S and Cordelieres, F P. J. Microsc., 2006, 224, 213-232). For GR-GFP translocation assay, individual GFP and Hoescht images were loaded into a customized CellProfiler pipeline and colored to grey (Carpenter, A E et al. Genome Biol., 2006, 7, R100). Nuclei were distinguished from the Hoescht image via Otsu automatic three-class thresholding, with pixels of the middle intensity class assigned to background. Clumped objects were identified using Laplacian of Gaussian modeling and separated by shape. The nuclear region was defined as the diameter of the Hoescht objects shrunken by 1 μm, while the cytosolic ring region was defined as the region between the nuclear diameter and the nuclear diameter expanded 2 μm. The translocation ratio was defined as the mean GFP signal inside the nuclear region divided by the mean GFP signal within the cytosolic region measured per cell, and 30-70 cells from 15-30 images were captured for each condition tested.

Confocal Microscopy.

To examine the co-localization between rhodamine-labeled cyclic peptide (cFΦR4Rho) and Rab5+ or Rab7+ endosomes, HeLa cells transfected with Rab5-GFP or Rab7-GFP were plated (200 μL, 104 cells/well, 96-well glass bottom MatriPlates) the day prior to the experiment. On the day of experiment, HeLa cells were treated with 1 μM cFΦR4Rho in DMEM media supplemented with 300 nM Hoescht 33342 for 30 min. After that, the cells were washed with HKR buffer (10 mM HEPES, pH 7.4, 140 mM NaCl, 2 mM KCl, 1 mM CaCl2), 1 mM MgCl2) and imaged using a PerkinElmer LiveView spinning disk confocal microscope.

For GR translocation assay, HeLa cells transfected with GR-GFP were plated as described above (Holub, J M et al. Biochemistry, 2013, 50, 9036-6046). The cells were treated for 30 min with DMEM media containing 1 μM Dex or Dex-peptide conjugate and 300 nM Hoescht 33342 and imaged using a Zeiss Axiovert 200M epifluorescence microscope outfitted with Ziess Axiocam mRM camera and an EXFO-Excite series 120 Hg arc lamp. To examine the effect of endocytosis inhibitors, transfected HeLa cells were pretreated for 30 min with clear DMEM containing the inhibitors before incubation with Dex or Dex-peptide conjugates. To test whether Rab5 activity is required for endosomal escape, HeLa cells were transfected with GR-GFP and DsRed-Rab5 WT or DsRed-Rab5Q79L before treatment with Dex or Dex-peptide conjugate and imaged as described above (Appelbaum, J S et al. Chem. Biol., 2012, 19, 819-830).

To examine the internalization of rhodamine-labeled peptides, 5×104 HEK293 cells were plated in a 35 mm glass-bottomed microwell dish (MatTek). On the day of experiment, the cells were incubated with the peptide solution (5 μM) and 0.5 mg/mL dextranFITC at 37° C. for 2 h. The cells were gently washed with DPBS twice and imaged on a Visitech Infinity 3 Hawk 2D-array live cell imaging confocal microscope. To detect the internalization of pCAP-containing peptides, HEK293 cells were similarly plated and incubated with the peptide solution (5 μM) at 37° C. for 60 min. After removal of the medium, the cells were gently washed with DPBS containing sodium pervanadate (1 mM) twice and incubated for 10 min in DPBS containing 5 μM nuclear staining dye DRAQ5. The resulting cells were washed with DPBS twice and imaged on a spinning disk confocal microscope (UltraView Vox CSUX1 system). To monitor GFP internalization, 5×104 HEK293 cells were seeded in a 35 mm glass-bottomed microwell dish and cultured overnight. Cells were treated with cFΦR4-S-S-GFP (1 μM) at 37° C. for 2 h. After removal of the medium, the cells were incubated in DPBS containing 5 μM DRAQ5 for 10 min. The cells were washed with DPBS twice and imaged on a Visitech Infinity 3 Hawk 2D-array live cell imaging confocal microscope.

Flow Cytometry.

To quantify the delivery efficiencies of pCAP-containing peptides, HeLa cells were cultured in six-well plates (5×105 cells per well) for 24 h. On the day of experiment, the cells were incubated with 10 μM pCAP-containing peptide in clear DMEM with 1% FBS at 37° C. for 2 h. The cells were washed with DPBS containing 1 mM sodium pervanadate, detached from plate with 0.25% trypsin, suspended in DPBS containing 1% bovine serum albumin, and analyzed on a BD FACS Aria flow cytometer with excitation at 355 nm. Data were analyzed with Flowjo software (Tree Star).

To estimate the effect of cFΦR4 on endocytosis, HeLa cells were seeded in six-well plates (5×105 cells per well) and allowed to adhere overnight. Following adherence, cells were treated with clear DMEM containing no supplement, 1 μM cFΦR4 peptide, 100 μM dextranAlexa488 (Life Technologies, D-22910), or both 1 μM cyclic peptide and 100 μM dextranAlexa488 for 30 min under standard cell culture conditions. The cells were washed with DPBS twice, removed from the plate with 0.25% trypsin, diluted into clear DMEM containing 10% FBS, pelleted at 300 g for 5 min, washed once with DPBS and resuspended in 200 μL of DPBS. Whole-cell dextran uptake was analyzed on a BD Accuri C6 flow cytometer using the manufacturer FL1 laser and filter set.

Immunoblotting.

NIH 3T3 cells were cultured in full growth media to reach 80% confluence. The cells were starved in serum free media for 3 h and treated with different concentrations of cFΦR4-PTP1B or untagged PTP1B for 2 h, followed by 30 min incubation in media supplemented with 1 mM sodium pervanadate. The solutions were removed and the cells were washed with cold DPBS twice. The cells were detached and lysed in 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% NP-40, 10 mM sodium pyrophosphate, 5 mM iodoacetic acid, 10 mM NaF, 1 mM EDTA, 2 mM sodium pervanadate, 0.1 mg/mL phenylmethanesulfonyl fluoride, 1 mM benzamidine, and 0.1 mg/mL trypsin inhibitor. After 30 min incubation on ice, the cell lysate was centrifuged at 15,000 rpm for 25 min in a microcentrifuge. The total cellular proteins were separated by SDS-PAGE and transferred electrophoretically to PVDF membrane, which was immunoblotted using anti-pY antibody 4G10.

Serum Stability Test.

The stability tests were carried by modifying a previously reported procedure (Nguyen, L T et al. PLoS One, 2010, 5, e12684). Diluted human serum (25%) was centrifuged at 15,000 rpm for 10 min, and the supernatant was collected. A peptide stock solution was diluted into the supernatant to a final concentration of 5 μM for cFΦR4 and Antp and 50 μM for peptides R9 and Tat and incubated at 37° C. At various time points (0-6 h), 200-μL aliquots were withdrawn and mixed with 50 μL of 15% trichloroacetic acid and incubated at 4° C. overnight. The final mixture was centrifuged at 15,000 rpm for 10 min in a microcentrifuge, and the supernatant was analyzed by reversed-phase HPLC equipped with a C18 column (Waters). The amount of remaining peptide (%) was determined by integrating the area underneath the peptide peak (monitored at 214 nm) and compared with that of the control reaction (no serum).

Cytotoxicity Assay.

MTT assays were performed to evaluate cyclic peptide's cytotoxicity against several mammalian cell lines (Mosmann, T. J. Immunol. Methods, 1983, 65, 55-63). One hundred μL of MCF-7, HEK293, H1299, H1650, A549 (1×105 cells/mL) cells were placed in each well of a 96-well culture plate and allowed to grow overnight. Varying concentrations of the peptide (5 or 50 μM) were added to the each well and the cells were incubated at 37° C. with 5% CO2 for 24 to 72 h. Ten μL of MTT stock solution was added into each well. Addition of 10 μL of the solution to the growth medium (no cell) was used as a negative control. The plate was incubated at 37° C. for 4 h. Then 100 μL of SDS-HCl solubilizing buffer was added into each well, and the resulting solution was mixed thoroughly. The plate was incubated at 37° C. for another 4 h. The absorbance of the formazan product was measured at 570 nm using a Molecular Devices Spectramax M5 plate reader. Each experiment was performed in triplicates and the cells without any peptide added were treated as control.

cFΦR4 Binds to Membrane Phospholipids.

It was previously observed that incubation of 1 μM FITC-labeled cyclic peptide cFΦR4FITC with vesicles containing negatively charged phospholipids (90% phosphatidylcholine (PC) and 10% phosphatidylglycerol (PG)) resulted in quenching of the peptide fluorescence, consistent with direct binding of cFΦR4 to phospholipids (Qian, Z et al. ACS Chem. Biol., 2013, 8, 423-431). To test the potential role of membrane binding during endocytic uptake of CPPs, SUVs that mimic the outer membrane of mammalian cells (45% PC, 20% phosphatidylethanolamine, 20% sphingomyelin, and 15% cholesterol) were prepared and tested for binding to FITC-labeled cFΦR4, R9, and Tat (each at 100 nM) by a fluorescence polarization (FP) assay. cFΦR4 bound to the neutral SUVs with an EC50 value (lipid concentration at which half of cFΦR4FITC is bound) of 2.1±0.1 mM (FIG. 5A). R9 showed much weaker binding to the artificial membrane (EC50>10 mM), whereas Tat did not bind at all. Next, the CPPs were tested for binding to heparan sulfate, which was previously proposed to be the primary binding target of cationic CPPs (Nakase, I et al. Biochemistry, 2007, 46, 492-501; Rusnati, M et al. J. Biol. Chem., 1999, 274, 28198-28205; Tyagi, M et al. J. Biol. Chem., 2001, 276, 3254-3261; Ziegler, A and Seelig, J. Biophys. J., 2004, 86, 254-263; Goncalves, E et al. Biochemistry, 2005, 44, 2692-2702; Ziegler, A. Adv. Drug Delivery Rev., 2008, 60, 580-597). R9 and Tat both bound to heparan sulfate with high affinity, having EC50 values of 144 and 304 nM, respectively (FIG. 5B). Under the same condition, cFΦR4 showed no detectable binding to heparan sulfate. These results are in agreement with the previous observations that non-amphipathic cationic CPPs (e.g., Tat and R9) bind tightly with cell surface proteoglycans (e.g. heparan sulfate) but only weakly with membrane lipids (Ziegler, A. Adv. Drug Delivery Rev., 2008, 60, 580-597). The insufficient number of positive charges of cFΦR4 is likely responsible for its lack of strong electrostatic interaction with heparan sulfate. On the other hand, the amphipathic nature and the more rigid cyclic structure of cFΦR4 should facilitate its binding to neutral lipid membranes. These data, together with the inhibition pattern by various endocytic inhibitors described above, suggest that cFΦR4 can bind directly to the plasma membrane phospholipids and can be internalized by all of the endocytic mechanisms in a piggyback manner.

Intracellular Delivery of Peptidyl Cargos.

Since endocyclic delivery by cFΦR4 is limited to a heptapeptide or smaller cargos (Qian, Z et al. ACS Chem. Biol., 2013, 8, 423-431), in this study the ability of cFΦR4 to deliver cargos of varying sizes and physicochemical properties attached to the Gln side chain (FIG. 1B, exocyclic delivery) was tested. First, positively charged (RRRRR), neutral (AAAAA), hydrophobic (FFFF), and negatively charged peptides [DE(pCAP)LI] were covalently attached to cFΦR4. The first three peptides were labeled with rhodamine B at a C-terminal lysine side chain (FIG. 2), and their internalization into HEK293 cells was examined by live-cell confocal microscopy. Cells incubated for 2 h with 5 μM peptide cFΦR4-A5 (FIG. 6A) or cFΦR4-R5 (FIG. 6B) showed evidence of both punctate and diffuse fluorescence, with the latter distributed almost uniformly throughout the cell. In contrast, the fluid phase endocytic marker dextranFITC displayed predominantly punctate fluorescence, indicative of endosomal localization. The diffuse rhodamine fluorescence suggests that a fraction of the peptides reached the cytosol and nucleus of the cells. Co-incubation of cells with cFΦR4 (1 μM) and dextranAlexa488 increased the internalization of the endocytic marker by 15% (FIG. 7), suggesting that cFΦR4 can activate endocytosis in cultured cells. cFΦR4-F4 was not tested due to its poor aqueous solubility.

Peptide cFΦR4-DE(pCAP)LI (cFΦR4-PCP; FIG. 2) was designed to test the ability of cFΦR4 to deliver negatively charged cargos as well as to compare the cytoplasmic delivery efficiency of cFΦR4 with those of other widely used CPPs such as R9, Tat, and penetratin (Antp). Thus, untagged PCP [Ac-DE(pCap)LI-NH2] and PCP conjugated to R9 (R9-PCP), Tat (Tat-PCP), or Antp (Antp-PCP) were also prepared. Note that cFΦR4-PCP carries a net charge of zero at physiological pH. pCAP is non-fluorescent, but upon entering the cell interior, should be rapidly dephosphorylated by endogenous protein tyrosine phosphatases (PTPs) to produce a fluorescent product, coumaryl aminopropionic acid (CAP, excitation 355 nm; emission 450 nm) (Mitra, S and Barrios, A M. Bioorg. Med. Chem. Lett., 2005, 15, 5124-5145; Stanford, S M et al. Proc. Natl. Acad. Sci. U.S.A., 2012, 109, 13972-13977). When assayed against a PTP panel in vitro, all four CPP-PCP conjugates were efficiently dephosphorylated (Table 8). This assay detects only the CPP-cargo inside the cytoplasm and nucleus, where the catalytic domains of all known mammalian PTPs are localized (Alonso, A et al. Cell, 2004, 117, 699-711). Further, CAP is fluorescent only in its deprotonated state (pKa=7.8); even if some dephosphorylation occurs inside the endosome (pH 6.5-4.5) or lysosome (pH 4.5), it would contribute little to the total fluorescence (FIG. 8). Treatment of HEK293 cells with 5 μM cFΦR4-PCP for 60 min resulted in diffuse blue fluorescence throughout the cell, suggesting that cFΦR4-PCP reached the cell interior, whereas the untagged PCP failed to enter cells under the same condition (FIG. 9A). When HEK293 cells were pretreated with the PTP inhibitor sodium pervanadate for 1 h prior to incubation with cFΦR4-PCP (5 μM), the CAP fluorescence in the cells diminished to background levels. HEK293 cells treated with R9-PCP, Antp-PCP, or Tat-PCP under identical conditions showed weak fluorescence, consistent with the poor ability of these peptides to access the cell interior (FIG. 9A). To quantify the relative intracellular PCP delivery efficiency, HeLa cells were treated with each peptide and analyzed by fluorescence activated cell sorting (FIG. 9B). cFΦR4-PCP was most efficiently internalized by the HeLa cells, with a mean fluorescence intensity (MFI) of 3510 arbitrary units (AU), whereas R9-PCP, Antp-PCP, Tat-PCP, and untagged PCP produced MFI values of 960, 400, 290, and 30 AU, respectively (FIG. 9C). Again, when cells were treated with cFΦR4-PCP in the presence of sodium pervanadate, the amount of CAP fluorescence was reduced to near background levels (70 AU). Thus, cFΦR4 is capable of delivering peptidyl cargos of varying physicochemical properties into the cytoplasm with efficiencies 3.7-12-fold higher than R9, Antp, and Tat.

TABLE 8
Kinetic Activities (kcat/KM, M−1 s−1) of Recombinant PTPs
against pCAP-Containing Peptidesa
PTPcFΦR4-PCPTat-PCPR9-PCPAntp-PCP
PTP1B37100138001470017400
TCPTP2780560457970
SHP2740022902482210
CD453510021800294022300
VHR2460146062402030
akcat/KM was measured as previously described (Ren, L et al. Biochemistry, 2011, 50, 2339-2356).

Intracellular Delivery of Cyclic Peptides.

In recent years, there has been much interest in cyclic peptides as therapeutic agents and biomedical research tools (Driggers, E M et al. Nat. Rev. Drug Discov., 2008, 7, 608-624; Marsault, E and Peterson, M L. J. Med. Chem., 2011, 54, 1961-2004). For example, cyclic peptides are effective for inhibition of protein-protein interactions (Lian, W et al. J. Am. Chem. Soc., 2013, 135, 11990-11995; Liu, T et al. ACS Comb. Sci., 2011, 13, 537-546; Dewan, V et al. ACS Chem. Biol., 2012, 7, 761-769; Wu, X et al. Med. Chem. Commun., 2013, 4, 378-382), which are challenging targets for conventional small molecules. A major obstacle in developing cyclic peptide therapeutics is that they are generally impermeable to the cell membrane (Kwon, Y U and Kodadek, T. Chem. Biol., 2007, 14, 671-677; Rezai, T et al. J. Am. Chem. Soc., 2006, 128, 2510-2511; Chatterjee, J et al. Acc. Chem. Res., 2008, 41, 1331-1342). The attempt to deliver cyclic peptides by cFΦR4 by the endocyclic method had only limited success; increase in the cargo size from 1 to 7 residues led to progressively poorer cellular uptake, likely because the larger, more flexible rings bind more poorly to the cell membrane (Qian, Z et al. ACS Chem. Biol., 2013, 8, 423-431). To overcome this limitation, a bicyclic peptide system was explored, in which one ring contains a CPP motif (e.g., FΦR4) while the other ring consists of peptide sequences specific for the desired targets (FIG. 1C). The bicyclic system should in principle be able to accommodate cargos of any size, because the cargo does not change the structure of the CPP ring and should have less impact on its delivery efficiency. The additional rigidity of a bicyclic structure should also improve its metabolic stability as well as the target-binding affinity and specificity. The bicyclic peptides were readily synthesized by forming three amide bonds between a trimesoyl scaffold and three amino groups on the corresponding linear peptide (i.e., the N-terminal amine, the side chain of a C-terminal diaminopropionic acid (Dap), and the side chain of a lysine (or ornithine, Dap) imbedded in between the CPP and target-binding motifs) (Lian, W et al. J. Am. Chem. Soc., 2013, 135, 11990-11995). To test the validity of this approach, FΦR4 was chosen in the C-terminal ring as the CPP moiety and peptides of different lengths and charges (AAAAA, AAAAAAA, RARAR, or DADAD) were chosen as cargo (Table 8, compounds 13-16). For comparison, two monocyclic peptides containing FΦR4 as transporter and peptides A5 and A7 as cargos (Table 8, compounds 17 and 18) were also prepared. All of the peptides were labeled at a C-terminal lysine side chain with rhodamine B (FIG. 2) and their internalization into HEK293 cells was examined by live-cell confocal microscopy. Treatment of cells with 5 μM peptide for 2 h resulted in efficient internalization of all six peptides (FIG. 10), although FACS analysis indicated that the uptake of bicyclo(FΦR4-A5)Rho was ˜3-fold more efficient than the corresponding monocyclic peptide (compound 17). The intracellular distribution of the internalized peptides was quite different between the bicyclic and monocyclic peptides. While the four bicyclic peptides showed evidence for their presence in both the cytoplasm/nucleus (as indicated by the diffuse rhodamine fluorescence) and the endosomes (as indicated by the fluorescence puncta), the monocyclic peptides exhibited predominantly punctate fluorescence that overlapped with that of the endocytic marker dextranFITC. In all cases, the endocytic marker displayed only punctate fluorescence, indicating that the endosomes were intact in the cells treated with the peptides. These results indicate that the increased structural rigidity of the bicyclic peptides facilitates both the initial uptake by endocytosis and endosomal release, presumably because of their improved binding to the plasma and endosomal membranes. The bicyclic system may provide a general strategy for intracellular delivery of cyclic and bicyclic peptides.

Intracellular Delivery of Protein Cargos.

To test whether cFΦR4 is capable of transporting full-length proteins into mammalian cells, GFP was attached to the N-terminus of cFΦR4 through a disulfide bond (FIG. 11A and FIG. 3). GFP was chosen because of its intrinsic fluorescence. The disulfide exchange reaction is highly specific, efficient, and reversible; upon entering the cytoplasm, the CPP-S-S-protein conjugate can be rapidly reduced to release the native protein. Although cFΦR4 can be directly attached to a native or engineered surface cysteine residue(s) on a cargo protein, a GFP variant containing a 12-amino acid ybbR tag at its N-terminus was used and phosphopantetheinyl transferase Sfp was used to enzymatically attach cFΦR4 to the ybbR tag (Yin, J et al. Proc. Natl. Acad. Sci. U.S.A., 2005, 102, 15815-15820). This permitted the attachment of a single cFΦR4 unit to GFP in a site-specific manner. For comparison, a Tat-S-S-GFP conjugate was generated in the same manner. Incubation of HEK293 cells in the presence of 1 μM cFΦR4-S-S-GFP resulted in accumulation of green fluorescence inside the cells (FIG. 11B). The fluorescence signal was diffuse and present throughout the entire cell volume, but with higher concentrations in the nucleus. Some of the cells contained small spots of intense green fluorescence (indicated by arrows in FIG. 11B), which may represent endosomally sequestered cFΦR4-S-S-GFP or aggregated GFP inside the cell. The untagged GFP was unable to enter cells, whereas Tat-S-S-GFP entered cells less efficiently than cFΦR4-S-S-GFP (FIG. 11B); FACS analysis of HaLa cells treated with 1 μM protein revealed a 5.5-fold higher total intracellular fluorescence for the latter. The fluorescence puncta in the cell periphery as well as lack of any detectable fluorescence in the nuclear region of Tat-S-S-GFP treated cells indicate that Tat-S-S-GFP is mostly entrapped in the endosomes, in agreement with previous reports (Kaplan, I M et al. J. Controlled Release, 2005, 102, 247-253). Thus, with a protein as cargo, cFΦR4 also has higher efficiency than Tat with regard to both initial uptake and endosomal escape.

To demonstrate the generality of cFΦR4 for protein delivery, a functional enzyme, the catalytic domain of PTP1B (amino acids 1-321), was chosen to be delivered into the cell interior. To show that a non-cleavable linkage is also compatible with the delivery method, cFΦR4 was conjugated to ybbR-tagged PTP1B via a thioether bond (cFΦR4-PTP1B) (FIG. 4). In vitro assay using p-nitrophenyl phosphate as substrate showed that addition of the cFΦR4 tag does not affect the catalytic activity of PTP1B (Table 9). NIH 3T3 cells were incubated for 2 h in the presence of untagged PTP1B or cFΦR4-PTP1B and their global pY protein levels were analyzed by anti-pY western blotting (FIG. 12A). Treatment of the cells with cFΦR4-PTP1B, but not untagged PTP1B, resulted in concentration-dependent decrease in pY levels of most, but not all, proteins. The total cellular protein levels, as detected by Coomassie blue staining, were unchanged (FIG. 12B), indicating that the observed decrease in pY levels was due to dephosphorylation of the pY proteins by cFΦR4-PTP1B and/or secondary effects caused by the introduction of cFΦR4-PTP1B (e.g., inactivation of cellular protein tyrosine kinases). Interestingly, different proteins exhibited varying dephosphorylation kinetics. Several proteins in the 150-200 kD range were completely dephosphorylated upon the addition of 62 nM cFΦR4-PTP1B, whereas proteins of ˜80 kD remained phosphorylated at 500 nM cFΦR4-PTP1B. The changes in the pY pattern are consistent with the broad substrate specificity of PTP1B (Ren, L et al. Biochemistry, 2011, 50, 2339-2356) and very similar to that caused by overexpression of PTP1B inside the cytosol of mammalian cells (LaMontagne Jr., K R et al. Proc. Natl. Acad. Sci. U.S.A., 1998, 95, 14094-14099). These results indicate that cFΦR4 can deliver PTP1B into the interior of NIH 3T3 cells in the catalytically active form and to sufficient levels to perturb the cell signaling process. cFΦR4 thus provides a tool for introducing other functional proteins, especially proteins that cannot be genetically expressed (e.g., toxic and chemically modified proteins), into mammalian cells in order to study their cellular functions.

TABLE 9
Kinetic Activities (kcat/KM, M−1 s−1) of PTP1B and
cFΦR4-PTP1B against pNPPa
enzymekcat/KM (M−1 s−1)
PTP1B1340
cFΦR4-PTP1B1600
apNPP = p-nitrophenyl phosphate; kcat/KM was measured as previously described (Ren, L et al. Biochemistry, 2011, 50, 2339-2356).

Stability and Cytotoxicity of cFΦR4.

The relative stability of cFΦR4, R9, Tat, and Antp (Table 8, compounds 19-22) against proteolytic degradation was determined by incubating the CPPs in 25% human serum at 37° C. and following the disappearance of the full-length peptides by reversed-phase HPLC. The cationic tryptophan-containing peptide, Antp, was least stable among the four CPPs; it was degraded at a half-life of <20 min and was completely digested after 2 h (FIG. 13A). R9 and Tat were slightly more stable than Antp, having half-lives of ˜30 min. In contrast, cFΦR4 was remarkably stable against serum proteases. There was less than 10% degradation after 6 h of incubation; after 24 h of incubation in the serum, >70% of cFΦR4 remained intact. Numerous other studies have also demonstrated that cyclization of peptides increases their proteolytic stabilities (Nguyen, L T et al. PLoS One, 2010, 5, e12684). The potential cytotoxicity of cFΦR4 was assessed by MTT assays with five different human cell lines (HEK293, MCF-7, A549, H1650, and H1299). After 24 or 48 h of incubation with up to 50 μM cFΦR4, there was no significant growth inhibition for any of the cell lines (FIG. 13B and FIG. 14). After 72 h, a slight growth inhibition (up to 20%) was observed at 50 μM (FIG. 14). Thus, cFΦR4 is relatively nontoxic to mammalian cells.

In this study, it was demonstrated that cFΦR4 can be effective for exocyclic delivery of small-molecule, peptide, and protein cargos into the cytoplasm and nucleus of mammalian cells. By using a pCAP-containing peptide as cargo/reporter, it was shown that cFΦR4 can be 3.7-12-fold more efficient than R9, Tat, and Antp for cytoplasmic cargo delivery, making cFΦR4 one of the most active CPPs known to date. Although modification of polybasic CPPs such as addition of hydrophobic acyl groups has previously been reported to enhance cellular uptake by a similar magnitude (Pham, W et al. Chembiochem, 2004, 5, 1148-1151), these previous studies have not established whether the enhanced uptake translates into a similar increase in the cytoplasmic CPP concentration. The pCAP-based reporter system described herein can provide a simple, robust method to quantitatively assess the cytoplasmic delivery efficiency of other CPPs. Several lines of evidence indicate that cFΦR4 can enter cells through multiple endocytic mechanisms, including its failure to enter cells at 4° C. or in the presence of sodium azide, partial overlap between the fluorescence puncta of cFΦR4Rho and the fluid phase endocytic marker dextranFITC, colocalization of cFΦR4Rho and endosomal proteins Rab5 and Rab7, and decreased cFΦR4Dex uptake upon administration of endocytic inhibitors. The minimal effect of the PI3K inhibitor wortmannin and the Rab5 Q79L mutation on the cytoplasmic delivery of cFΦR4, in addition to the strong colocalization observed between cFΦR4 and Rab5+ endosomes, suggest that cFΦR4 can escape from early endosomes (FIG. 15). In comparison, Tat has been demonstrated to enter cells through endocytosis and release from late endosomes, while R9 escapes endosomes prior to Rab7 recruitment (Appelbaum, J S et al. Chem. Biol., 2012, 19, 819-830). Early endosomal release can offer advantages, especially for peptide and protein cargos, since it can minimize cargo degradation by late endosomal and lysosomal proteases and denaturation caused by acidification during endosomal maturation. Indeed, both GFP and PTP1B delivered into the cytoplasm by cFΦR4 were in their folded, active forms, as evidenced by the green fluorescence and the ability to dephosphorylate intracellular pY proteins, respectively. Additionally, due to its more rigid structure, cFΦR4 can be more stable against proteolytic degradation than linear peptides, and due to its smaller size, cFΦR4 can be less expensive to synthesize and potentially less likely to interfere with the cargo function. These properties can make cFΦR4 a useful transporter for cytosolic delivery of small-molecule to protein cargos. Direct protein delivery can provide a useful research tool, e.g., for studying the cellular function of a protein, as it can offer improved temporal control over DNA transfection and subsequent gene expression and can allow delivery of chemically modified proteins and proteins whose overexpression can cause toxicity. The ability of cFΦR4 to escape from early endosomes and its simple structure can also provide an excellent system for elucidating the mechanism of endosomal escape and the factors that influence the escape efficiency.

Full text: Click here
Patent 2022

Example 3

Cyclic peptides have great potential as therapeutic agents and research tools but are generally impermeable to the cell membrane. Fusion of the cyclic peptides with a cyclic cell-penetrating peptide can produce bicyclic peptides that can be cell permeable and can retain the ability to recognize specific intracellular targets. Application of this strategy to protein tyrosine phosphatase 1B and peptidyl prolyl cis-trans isomerase Pin1 resulted in potent, selective, proteolytically stable, and biologically active inhibitors against the enzymes.

Cyclic peptides (and depsipeptides) exhibit a wide range of biological activities (Pomilio, A B et al. Curr. Org. Chem. 2006, 10, 2075-2121). Several innovative methodologies have recently been developed to synthesize cyclic peptides, either individually (Meutermans, W D F et al. J. Am. Chem. Soc. 1999, 121, 9790-9796; Schafmeister, C E et al. J. Am. Chem. Soc. 2000, 122, 5891-5892; Sun, Y et al. Org. Lett. 2001, 3, 1681-1684; Kohli, R M et al. Nature 2002, 418, 658-661; Qin, C et al. J. Comb. Chem. 2004, 6, 398-406; Turner, R A et al. Org. Lett. 2007, 9, 5011-5014; Hili, R et al. J. Am. Chem. Soc. 2010, 132, 2889-2891; Lee, J et al. J. Am. Chem. Soc. 2009, 131, 2122-2124; Frost, J R et al. ChemBioChem 2013, 14, 147-160) or combinatorially (Eichler, J et al. Mol. Divers. 1996, 1, 233-240; Giebel, L B et al. Biochemistry 1995, 34, 15430-15435; Scott, C P et al. Proc. Nat. Acad. Sci. USA 1999, 96, 13638-13643; Millward, S W et al. J. Am. Chem. Soc. 2005, 127, 14142-14143; Sako, Y et al. J. Am. Chem. Soc. 2008, 130, 7232-7234; Li, S et al. Chem. Commun. 2005, 581-583; Joo, S H et al. J. Am. Chem. Soc. 2006, 128, 13000-13009; Heinis, C et al. Nat. Chem. Biol. 2009, 5, 502-507; Tse, B N et al. J. Am. Chem. Soc. 2008, 130, 15611-15626), and screen them for biological activity. A particularly exciting application of cyclic peptides is the inhibition of protein-protein interactions (PPIs) (Leduc, A M et al. Proc. Nat. Acad. Sic. USA 2003, 100, 11273-11278; Millward, S W et al. ACS Chem Biol 2007, 2, 625-634; Tavassoli, A et al. ACS Chem. Biol. 2008, 3, 757-764; Wu, X et al. Med. Chem. Commun. 2013, 4, 378-382; Birts, C N et al. Chem. Sci. 2013, 4, 3046-3057; Kawakami, T et al. ACS Chem. Biol. 2013, 8, 1205-1214; Lian, W et al. J. Am. Chem. Soc. 2013, 135, 11990-11995), which remain challenging targets for conventional small molecules. However, a major limitation of cyclic peptides is that they are generally impermeable to the cell membrane, precluding any application against intracellular targets, which include most of the therapeutically relevant PPIs. Although formation of intramolecular hydrogen bonds (Rezai, T et al. J. Am. Chem. Soc. 2006, 128, 14073-14080) or Nα-methylation of the peptide backbone (Chatterjee, J et al. Acc. Chem. Res. 2008, 41, 1331-1342; White, T R et al. Nat. Chem. Biol. 2011, 7, 810-817) can improve the membrane permeability of certain cyclic peptides, alternative strategies to increase the cell permeability of cyclic peptides are clearly needed.

Protein-tyrosine phosphatase 1B (PTP1B) is a prototypical member of the PTP superfamily and plays numerous roles during eukaryotic cell signaling. Because of its role in negatively regulating insulin and leptin receptor signaling, PTP1B is a valid target for treatment of type II diabetes and obesity (Elchelby, M et al. Science 1999, 283, 1544-1548; Zabolotny, J M et al. Dev Cell 2002, 2, 489-495). A large number of PTP1B inhibitors have been reported (He, R et al. in New Therapeutic Strategies for Type 2 Diabetes: Small Molecule Approaches. Ed. R. M. Jones, RSC Publishing 2012, pp 142), however, none of them have succeeded in the clinic. Designing PTP inhibitors is challenging because most of the phosphotyrosine (pY) isosteres, such as difluorophosphonomethyl phenylalanine (F2Pmp) (Burke Jr., T R et al. Biochem. Biophys. Res. Commun. 1994, 204, 129-134), are impermeable to the cell membrane. Additionally, because all PTPs share a similar active site, achieving selectivity for a single PTP has been difficult. Herein, a potentially general approach to designing cell-permeable cyclic peptidyl inhibitors against intracellular proteins such as PTP1B is reported.

Materials.

Fmoc-protected amino acids were purchased from Advanced ChemTech (Louisville, Ky.), Peptides International (Louisville, Ky.), or Aapptec (Louisville, Ky.). Fmoc-F2Pmp-OH was purchased from EMD Millipore (Darmstadt, Germany). Aminomethyl-ChemMatrix resin (0.66 mmol/g) was from SJPC (Quebec, Canada). Rink resin LS (100-200 mesh, 0.2 mmol/g) and N-(9-fluorenylmethoxycarbonyloxy) succinimide (Fmoc-OSu) were purchased from Advanced ChemTech. O-Benzotriazole-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HBTU), 2-(7-aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU), 1-hydroxybenzotriazole hydrate (HOBt) were purchased from Aapptec. Phenyl isothiocyanate in 1-mL sealed ampoules, fluorescein isothiocyanate (FITC), rhodamine B-labeled dextran (dextranRho) were purchased from Sigma-Aldrich. Cell culture media, fetal bovine serum (FBS), penicillin-streptomycin, 0.25% trypsin-EDTA, Dulbecco's phosphate-buffered saline (DPBS) (2.67 mM potassium chloride, 1.47 mM potassium phosphate monobasic, 137 mM sodium chloride, 8.06 mM sodium phosphate dibasic.), and anti-phospho-IR/IGF1R antibody were purchased from Invitrogen (Carlsbad, Calif.). Nuclear staining dye DRAQ5™ and anti-β-actin antibody were purchased from Thermo Scientific (Rockford, Ill.). Antibody 4G10 was purchased from Millipore (Temecula, Calif.). All solvents and other chemical reagents were obtained from Sigma-Aldrich (St. Louis, Mo.) and were used without further purification unless noted otherwise.

Cell Culture.

A549, HEK293, and HepG2 cells were maintained in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% FBS in a humidified incubator at 37° C. with 5% CO2.

Protein Expression, Purification and Labeling.

The gene coding for the catalytic domain of PTP1B (amino acids 1-321) was amplified by the polymerase chain reaction using PTP1B cDNA as template and oligonucleotides 5′-ggaattccatatggagatggaaaaggagttcgagcag-3′ and 5′-gggatccgtcgacattgtgtggctccaggattcgtttgg-3′ as primers. The resulting DNA fragment was digested with endonucleases Nde I and Sal I and inserted into prokaryotic vector pET-22b(+)-ybbR (Yin, J et al. Proc. Natl. Acad. Sci. U.S.A. 2005, 102, 15815-15820). This cloning procedure resulted in the addition of a ybbR tag (VLDSLEFIASKL) to the N-terminus of PTP1B. Expression and purification of the ybbR-tagged PTP1B were carried out as previously described (Ren, L et al. Biochemistry 2011, 50, 2339-2356). Texas Red labeling of PTP1B was carried out by treating the ybbR-tagged PTP1B protein (80 μM) in 50 mM HEPES, pH 7.4, 10 mM MgCl2 with Sfp phosphopantetheinyl transferase (1 μM) and Texas Red-CoA (100 μM) for 30 min at room temperature (Yin, J et al. Proc. Natl. Acad. Sci. U.S.A. 2005, 102, 15815-15820). The reaction mixture was passed through a G-25 fast-desalting column equilibrated in 30 mM HEPES, pH 7.4, 150 mM NaCl to remove any free dye molecules. The full-length human S16A/Y23A mutant Pin1 was expressed and purified from E. coli as previously described (Liu, T et al. J. Med. Chem. 2010, 53, 2494-2501).

Library Synthesis.

The cyclic peptide library was synthesized on 1.35 g of aminomethyl-ChemMatrix resin (0.57 mmol/g). The library synthesis was performed at room temperature unless otherwise noted. The linker sequence (BBM) was synthesized using standard Fmoc chemistry. The typical coupling reaction contained 5 equiv of Fmoc-amino acid, 5 equiv of HBTU and 10 equiv of diisopropylethylamine (DIPEA) and was allowed to proceed with mixing for 2 h. The Fmoc group was removed by treatment twice with 20% (v/v) piperidine in DMF (5+15 min), and the beads were exhaustively washed with DMF (6×). To spatially segregate the beads into outer and inner layers, the resin (after removal of N-terminal Fmoc group) was washed with DMF and water, and soaked in water overnight. The resin was quickly drained and suspended in a solution of Fmoc-Glu(δ-NHS)-OAll (0.10 equiv), Boc-Met-OSu (0.4 equiv) and N-methylmorpholine (2 equiv) in 20 mL of 1:1 (v/v) DCM/diethyl ether (Joo, S H et al. J. Am. Chem. Soc. 2006, 128, 13000-13009). The mixture was incubated on a carousel shaker for 30 min. The beads were washed with 1:1 DCM/diethyl ether (3×) and DMF (8×). Next, the Fmoc group was removed by piperidine treatment. Then, Fmoc-Arg(Pbf)-OH (4×), Fmoc-Nal-OH, and Fmoc-Phe-OH were sequentially coupled by standard Fmoc chemistry to half of the resin. The other half was coupled with the same amino acids in the reverse sequence. The resin was combined and the random sequence was synthesized by the split-and-pool method using 5 equiv of Fmoc-amino acids, 5 equiv HATU and 10 equiv DIPEA as the coupling agent. The coupling reaction was repeated once to ensure complete coupling at each step. For random positions, a 24-amino acid set was selected based on their structural diversity, metabolic stability, and commercial availability, including 10 proteinogenic α-L-amino acids (Ala, Asp, Gln, Gly, His, Ile, Ser, Trp, Pro, and Tyr), 5 nonproteinogenic α-L-amino acids (L-4-fluorophenylalanine (Fpa), L-homoproline (Pip), L-norleucine (Nle), L-phenylglycine (Phg) and L-4-(phosphonodifluoromethyl)phenylalanine (F2Pmp)), and nine α-D-amino acids (D-2-naphthylalanine (D-Nal), D-Ala, D-Asn, D-Glu, D-Leu, D-Phe, D-Pro, D-Thr, and D-Val). To differentiate isobaric amino acids during PED-MS analysis, 4% (mol/mol) of CD3CO2D was added to the coupling reactions of D-Ala, D-Leu, and D-Pro, while 4% CH3CD2CO2D was added to the Nle reactions. Fmoc-F2Pmp-OH (0.06 equiv) and Fmoc-Tyr-OH (0.54 equiv) was placed in the middle of the random positions using HATU/DIPEA. After the entire sequence was synthesized, the allyl group on the C-terminal Glu residue was removed by treatment with a DCM solution containing tetrakis(triphenylphosphine)palladium [Pd(PPh3)4, 0.25 equiv] and phenylsilane (5 equiv) for 15 min (3×). The beads were sequentially washed with 0.5% (v/v) DIPEA in DMF, 0.5% (w/v) sodium dimethyldithiocarbamate hydrate in DMF, DMF (3×), DCM (3×), and DMF (3×). The Fmoc group on the N-terminal random residue was removed by piperidine as described above. The beads were washed with DMF (6×), DCM (3×), and 1 M HOBt in DMF (3×). For peptide cyclization, a solution of PyBOP/HOBt/DIPEA (5, 5, 10 equiv, respectively) in DMF was mixed with the resin and the mixture was incubated on a carousel shaker for 3 h. The resin was washed with DMF (3×) and DCM (3×) and dried under vacuum for >1 h. Side-chain deprotection was carried out with a modified reagent K 78.5:7.5:5:5:2.5:1:1 (v/v) TFA/phenol/water/thioanisole/ethanedithiol/anisole/triisopropylsilane) for 3 h. The resin was washed with TFA and DCM and dried under vacuum before storage at −20° C.

Library Screening and Peptide Sequencing.

Library resin (100 mg, ˜300,000 beads) was swollen in DCM, washed extensively with DMF, doubly distilled H2O, and incubated in 1 mL of blocking buffer (PBS, pH 7.4, 150 mM NaCl, 0.05% Tween 20 and 0.1% gelatin) containing 20 nM Texas red-labeled PTP1B at 4° C. for 3 h. The beads were examined under an Olympus SZX12 microscope equipped with a fluorescence illuminator (Olympus America, Center Valley, Pa.) and the most intensely fluorescent beads were manually collected as positive hits. Beads containing encoding linear peptides were individually sequenced by partial Edman degradation-mass spectrometry (PED-MS) (Liu, T et al. J. Med. Chem. 2010, 53, 2494-2501).

Individual Peptide Synthesis and Labeling.

Monocyclic and bicyclic peptides were synthesized on Rink Resin LS (0.2 mmol/g) using standard Fmoc chemistry. For monocyclic peptides, after the last (N-terminal) residue was coupled, the allyl group on the C-terminal Glu residue was removed by treatment with Pd(PPh3)4 and phenylsilane (0.1 and 10 equiv, respectively) in anhydrous DCM (3×15 min). The N-terminal Fmoc group was removed by treatment with 20% (v/v) piperidine in DMF and the peptide was cyclized by treatment with PyBOP/HOBt/DIPEA (5, 5, and 10 equiv) in DMF for 3 h. For bicyclic peptides, the N-terminal Fmoc group was removed with piperidine and a trimesic acid was coupled on the N-terminal amine using HBTU as a coupling agent. The allyloxycarbonyl groups on the side chains of two Dap residues were removed by treatment with Pd(PPh3)4 and phenylsilane (0.1 and 10 equiv, respectively) in anhydrous DCM for 2 h. The resulting peptide was cyclized with PyBOP as described above. The peptides were deprotected and released from the resin by treatment with 82.5:5:5:5:2.5 (v/v) TFA/thioanisole/water/phenol/ethanedithiol for 2 h. The peptides were triturated with cold ethyl ether (3×) and purified by reversed-phase HPLC on a C18 column. The authenticity of each peptide was confirmed by MALDI-TOF mass spectrometry. Peptide labeling with FITC was performed by dissolving the purified peptide (˜1 mg) in 300 μL of 1:1:1 (vol/vol) DMSO/DMF/150 mM sodium bicarbonate (pH 8.5) and mixing with 10 μL of FITC in DMSO (100 mg/mL). After 20 min at room temperature, the reaction mixture was subjected to reversed-phase HPLC on a C18 column to isolate the FITC-labeled peptide.

PTP Inhibition Assay.

PTP assays were performed in a quartz microcuvette (total volume 150 μL). The reaction mixture contains 100 mM Tris-HCl, pH 7.4, 50 mM NaCl, 2 mM EDTA, 1 mM TCEP, 0-1 μM of PTP inhibitor, and 500 μM para-nitrophenyl phosphate (pNPP). The enzymatic reaction was initiated by the addition of PTP (final concentration 15-75 nM) and monitored continuously at 405 nm on a UV-VIS spectrophotometer. Initial rates were calculated from the reaction progress curves (typically <60 s). The half-maximal inhibition constant (IC50) was defined as the concentration of an inhibitor that reduced the enzyme activity to 50% and was obtained by plotting the rates (V) against the inhibitor concentration [I] and fitting the data against the equation

V = V 0 ( 1 + [ I ] IC 5 0 ) where V0 is the enzymatic reaction rate in the absence of inhibitor. The inhibition constant (Ki) was determined by measuring the initial rates at fixed enzyme concentration (15 nM) but varying concentrations of pNPP (0-24 mM) and inhibitor (0-112 nM). The reaction rate (V) was plotted against the pNPP concentration ([S]) and fitted against the equation

1 V = K × 1 [ S ] + 1 V max to obtain the Michaelis constant K. The Ki value was obtained by plotting the K values against the inhibitor concentration [I] and fitted to equation

K K 0 = 1 + [ I ] K i where K0 is the Michaelis constant in the absence of inhibitor ([I]=0).

Confocal Microscopy.

Approximately 5×104 A549 cells were seeded in 35-mm glass-bottomed microwell dish (MatTek) containing 1 mL of media and cultured for one day. A549 cells were gently washed with DPBS once and treated with the FITC-labeled PTP1B inhibitors (5 μM), dextranRho (1 mg mL−1) in growth media for 2 h at 37° C. in the presence of 5% CO2. The peptide-containing media was removed and the cells were washed with DPBS three times and incubated for 10 min in 1 mL of DPBS containing 5 μM DRAQ5. The cells were again washed with DPBS twice. Then the cells were imaged on a Visitech Infinity 3 Hawk 2D-array live cell imaging confocal microscope (with a 60× oil immersion lens) at 37° C. in the presence of 5% CO2. Live-cell confocal microscopic imaging of HEK293 cells after treatment with FITC-labeled Pin1 inhibitors were similarly conducted.

Immunoblotting.

A549 cells were cultured in full growth media to reach 80% confluence. The cells were starved in serum free media for 3 h and treated with varying concentrations of PTP1B inhibitors for 2 h, followed by 30 min incubation in media supplemented with 1 mM sodium pervanadate. The solutions were removed and the cells were washed with cold DPBS twice. The cells were detached and lysed in 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% NP-40, 10 mM sodium pyrophosphate, 5 mM iodoacetic acid, 10 mM NaF, 1 mM EDTA, 2 mM sodium pervanadate, 0.1 mg/mL phenylmethanesulfonyl fluoride, 1 mM benzamidine, and 0.1 mg/mL trypsin inhibitor. After 30 min incubation on ice, the cell lysate was centrifuged at 15,000 rpm for 25 min in a microcentrifuge. The total cellular proteins were separated by SDS-PAGE and transferred electrophoretically to a PVDF membrane, which was immunoblotted using anti-phosphotyrosine antibody 4G10. The same samples were analyzed on a separate SDS-PAGE gel and stained by Coomassie brilliant blue to ascertain equal sample loading in all lanes.

To test the inhibitor's effect on insulin signaling pathway, HepG2 cells were cultured to reach 80% confluence. The cells were starved for 4 h in serum free DMEM media before being treated with PTP1B inhibitor (2 h), followed by stimulation with 100 nM insulin for 5 min. The samples were analyzed by SDS-PAGE as described above and immunoblotted using anti-phospho-IR/IGF1R antibody. The PVDF membrane was also probed by anti-β-actin antibody as the loading control.

Serum Stability Test.

The stability tests were carried by modifying a previously reported procedure (Nguyen, L T et al. PLoS One 2010, 5, e12684). Diluted human serum (25%) was centrifuged at 15,000 rpm for 10 min, and the supernatant was collected. A peptide stock solution was diluted into the supernatant to a final concentration of 5 μM and incubated at 37° C. At various time points (0-24 h), 200-μL aliquots were withdrawn and mixed with 50 μL of 15% trichloroacetic acid and incubated at 4° C. overnight. The final mixture was centrifuged at 15,000 rpm for 10 min in a microcentrifuge, and the supernatant was analyzed by reversed-phase HPLC equipped with a C18 column. The amount of remaining peptide (%) was determined by integrating the area underneath the peptide peak (monitored at 214 nm) and comparing with that of the control reaction (no serum).

Fluorescence Anisotropy.

FA experiments were carried out by incubating 100 nM FITC-labeled peptide with varying concentrations of protein in 20 mM HEPES (pH 7.4), 150 mM NaCl, 2 mM magnesium acetate, and 0.1% bovine serum albumin (BSA) for 2 h at room temperature. The FA values were measured on a Molecular Devices Spectramax M5 plate reader, with excitation and emission wavelengths at 485 and 525 nm, respectively. Equilibrium dissociation constants (KD) were determined by plotting the FA values as a function of protein concentration and fitting the curve to the following equation:

Y = ( A min + ( A max × Q b Q f - A min ) ( ( L + x + K D ) - ( ( L + x + K D ) 2 - 4 L x ) 2 L ) ) ( 1 + ( Q b Q f - 1 ) ( ( L + x + K D ) - ( ( L + x + K D ) 2 - 4 L x ) 2 L ) ) where Y is the FA value at a given protein concentration x, L is the peptide concentration, Qb/Qf is the correction factor for fluorophore-protein interaction, Amax is the maximum FA value when all of the peptides are bound to protein, while Amin is the minimum FA value when all of the peptides are free. FA competition assay was performed by incubating 100 nM FITC-labeled Pin1 inhibitor 5 with 1 μM Pin1, followed by the addition of 0-5 μM unlabeled inhibitor. The FA values were measured similarly on a pate reader. IC50 values were obtained by plotting the FA values against the competitor concentration and curve fitting using the four-parameter dose-response inhibition equation (Prism 6, GraphPad).

A class of cell-penetrating peptides (CPPs), cyclo(Phe-Nal-Arg-Arg-Arg-Arg-Gln) (cFΦR4, where Φ or Nal is L-naphthylalanine), were recently discovered (Qian, Z et al. ACS Chem. Biol. 2013, 8, 423-431). Unlike previous CPPs, which are typically linear peptides and predominantly entrapped in the endosome, cFΦR4 can efficiently escape from the endosome into the cytoplasm. Short peptide cargos (1-7 aa) could be delivered into mammalian cells by directly incorporating them into the cFΦR4 ring. The possibility of developing bifunctional cyclic peptides containing both cell-penetrating and target-binding sequences as cell-permeable inhibitors against intracellular proteins was examined. To generate specific inhibitors against PTP1B, a one-bead-two-compound library was synthesized on spatially segregated ChemMatrix resin (Liu, R et al. J. Am. Chem. Soc. 2002, 124, 7678-7680), in which each bead displayed a bifunctional cyclic peptide on its surface and contained the corresponding linear peptide in its interior as an encoding tag (FIG. 23 and FIG. 24). The bifunctional cyclic peptides all featured the amphipathic CPP motif FΦR4 (or its inverse sequence RRRRΦF) on one side and a random pentapeptide sequence (X1X2X3X4X5) on the other side, where X2 represents a 9:1 (mol/mol) mixture of Tyr and F2Pmp while X1 and X3-X5 are any of the 24 amino acids that included 10 proteinogenic L-amino acids (Ala, Asp, Gln, Gly, His, Ile, Pro, Ser, Tyr, Trp), 5 unnatural α-L-amino acids (F2Pmp, L-4-fluorophenylalanine (Fpa), L-norleucine (Nle), L-phenylglycine (Phg), L-pipecolic acid (Pip)), and 9 α-D-amino acids (D-Ala, D-Asn, D-Glu, D-Leu, L-β-naphthylalanine (D-Nal), D-Phe, D-Pro, D-Thr, and D-Val). The use of 9:1 Tyr/F2Pmp ratio at the X2 position, together with a 5-fold reduction of the surface peptide loading, reduced the amount of F2Pmp-containing peptides at the bead surface by 50-fold, increasing the stringency and minimizing nonspecific binding during library screening (Chen, X et al. J. Comb. Chem. 2009, 11, 604-611). Screening of the library (theoretical diversity 6.6×105) against Texas red-labeled PTP1B resulted in 65 positive beads, which were individually sequenced by partial Edman degradation-mass spectrometry (PED-MS) (Thakkar, A et al. Anal. Chem. 2006, 78, 5935-5939) to give 42 complete sequences (Table 12). Interestingly, most of the selected PTP1B inhibitors contained the inverse CPP motif (RRRRΦF).

TABLE 12
Peptide Sequences Selected from Cyclic Peptide
Library against PTP1Ba.
SEQ IDBead
NO.No.Sequence
136 1Pro-Pip-Gly-F2Pmp-Tyr-Arg
137 2Ser-Pip-Ile-F2Pmp-F2Pmp-Arg
138 3Ile-His-Ile-F2Pmp-Ile-Arg
139 4Ala-D-Ala-Ile-F2Pmp-Pip-Arg
140 5Fpa-Ser-Pip-F2Pmp-D-Val-Arg
141 6Pip-D-Asn-Pro-F2Pmp-Ala-Arg
142 7Tyr-Phg-Ala-F2Pmp-Gly-Arg
143 8Ala-His-Ile-F2Pmp-D-Ala-Arg
144 9Gly-D-Asn-Gly-F2Pmp-D-Pro-Arg
14510D-Phe-Gln-Pip-F2Pmp-Ile-Arg
14611Ser-Pro-Gly-F2Pmp-His-Arg
14712Pip-Tyr-Ile-F2Pmp-His-Arg
14813*Ser-D-Val-Pro-F2Pmp-His-Arg
14914Ala-Ile-Pro-F2Pmp-D-Asn-Arg
15015Fpa-Ser-Ile-F2Pmp-Gln-Phe
15116Ala-D-Aa-Phg-F2Pmp-D-Phe-Arg
15217D-Asn-D-Thr-Phg-F2Pmp-Phg-Arg
15318*Ile-Pro-Phg-F2Pmp-Nle-Arg
15419Gln-Pip-Fpa-F2Pmp-Pip-Arg
15520D-Asn-Ala-Fpa-F2Pmp-Gly-Arg
15621D-Asn-D-Thr-Tyr-F2Pmp-Ala-Arg
15722D-Glu-Ala-Phg-F2Pmp-D-Val-Arg
15823Ile-D-Val-Phg-F2Pmp-Ala-Arg
15924Tyr-D-Thr-Phg-F2Pmp-Ala-Arg
16025D-Asn-Pip-Phg-F2Pmp-Ile-Arg
16126Pip-D-Asn-Trp-F2Pmp-His-Arg
16227Tyr-Pip-D-Val-F2Pmp-Ile-Arg
16328D-Asn-Ser-D-Ala-F2Pmp-Gly-Arg
16429*D-Thr-D-Asn-D-Val-F2Pmp-D-Ala-
Arg
16530D-Asn-D-Thr-D-Val-F2Pmp-D-Thr-
Arg
16631Ser-Ile-D-Thr-F2Pmp-Tyr-Arg
16732D-Asn-Fpa-D-Asn-F2Pmp-D-Leu-
Arg
16833Tyr-D-Asn-D-Asn-F2Pmp-Nle-Arg
16934D-Asn-Tyr-D-Asn-F2Pmp-Gly-Arg
17035Ala-Trp-D-Asn-F2Pmp-Ala-Arg
17136D-Val-D-Thr-His-F2Pmp-Tyr-Arg
17237Pro-Phg-His-F2Pmp-Pip-Arg
17338D-Asn-Phg-His-F2Pmp-Gly-Arg
17439Pro-Ala-His-F2Pmp-Gly-Arg
17540Ala-Tyr-His-F2Pmp-Ile-Arg
17641D-Asn-Pip-D-Glu-F2Pmp-Tyr-Arg
17742D-Val-Ser-Ser-F2Pmp-D-Thr-Arg
aFpa, L-4-fluorophenylalanine; Pip, L-homoproline; Nle, L-norleucine; Phg, L-phenylglycine; F2Pmp, L-4-(phosphonodifluoromethyl)phenylalanine.
*Sequences subjected to further analysis.

Three hit sequences (D-Thr-D-Asn-D-Val-F2Pmp-D-Ala-Arg-Arg-Arg-Arg-Nal-Phe-Gln (inhibitor 1), Ser-D-Val-Pro-F2Pmp-His-Arg-Arg-Arg-Arg-Nal-Phe-Gln (inhibitor 2), and Ile-Pro-Phg-F2Pmp-Nle-Arg-Arg-Arg-Arg-Nal-Phe-Gln (inhibitor 3)) were resynthesized and purified by HPLC. All three peptides are competitive PTP1B inhibitors (Table 13), with peptide 2 being most potent (K1=54 nM) (FIG. 25). Confocal microscopic analysis of human cells treated with fluorescein isothiocyanate (FITC)-labeled inhibitor 2 indicated poor cellular uptake of the peptide (FIG. 26a). It has previously been shown that as the size of the cargo inserted into the cFΦR4 ring increases, the cellular uptake efficiency of the cyclic peptides decreases (Qian, Z et al. ACS Chem. Biol. 2013, 8, 423-431). Larger rings can be more conformationally flexible and may bind less tightly to the cell surface receptors (e.g., membrane phospholipids) during endocytosis. The negatively charged F2Pmp may also interact intramolecularly with the FΦR4 motif and interfere with its CPP function.

TABLE 13
Potency of Selected Monocyclic Peptide Inhibitors against PTP1B
SEQMonocyclicIC50
ID NOInhibitorSequence(nM)
1781cyclo(D-Thr-D-Asn-D-Val-F2Pmp-D-Ala-Arg-Arg-~100
Arg-Arg-Nal-Phe-Gln)
1792cyclo(Ser-D-Val-Pro-F2Pmp-His-Arg-Arg-Arg-Arg- ~30
Nal-Phe-Gln)
1803cyclo(Ile-Pro-Phg-F2Pmp-Nle-Arg-Arg-Arg-Arg-Nal-~200
Phe-Gln)

To improve the cell permeability of inhibitor 2, a bicyclic system in which the CPP motif is placed in one ring whereas the target-binding sequence constitutes the other ring (FIG. 23) was explored. The bicyclic system keeps the CPP ring to a minimal size which, according to the previously observed trend (Qian, Z et al. ACS Chem. Biol. 2013, 8, 423-431), can result in more efficient cellular uptake. The bicyclic system should be able to accommodate cargos of any size, because incorporation of the latter does not change the size of CPP ring and, therefore, should not affect the delivery efficiency of the cyclic CPP. The use of a rigid scaffold (e.g., trimesic acid) may also help keep the CPP and cargo motifs away from each other and minimize any mutual interference. The smaller rings of a bicyclic peptide, compared to its monocyclic counterpart, can result in greater structural rigidity and improved metabolic stability.

To convert the monocyclic PTP1B inhibitor 2 into a bicyclic peptide, the Gln residue (used for attachment to the solid support and peptide cyclization) was replaced with (S)-2,3-diaminopropionic acid (Dap) and a second Dap residue was inserted at the junction of CPP and PTP1B-binding sequences (C-terminal to His) (FIG. 23). Synthesis of the bicycle was accomplished by the formation of three amide bonds between a trimesic acid and the N-terminal amine and the side chains of the two Dap residues (FIG. 27) (Lian, W et al. J. Am. Chem. Soc. 2013, 135, 11990-11995). Briefly, the linear peptide was synthesized on Rink amide resin using the standard Fmoc chemistry and NO-alloxycarbonyl (Alloc)-protected Dap. After removal of the N-terminal Fmoc group, the exposed amine was acylated with trimesic acid. Removal of the Alloc groups with Pd(PPh3)4 followed by treatment with PyBOP afforded the desired bicyclic structure. To facilitate labeling with fluorescent probes, a lysine was added to the C-terminus. The bicyclic peptide (peptide 4) was deprotected by TFA and purified to homogeneity by HPLC.

Bicyclic peptide 4 can act as a competitive inhibitor of PTP1B, with a KI value of 37 nM (FIG. 26b). It can be highly selective for PTP1B. When assayed against p-nitrophenyl phosphate as a substrate (500 μM), inhibitor 4 had IC50 values of 30 and 500 nM for PTP1B and TCPTP, respectively (FIG. 26c and Table 14). It exhibited minimal inhibition of any of the other PTPs tested (≤10% inhibition of HePTP, SHP-1, PTPRC, PTPH1, or PTPRO at 1 μM inhibitor concentration). Inhibitor 4 has improved cell permeability over peptide 2, as detected by live-cell confocal microscopy of A549 cells treated with FITC-labeled inhibitor 4 (FIG. 26a). The treated cells showed both diffuse fluorescence throughout the cytoplasm and nucleus as well as fluorescence puncta, indicating that a fraction of the inhibitors reached the cytoplasm and nucleus while the rest was likely entrapped in the endosomes. Incubation of inhibitor 4 in human serum for 24 h at 37° C. resulted in ˜10% degradation, whereas 91% of inhibitor 2 was degraded under the same condition (FIG. 28). Overall, inhibitor 4 compares favorably with the small-molecule PTP1B inhibitors reported to date (Qian, Z et al. ACS Chem. Biol. 2013, 8, 423-431) with respect to potency, selectivity over the highly similar TCPTP (17-fold), cell permeability, and stability.

TABLE 14
Selectivity of Bicyclic Inhibitor 4 against Various PTPsa
PTPPTP1BTCPTPHePTPPTPRCSHP1PTPROPTPH1
IC5030 ± 4500 ± 250NANANANANA
(nM)
aNA, no significant inhibition at 1 μM inhibitor.

Inhibitor 4 was next tested for its ability to perturb PTP1B function during cell signaling. Treatment of A549 cells with inhibitor 4 (0-5 μM) resulted in dose-dependent increases in the phosphotyrosine (pY) levels of a large number of proteins, consistent with the broad substrate specificity of PTP1B (Ren, L et al. Biochemistry 2011, 50, 2339) (FIG. 29a). Analysis of the same samples by Coomassie blue staining showed similar amounts of proteins in all samples (FIG. 29b), indicating that the increased pY levels reflected increased phosphorylation (or decreased PTP reaction) instead of changes in the total protein levels. Remarkably, the increase in tyrosine phosphorylation was already apparent at 8 nM inhibitor 4. Interestingly, further increase in inhibitor concentration beyond 1 μM reversed the effect on tyrosine phosphorylation, an observation that was also made previously by Zhang and co-workers with a different PTP1B inhibitor (Xie, L et al. Biochemistry 2003, 42, 12792-12804). To obtain further evidence that the intracellular PTP1B was inhibited by peptide 4, the pY level of insulin receptor (IR), a well-established PTP1B substrate in vivo (Elchelby, M et al. Science 1999, 283, 1544-1548; Zabolotny, J M et al. Dev Cell 2002, 2, 489-495), was monitored by immunoblotting with specific antibodies against the pY1162pY1163 site. Again, treatment with inhibitor 4 caused dose-dependent increase in insulin receptor phosphorylation up to 1 μM inhibitor and the effect leveled off at higher concentrations (FIG. 29c,d). Taken together, these data indicate that bicyclic inhibitor 4 can efficiently enter mammalian cells and can inhibit PTP1B in vivo. The decreased phosphorylation at higher inhibitor concentrations may be caused by nonspecific inhibition of other PTPs (which may in turn down regulate protein tyrosine kinases). It may also reflect the pleiotropic roles played by PTP1B, which can both negatively and positively regulate the activities of different protein kinases (Lessard, L et al. Biochim. Biophys. Acta 2010, 1804, 613).

To test the generality of the bicyclic approach, it was applied to design cell permeable inhibitors against peptidyl prolyl cis-trans isomerase Pin1, a potential target for treatment of a variety of human diseases including cancer (Lu, K P and Zhou, X Z. Nat. Rev Mol. Cell Biol. 2007, 8, 904-916), for which potent, selective, and biologically active inhibitors are still lacking (More, J D and Potter, A. Bioorg. Med. Chem. Lett. 2013, 23, 4283-91). Thus, a previously reported monocyclic peptide (5), which is a potent inhibitor against Pin1 in vitro (KD 258 nM) but membrane impermeable (Liu, T et al. J. Med. Chem. 2010, 53, 2494-2501), was fused with cFΦR4 (FIG. 30). In addition, the L-Tyr at the pThr+3 position was replaced with an Arg to improve the aqueous solubility. The resulting bicyclic peptide 6 bound Pin1 with a KD value of 131 nM (Table 15 and FIG. 31). Insertion of a D-Ala at the pThr+5 position to increase the separation between the Pin1-binding and cell-penetrating motifs improved the inhibitor potency by ˜2-fold (KD=72 nM for inhibitor 7). Inhibitor 7 competed with FITC-labeled inhibitor 5 for binding to Pin1 (FIG. 32), indicating that they both can bind to the Pin1 active site. Substitution of D-Thr for D-pThr of inhibitor 7 reduced its potency by ˜10-fold (KD=620 nM for inhibitor 8, Table 16), whereas further replacement of the pipecolyl residue with D-Ala abolished Pin1 inhibitory activity (peptide 9). The bicyclic inhibitors 7-9 were cell permeable (FIG. 33). Treatment of HeLa cells with inhibitor 7 resulted in time- and dose-dependent inhibition of cell growth (45% inhibition after 3-day treatment at 20 μM inhibitor 7), whereas the monocyclic inhibitor 5 and inactive peptide 9 had no effect (FIG. 34). Peptide 8 also inhibited cell growth, but to a lesser extent than inhibitor 7.

TABLE 15
Dissociation Constants of Monocyclic and Bicyclic Peptides against Pin1 as
Determined by FA Analysis
SEQPin1
ID NOInhibitorSequenceaKD (nM)
1815cyclo(D-Ala-Sar-D-pThr-Pip-Nal-Tyr-Gln)-Lys-NH2258 ± 65
1826bicyclo[Tm(D-Ala-Sar-D-pThr-Pip-Nal-Arg-Ala)-Dap-131 ± 44
(Phe-Nal-Arg-Arg-Arg-Arg-Dap)]-Lys-NH2
1837bicyclo[Tm(D-Ala-Sar-D-pThr-Pip-Nal-Arg-Ala-D-Ala)- 72 ± 21
Dap-(Phe-Nal-Arg-Arg-Arg-Arg-Dap)]-Lys-NH2
1848bicyclo[Tm(D-Ala-Sar-D-Thr-Pip-Nal-Arg-Ala-D-Ala)-620 ± 120
Dap-(Phe-Nal-Arg-Arg-Arg-Arg-Dap)]-Lys-NH2
1859bicyclo[Tm(D-Ala-Sar-D-Thr-D-Ala-Nal-Arg-Ala-D-Ala)->>6000
Dap-(Phe-Nal-Arg-Arg-Arg-Arg-Dap)]-Lys-NH2
aDap, L-2,3-diaminopropionic acid; Nal, L-β-naphthylalanine; Pip, L-pipecolic acid; Sar, sarcosine; Tm, trimesic acid. For FA analysis, all peptides were labeled at the C-terminal lysine side-chain with FITC.

In conclusion, a potentially general approach to designing cell-permeable bicyclic peptides against intracellular targets was developed. These preliminary studies show that replacement of the PTP1B-binding motif with other peptide sequences of different physicochemical properties also resulted in their efficient delivery into cultured mammalian cells. The availability of a general intracellular delivery method should greatly expand the utility of cyclic peptides in drug discovery and biomedical research.

Full text: Click here
Patent 2022

Top products related to «Rhodamine-dextran-amine dye»

Sourced in United States
The MW 3000 is a laboratory instrument designed for molecular weight determination. It utilizes advanced technology to precisely measure the molecular weights of a variety of samples, including polymers, proteins, and other macromolecules. The core function of the MW 3000 is to provide accurate and reliable molecular weight analysis data to support research and development activities in various scientific fields.

More about "Rhodamine-dextran-amine dye"