WO2024250037A1 - Thérapie cellulaire immunitaire ciblée à l'aide de trail et d'apoptose médiée par le tnf - Google Patents
Thérapie cellulaire immunitaire ciblée à l'aide de trail et d'apoptose médiée par le tnf Download PDFInfo
- Publication number
- WO2024250037A1 WO2024250037A1 PCT/US2024/032321 US2024032321W WO2024250037A1 WO 2024250037 A1 WO2024250037 A1 WO 2024250037A1 US 2024032321 W US2024032321 W US 2024032321W WO 2024250037 A1 WO2024250037 A1 WO 2024250037A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cell
- cells
- immune
- genetically modified
- subject
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
- C12N5/0638—Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5011—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2502/00—Coculture with; Conditioned medium produced by
- C12N2502/30—Coculture with; Conditioned medium produced by tumour cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2513/00—3D culture
Definitions
- Targeted cellular therapies for cancer treatment such as CAR-NK and CAR-T cell treatments, show some promise and have been approved for clinical use. However, these treatments are not universally effective for patients, and possess potential side effects that can present their own health risk to patients. There remains a need to develop safer and more effective uses of CAR-NK and CAR-T cells to treat cancer and infectious diseases.
- the present technology provides genetically modified immune cells, such as CAR-T and CAR-NK cells, with superior efficiency of killing target cells, such as cancer cells.
- a genetically modified lymphocyte model was developed that activates the tumor necrosis factor (TNF) pathway to kill cancer cells in combination with checkpoint inhibitors.
- TNF tumor necrosis factor
- the modified immune cells kill cancer cells more efficiently, and are expected to reduce resource demands, reduce side effects, and improve patient recovery rates.
- One aspect of the present technology provides a genetically modified immune cell expressing a chimeric antigen receptor (CAR) and also expressing an elevated level of a ligand for a tumor necrosis factor (TNF) receptor.
- CAR chimeric antigen receptor
- TNF tumor necrosis factor
- the genetically modified cell takes advantage of dual pathways for killing cancer cells, cellular pathogens, and cells infected with viruses.
- the CAR-mediated pathway promotes killing action by perforin and granzymes released by cytotoxic T lymphocytes and natural killer cells, as well as the TNF receptor (i.e., death domain) mediated target cell killing by these same cells.
- the present technology especially when combined with single cell image analysis, microfluidics based cell sorting, and transcriptomic analysis makes it possible to adapt genetic modifications to cells of a patient, in response to significant patient to patient variation in gene expression of T cells and NK cells.
- kits containing the genetically modified cell described above and one or more bispecific antibodies or aptamers having a first binding specificity for a tumor antigen and a second binding specificity for an immune cell.
- kits containing one or more viral vectors for genetically modifying an immune cell to express a CAR and to elevate an expression level of a ligand for a TNF receptor, and instructions or one or more reagents for carrying out the genetic modification.
- Still another aspect of the present technology is a method of producing a genetically modified cell.
- the method includes: (a) harvesting immune cells from a subject; (b) optionally screening the harvested immune cells for their ability to kill tumor cells of the subject and selecting harvested immune cells having desired function; and (c) genetically modifying the harvested or screened and selected immune cells for use in anti-tumor therapy for the subject.
- Even another aspect of the present technology is a method of enhancing the killing of tumor cells in a subject in need thereof.
- the method includes administering a plurality of the genetically modified cells described above to the subject, whereby killing of tumor cells in the subject is enhanced compared to the absence of the genetically modified cells.
- the technology can also be summarized in the following list of features.
- a genetically modified immune cell expressing a chimeric antigen receptor (CAR) and an elevated level of a ligand for a tumor necrosis factor (TNF) receptor.
- CAR chimeric antigen receptor
- TNF tumor necrosis factor
- the genetically modified immune cell of feature 1 wherein the cell is a T-lymphocyte or a natural killer cell.
- the CAR comprises an amino acid sequence of a tumor antigen selected from the group consisting of CD-19, NY-ESO-1 , mesothelin, PSA, MART-1 , MART-2, Gp100, tyrosinase, p53, ras, MUC1 , SAP-1 , survivin, CEA, Ep-CAM, Her2, BRCA1/2, and combinations thereof.
- a tumor antigen selected from the group consisting of CD-19, NY-ESO-1 , mesothelin, PSA, MART-1 , MART-2, Gp100, tyrosinase, p53, ras, MUC1 , SAP-1 , survivin, CEA, Ep-CAM, Her2, BRCA1/2, and combinations thereof.
- the genetically modified immune cell of any of the preceding features wherein the cell further expresses a soluble immune modulator or a soluble immune checkpoint inhibitor.
- the soluble immune modulator or soluble immune checkpoint inhibitor is selected from the group consisting of anti-CTLA-4, anti-PD-1 , anti-PDL-1 , anti-LAG-3, anti-TIM 3, anti-B7-H3, anti-ICOS, IDO inhibitors, 4-1 BB, anti-CD47, anti-B7-H4, anti-OX-40, anti-TIGIT, and anti-CD160, and combinations thereof.
- step (b) comprises analysis of individual harvested immune cells for their ability to kill tumor cells when paired in aqueous microdroplets or cell spheroids in a microfluidic device, and optionally wherein the screening comprises transcriptomic analysis by sequencing of RNA from said harvested immune cells collected from the aqueous microdroplets or cell spheroids.
- step (b) is performed, and wherein the selecting comprises use of a sorting module in the microfluidic device.
- step (c) comprises transducing the cell with a viral vector comprising a gene for expressing said CAR and a gene for modifying expression of said TNF receptor ligand.
- a kit comprising the genetically modified cell of any of the preceding features and one or more bispecific antibodies or aptamers having a first binding specificity for a tumor antigen and a second binding specificity for an immune cell.
- a kit comprising one or more viral vectors for genetically modifying an immune cell to express a CAR and to elevate an expression level of a ligand for a TNF receptor, and instructions or one or more reagents for carrying out the genetic modification.
- a method of producing a genetically modified cell comprising:
- step (b) is performed, and wherein the screening comprises analysis of individual harvested immune cells for their ability to kill tumor cells when paired in aqueous microdroplets or cell spheroids in a microfluidic device, and optionally wherein the screening comprises transcriptomic analysis by sequencing of RNA from said harvested immune cells collected from the aqueous microdroplets or cell spheroids.
- step (b) is performed, and wherein the selecting comprises use of a sorting module in the microfluidic device.
- step (c) comprises transducing the cell with a viral vector comprising a gene for expressing said CAR and a gene for modifying expression of said TNF receptor ligand.
- a method of enhancing killing of tumor cells in a subject in need thereof comprising administering the genetically modified cell of any of features 1-10 to the subject, whereby killing of tumor cells in the subject is enhanced.
- Fig. 1 A shows the layout of a microfluidic device for forming aqueous microdroplets in an oil stream, co-encapsulating immune cells and target cells in microdroplets, optionally storing and observing the microdroplets in a microwell array, and sorting and harvesting cells based on fluorescence.
- Fig. 1 B shows details of the sorting junction (panel (1)), and droplet docking stations (panels (2) and (3)).
- Fig. 2A shows a schematic illustration of a droplet sorting junction based on cell fluorescence.
- Fig. 2B shows a schematic illustration of a protocol for separating active NK cells from inactive NK cells.
- Fig. 2C shows a schematic illustration of a protocol for separating serial killer NK cells from less active NK cells.
- Fig. 3A shows a schematic illustration of a protocol for separating activated T cells based on an intracellular Ca 2+ transient.
- Fig. 3B shows a representation of how an antigen can be presented to a CD8+ T cell to activate it using an MHC tetramer loaded with the antigen.
- Fig. 30 shows two intracellular calcium signals, each indicating a sorted, activated T cell.
- Fig. 3D shows the time course of an intracellular calcium signal in a CD8+ T cell (left side of pair) activated by contact with antigen.
- Fig. 4, left shows the layout of a microfluidic device for forming, docking, observing, and collecting cell spheroids containing cancer cells and immune cells.
- the middle panel shows a higher magnification image of the docking array of the device containing cell spheroids.
- the right image is a still higher magnification of one spheroid at different times.
- Fig. 5 shows the relative expression levels of selected genes under the conditions indicated.
- Fig. 6A shows a volcano plot that demonstrates the total quantity of genes upregulated, downregulated, and unchanged between rCHOP and rituximab treated cells.
- Fig. 6B shows the relative expression levels of selected genes related to lipid metabolism after treatment with rituximab or rCHOP.
- Fig. 6C shows the relative expression levels of selected genes related to immune activity after treatment with rituximab or rCHOP.
- Fig. 6D shows the upregulation of the indicated biological processes.
- Fig. 6E shows the upregulation of the indicated cellular components.
- Fig. 6F shows the upregulation of the indicated molecular factors.
- Fig. 7A shows the relative expression of several genes of different indicated groups of NK cells for each of four donors.
- Figs. 7B-1 and 7B-2 shows relative gene expression levels for NK killers vs. non-killers for each of four donors.
- Fig. 7C shows the Iog2 fold change in expression for cytotoxicity-related genes for each of four donors after treatment with a CD3/CD19 bispecific antibody.
- Fig. 8A shows the relative expression levels for each of the genes indicated for each of four donors after treatment with a CD3/CD19 bispecific antibody.
- Fig. 8B shows the Iog2 fold change in expression for a series of T cell activation genes for each of four donors after treatment with the bispecific antibody.
- Fig. 8C shows the log 2 fold change in expression for the indicated genes for each of four donors after treatment with the bispecific antibody.
- CAR-NK cells are genetically modified to express surface receptors that target specific tumor cell markers, and upon binding release perforin and granzymes to kill the target cell. 1 While highly effective at killing target cells, perforin and granzyme are released into solution and also can kill healthy cells, including immune cells. 2 Additionally, the granzyme/perforin pathway has been shown to only effectively kill a single target cell, and in subsequent interactions other signaling mechanisms are used. 3 This mechanism transition is useful, given the limited quantity of the secreted factors that a cell can carry, and the time required to replenish cellular stores.
- the ability of a cytotoxic immune cell to kill multiple target cells is a highly desired feature for a cell-based immune therapy, due to the inability to deliver CAR-NK cells to a tumor at a 1 :1 ratio.
- the inventors have also studied transcriptomic features of CAR-NK and Wt-NK cells, separated using a microfluidic system into populations that could or could not kill target cancer cells. They found an expected high upregulation of perforin, granzymes, and IFNy in the CAR- NK killers; however, this was not mirrored in the Wt-NK killers. Instead, minor increases in expression of TNF factors were found. An increase in TNF and TNFSF10 expression by NK cells, and of TNFRSF1A and 21 on target cells in the Wt-killer NK cells compared to nonkillers, indicated that cytotoxicity is driven by the TNF pathway.
- CAR-NK cells showed a much higher expression of certain exhaustion markers, including KLRG1 , CD160 and HACVR2 (TIM3).
- KLRG1 KLRG1 , CD160 and HACVR2
- TIM3 HACVR2
- the wildtype NK cells showed relatively low expression of these markers, indicating that these cells are able to kill additional target cells.
- an upregulation of PDL1 expression was also observed in conditions where target cells were not killed, indicating that this signaling interferes both with CAR- and TNF-mediated apoptosis mechanisms.
- the present technology provides an immune cell, such as a T lymphocyte or NK cell, that is genetically modified to express a cancer antigen targeting Fc receptor (i.e., a CAR specific for a cancer antigen) and also modified to increase the expression of membrane-bound TNF ligands, such as TNFSF2 or TRAIL.
- a cancer antigen targeting Fc receptor i.e., a CAR specific for a cancer antigen
- membrane-bound TNF ligands such as TNFSF2 or TRAIL.
- these modifications are supplemented with a checkpoint inhibition mechanism, such as a PD1 or PDL1 inhibiting mechanism, either by further genetic modifications of the immune cell or by systematically delivering the checkpoint inhibitor during treatment.
- TNF signaling provides a slower but highly effective alternative to granzyme and perforin secretion, with reduced collateral damage to nearby healthy tissues.
- the scFV portion of a CAR is a powerful tool for bringing immune cells directly to their target cells.
- the use of anti-CD19 CARs in particular has proven useful for the treatment of lymphoma.
- CAR-NK cells can effectively find and connect with lymphoma cells.
- NK cells can produce multiple contact mediated kills without undergoing exhaustion. Combining this with PD1 receptor blocking is expected to greatly enhance the capability of cytotoxic immune cells to kill even resistant cancer cells.
- This design can be translated to other targets by changing the scFv portion of the CAR to recognize other antigens. Therefore genetically engineered immune cells of the present technology can be a powerful alternative to traditional CAR lymphocytes, particularly for solid tumors.
- the present technology provides genetically modified immune cells.
- the immune cells can be any type of immune cell that is part of the cell-mediated immunity (Th1) response, such as CD8+ T lymphocytes, cytotoxic T lymphocytes, and natural killer (NK) cells, and phagocytes such as macrophages.
- the immune cells are genetically modified to express an antigenspecific chimeric antigen receptor (CAR) whose antigen-binding specificity is selected to target a desired antigen, such as an antigen specific for cancer cells of a subject who has cancer, or a certain type of cancer, either a solid tumor, or a blood-born cancer, or other cancer.
- CAR chimeric antigen receptor
- one or more immune cells which can be cells of the human or other mammalian subject to be treated, or cells of another individual, is transduced with a viral vector, such as a lentiviral vector or other viral vector.
- a viral vector such as a lentiviral vector or other viral vector.
- the vector comprises a nucleic acid sequence that encodes the CAR, and also can comprise regulatory sequences such as a promoter, enhancer, and/or sequences that mediate insertion of the vector nucleic acid into the host cell’s genome.
- the structure of CARs is well known.
- a CAR has three domains: an extracellular scFv antibody domain that specifically binds the selected antigen, a membrane embedded domain, and a signal transduction domain within the cell, such as CD3zeta from the T cell receptor, which activates the cell upon binding of the scFv domain to its antigen.
- Any generation of CAR technology including any of the three generations currently available (Generations 2 and 3 add one or two co-stimulatory domains to the CD3zeta domain of Generation 1) can be used to prepare genetically modified immune cells as described herein.
- An example of a lentiviral vector for expressing a CAR in T cells can be found at en. vectorbuilder.com/resources/vector-system/pLV_Exp_CAR.html.
- the CAR expressed in an immune cell according to the present technology can have binding specificity for any desired antigen.
- Preferred are cancer cell-specific antigens or antigens specific for cellular pathogens.
- suitable cancer cell antigens include CD-19, NY-ESO-1 , mesothelin, PSA, MART-1 , MART-2, Gp100, tyrosinase, p53, ras, MUC1 , SAP-1 , survivin, CEA, Ep-CAM, Her2, and BRCA1/2.
- antigens specific for cellular pathogens include cell surface proteins, glycoproteins, or oligosaccharides of protozoans, parasites, and bacteria.
- epitopes to be targeted for binding by the CAR are well known and can be selected from the literature or by other means known to the person of ordinary skill.
- a viral vector used to transduce a cytotoxic immune cell contains one or more nucleic acid sequences that encode a membrane-bound TNF receptor ligand, in addition to sequences required for transduction and expressing the CAR.
- the viral vector can be a lentiviral vector or a gamma retroviral vector, for example.
- Nucleic acid sequences encoding TNF ligands such as TRAIL, FASL, TNF-a, LT, RANKL, APRIL, BAFF, LIGHT, and VEGI (see Dostert, C Cincinnati et al., The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond, Physiol. Rev.
- TNF ligands such as TRAIL, FASL, TNF-a that bind to a TNF receptor that contains a cytosolic death domain, such as DR4, DR5, Fas, or TNFR1.
- Some TNF receptor ligands exhibit cross-reactivity that allows them to bind to more than one TNF receptor.
- the corresponding TNF receptor will be found on the target cell, such as a cancer cell, and preferably is not significantly expressed on normal cells.
- Nucleic acid sequences encoding TNF ligands, including those described above, and suitable for expression from a viral vector in a cytotoxic immune cell are well known, and any such sequences can be used.
- the transgene for expressing the TNF receptor ligand can be positioned in the vector such that it is under control of the same promoter as that which controls CAR expression, or it can follow a different promoter.
- a variety of promoters are known which can be selected based on their ability to regulate expression in the type of host cell and/or their ability to cause expression of the TNF receptor ligand at a desired level in the host cell. It is preferred to express the TNF receptor ligand at a higher level in the genetically modified immune cell than it may be expressed normally in the absence of transduction by the viral vector.
- expression of the TNF ligand can be increased by 20%, 50% 100%, 1 .5- fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, or more compared to the non-transduced host cell, so that optimal effectiveness in killing of targeted cells is achieved,
- the viral vector used to transduce a cytotoxic immune cell and express a CAR and a TNF receptor ligand can optionally further include one or more nucleic acid sequences encoding a soluble immune modulator or checkpoint inhibitor.
- suitable soluble immune modulators or checkpoint inhibitors include anti-CTLA-4 (e.g., ipilimumab), anti-PD-1 (e.g., nivolumab, pembrolizumab), anti-PDL-1 (e.g., atezolizumab, avelumab, durvalumab), anti-LAG-3, anti-TIM 3, anti-B7-H3, anti-ICOS, IDO inhibitors, 4-1 BB, anti-CD47, anti-B7-H4, anti-OX-40, anti-TIGIT, and anti-CD160. Nucleic acid sequences suitable for expressing these checkpoint inhibitors are well known.
- the present technology includes the following novel features. (1) Utilization of the TNF pathway for mediating cytotoxicity in lymphocyte-induced killing of cancer cells. (2) Active targeting of tumor antigens by transducing immune cells with a CAR combined with a membrane-bound TNF ligand. (3) Combination with checkpoint inhibitor therapy to enhance killing efficacy. (4) A versatile therapy design that can be easily repurposed and produced for a range of different target cancer cells.
- Targeted immune cells for TNF-mediated tumor cell killing increases the efficiency of immune cells compared to previous designs. This means cancers can be treated in some cases with fewer regimens of therapy.
- TNF mediated apoptosis will help treat cancers that are resistant to the immune pathways used by other therapies.
- Membrane-bound TNF will not interact with cells that the lymphocyte is not directly in contact with. The treatment is expected to cause reduced damage to healthy tissues, and thus reduced side effects.
- Uses of the present technology include as a treatment for cancers with targetable antigens, such as CD19+ lymphomas.
- This technology also has the capability to target other cells of interest, such as autoreactive immune cells and cells with viral or bacterial infections. Therefore, in addition to treatment of cancer, the technology can be used for treatment of autoimmune and infectious diseases.
- Example 1 Methods for Imaging Cell Interactions in Droplets, Cell Sorting, and Collection.
- a Fluorescence Detection and Droplet Sorting was used.
- the system used a microfluidic device (Fig. 1A) with a high-throughput on-chip droplet docking array capable of analyzing 10,000 events.
- the docking array contained U-shaped microwells for on-demand trapping, extraction, and incubation of droplets on a single microfluidic chip (Fig. 1 B).
- the integrated FADS system allowed the following. (1) Rapid enrichment of droplets containing user-defined ratios of T-tumor cell pairs, increasing the efficiency of heterotypic co-encapsulation events.
- the FADS control system consisted of three main elements: excitation, detection, and sorting (Fig. 2A).
- the excitation was provided by three different laser lines (Opto Engine, 405nm, 488nm, and 561 nm 100mW laser lines). These allowed for blue (DAPI), green (FITC) and red (dsRed) fluorescence detection) combined in a laser combiner (Doric Lenses), collimated, and connected to the epi-fluorescence port of a standard fluorescence microscope. This system illuminated the sample (a microfluidic droplet in the sorter device) through the microscope objectives.
- the detection systems included a series of beam-splitters, dichroic mirrors (Semrock), and band-pass emission filters that selectively directed the emission wavelength to three dedicated photomultiplier detectors (PMTs, Hamamatsu). Simultaneously, trans-illumination was available from the trans-illumination port of the microscope, with the addition of an appropriate filter to filter out emission wavelengths from the illumination.
- the PMTs were wired to an I/O board (National Instruments) with a LabView- based program that monitored the PMT outputs and sent sort pulses when the detected signals were above a user-defined threshold.
- the pulse originated by the I/O board was amplified by a high-voltage amplifier (Trek Labs.) and connected to the microfluidic device electrodes.
- the droplet sorting segment of the microfluidic device included a straight channel where spaced droplets traveled single-file at 430 ul/hr, a detection region where the illumination-detection system is focused, and a sorting fork flanked by electrode channels filled with metal electrodes. Droplets were automatically sorted at the outlet based on detection of up to 3 distinct fluorescent markers at 170 droplets/min.
- Excitation Lasers Up to 3 solid-state diode lasers (Opto Engine) were focused in slits across the microfluidic channel corresponding to the excitation wavelengths of the three fluorophores (405nm, 488nm, 561 nm). In combination with the fluorescence detection filters, these were selected to be well spectrally separated to allow multiplexed detection with minimal cross-talk between channels. Beams were expanded with cylindrical lenses and then focused to a slit though a 40X microscope objective at different positions along the channel.
- PMTs Photomultiplier Tubes
- a set of 3 PMTs was used to detect the fluorescence signal from droplets. Emitted fluorescence was collected with a second microscope objective, split into 3 detection channels with 50-50 beam-splitters, and then filtered with appropriate band-pass filters corresponding to each fluorophore (centered at: 405nm, 488nm, 561 nm). The output of each PMT was pre-amplified and then acquired with a multi-function data acquisition board (National Instruments) connected to a personal computer. Software running on the PC allowed real-time detection of fluorescent spikes on each of the 3 detector channels.
- Sorting Junction After passing through the laser slits, droplets were diverted into collection outlets using dielectrophoresis. A microelectrode was deposited next to the channel at the sorting junction during fabrication and connected to a high voltage, programmable power supply (Stanford Research Systems). Software written in LabVIEW controlled the magnitude and frequency of the AC voltage applied across the electrodes based on droplet fluorescent signature, thereby controlling the magnitude of deflection and outlet channel. The amplitude of the AC potential was selected to be as small as possible to minimize the possibility of shearing the droplets.
- a white-light source and monochrome CCD camera allowed alignment of the microfluidic chip on the optical system, and alignment of orthogonal illumination of the flow channel with the 3 laser slits and detection optics.
- a device To sort droplets based on the cell-cell (or cell-drug) interactions, a device was developed that integrates the device with an array of U-shaped microwells.
- the U-shaped microwells enable on-demand trapping, extraction, incubation, and merging of droplets. This equips the device with the capability to sort droplets depending on the real-time monitored phenotype changes of the cells upon interactions.
- the cell- encapsulated droplets were first sorted and collected, and then the collected droplets were reinjected into the microwells. This results in the enrichment of cell-encapsulated droplets trapped and incubated in the microwells.
- a fluorescence detection technique was employed for monitoring the real-time interactions of the cells inside the droplets during an incubation period.
- the controlled and targeted delivery of cells or drugs was achieved through droplet merging in the microwells, and the extraction of droplets from the microwells was achieved by simply reversing the flow direction.
- the extracted droplets were then sorted by flowing the droplets through the sorting junction.
- the phase or fluorescence images of cells in droplets was captured using a Zeiss Axio Observer.ZI Microscope (Zeiss, Germany) equipped with a Hamamatsu digital camera C10600 Orca-R2, 10-40x objectives, and standard FITC/DAPI/TRITC/FR filters.
- the microfluidic device containing cells co-encapsulated in droplets was maintained in a humidified microscopic stage-top incubator at 37°C and 5% CO 2 for the duration of the experiment. All time-lapse images were obtained by automated software control, and the Zen imaging program (Zeiss) was set to travel to specific x-, y-, and z-positions at every 5 minutes for a total period of 6-8 hours for video microscopy.
- images were collected at 0 h to determine the initial occupancy of each droplet and every 5 min for 4-12h. Image processing and analysis was done with Imaged.
- E effector T cells
- T target cells
- Basal/inactive T cells were defined as those that did not kill target cells or only killed after prolonged coencapsulation.
- Active T cells killed one target rapidly, while “hyperactive” T cells (also referred to herein as “serial killers”) killed multiple targets sequentially. See Figs. 2B, 2C). Since cell loading in droplets is characterized by a Poisson distribution, the large-scale droplet array typically includes multiple E:T ratios in the same experimental run.
- CD8+ T cells typically cause the majority of target cell deaths, killing target cells serially, while the rest of immune cells were less cytotoxic.
- E:T ratios 1 :2, 1 :5, and 1 : 10 in 96-well plates to permit one effector cell to interact with multiple targets sequentially.
- the T cells were labeled with blue CellTracker CMAC Dye (ThermoFisher), while the target tumor cells (e.g., K562, HL-60, or Raji cells) were labeled with green Calcein AM dye. Then the cells were injected into the chip to generate droplets at a throughput of 200 droplets/sec. The two cell types were introduced through separate inlets to prevent cell interaction prior to co-encapsulation in droplets. The resulting droplets contained various numbers and types of cells. The mixed droplet population was flowed through the sorting system to enrich in droplets that contained one T cell labeled blue and one tumor cell labeled green for loading into the droplet array. Other droplets were considered waste material and removed.
- Blue CellTracker CMAC Dye ThermoFisher
- the target tumor cells e.g., K562, HL-60, or Raji cells
- green Calcein AM dye green Calcein AM dye
- Sorting after imaging in the array When the docking array was fully occupied by blue + green droplets, droplet generation was stopped and acquisition of dual-color fluorescence images of the array was begun. After incubating cell pairs in the array for4-16hrs, the droplets were released, and the sorting gate was set on green + blue channel positive, which selected the droplets containing both blue (T cells) and green (tumor cells).
- the time-lapse microscopybased approach allowed profiling of dynamic interaction histories at the single-cell level by quantifying the following parameters: (i) viability of each cell type; (ii) immune-complex formation; (iii) duration and frequency of contact; (iv) rate of association/dissociation; (v) change in motility; and (vi) timing of immune-mediated target cell lysis.
- viability of each cell type i.e., a cell type
- iii) immune-complex formation iii) duration and frequency of contact
- iv rate of association/dissociation
- change in motility and (vi) timing of immune-mediated target cell lysis.
- T cell did not kill the target cell, which retained its green fluorescence, the T cell was considered inactive.
- the droplets that did not undergo positive selection i.e.
- the droplets separated by negative selection at the second sorting gate consisted of two populations: (a) only blue cells, when the T (blue) cell has killed the tumor cell (i.e., loss of green), suggesting that there are hyperactive T cells in that population; and (b) only green cells, where the T (blue) cell has died. A minimal death rate of ⁇ 2% was observed during the duration of the experiments.
- Fluorescence-based droplet sorting of CD8+ T Cells to screen TCR binding affinity Since the amount of calcium released in a T cell is proportional to the number of T cell receptors (TCRs) bound, a fluorescence threshold was established to indicate T cells with highest antigen affinity. Upon TCR binding to antigen, calcium is released inside the T cell as part of the secondary messenger cascade. This produces a strong fluorescence signal, allowing sorting of antigen-bound T cells in droplets (Fig. 3A). A higher measured calcium response is expected to be associated with a stronger cytotoxic response when T cells are introduced to their corresponding antigen, such as an antigen present on cancer cells.
- a cancer cell antigen such as NY-ESO-1 peptides
- MHC tetramers can be conjugated with MHC tetramers, following established methods.
- Antigen-induced CD8+T cells also can be prepared as described in the literature, and mixed with antigen-conjugated MHC tetramers on-chip, and given 10 seconds to mix prior to running through the sorting array (Fig. 3B).
- An intracellular calcium stain such as Fura-10 AM (AAT Bioquest) can be used to identify the calcium spike following TCR binding to the antigen conjugate.
- the resulting fluorescent signal produced by the calcium release (Figs.
- 3C, 3D activates the sorter and leads to isolation of the activated CD8+ T cell.
- concentration of MHC tetramer can be adjusted, and the threshold of fluorescence signal for sorting also can be adjusted. This can ensure exclusive isolation of T cells with a strong response to the antigen.
- Microfluidic Device Fabrication Devices were fabricated using standard soft lithography techniques with PDMS (Fig. 4). Silicon wafers were spin coated with SU-8 2075, and UV crosslinked in two stages, one to make the perfusion channels and one to make the device design. Channel heights were made to approximately 120 pm, with perfusion channels an additional 40 pm. Devices were made with 10:1 PDMS and curing agent and bonded to glass slides with a Harrick Plasma Cleaner (Harrick Plasma Inc, Ithaca, NY). Devices were made hydrophobic by perfusing them with AQUAPEL (PPG Industries, Inc, Pittsburgh, PE) through the channels and allowing them to dry.
- AQUAPEL PPG Industries, Inc, Pittsburgh, PE
- DLBCL Diffuse large B-cells
- RPMI-1640 ATCC, Manassas, VA
- FBS FBS
- 1% antibotic/antimycotic mixture Gibco, Waltham, MA
- Peripheral blood NK cells were purchased from Stemcell Technologies (Vancouver, BC, Canada). NK cells were thawed the day prior to experiments and rested overnight in RPMI 1640.
- Alginate stocks were prepared at 4% in distilled water and filtered with a 0.22 pm filter. Vitrogel-RGD High Concentration was purchased from TheWell Biosciences (North Brunswick, NJ). Alginate solution was either mixed with RGD Vitrogel or added alone to glass vials with a stir bar. Cells were pelleted and resuspended in HBSS, then added to alginate while continuously stirring. Alginate was made to 1 % w/v final concentration, either without or supplemented with 7.5% v/v RGD+ Vitrogel.
- Viability imaging and Cell Preparation an Axio Observer microscope equipped with an automated stage and incubation chamber was used while maintaining cells at 37 °C and 5% CO2. The microscope was also equipped with a light source and filter set for fluorescence of DAPI, EGFP, dsRed and far-red channels.
- DLBCL were incubated with 10 pM CMAC CellTracker (ThermoFisher) in serum-free media for 45 minutes and washed twice with media prior to pelleting and resuspending in hydrogel.
- NK cells were incubated with 2 pM CFSE CellTracker (Thermofisher) and resuspended with DLBCL for coculture conditions.
- NK cells were loaded with DLBCL at a 1 :2 ratio (NK:DLBCL). NK cells were loaded at 5 to 7.5 million cells/mL, while DLBCL were loaded at 10 to 15 million cells/mL. Prior to viability imaging, devices were perfused with 8 pM ethidium homodimer (Biotium, Freemont, CA) for approximately 3 hours. Spheroids were differentiated between low density and high density, with high density having >5 cells/mm 2 .
- Immunofluorescence Imaging For immunofluorescence imaging, a Zeiss LSM 880 confocal microscope was used (Zeiss). The microscope was equipped with 405, 458, 488, 514, 561 , 594 and 640 nm excitation lasers, Airyscan superresolution, and spectral unmixing capabilities. For imaging, cells were not labeled with dyes prior to loading spheroids. For DLBCL, the antibodies used were anti-CD47 conjugated to PerCP-Cy 5.5, anti-PDL1 conjugated to Alexa Fluor 594, and anti CD20 conjugated to Brilliant Violet 421 (Biolegend).
- NK cells For NK cells, anti-SIRPa conjugated to FITC, anti-PD1 conjugated to Alexa Fluor 647, and anti-CD16 conjugated to PE were used for labeling (Biolegend). Prior to imaging, devices were infused with antibodies in serum-free RPMI- 1640 at 50 pL/h for 2 hours. After antibody labeling, cells were fixed by perfusion with BD fixation buffer for 1 hour, then washed with HBSS via perfusion for 1 hour. Images were taken at 20x magnification. Unlabeled spheroids loaded with an identical cell concentrations were used as controls for autofluorescence.
- DLBCL take a more resistant phenotype when under stress.
- the condition with DLBCL and NK displayed the highest levels of granzyme secretions, which were reduced with the addition of treatments.
- DLBCL + NK + rituximab showed high expression of numerous alternative factors associated with immune cell activation and inflammation. This result is reasonable, as rituximab binding activates the ADCC pathway in NK cells, which should produce a highly active and cytotoxic phenotype.
- Several of these secretions however, also have been shown to promote immune cell evasion in late stage cancers, and can lead to immune cell desensitization or exhaustion.
- Gal-1 and CD83 are found at increased levels. Gal-1 has been correlated to increased metastasis and immune evasion, while CD83 is known to decrease NK cell activation.
- ribosomal proteins Across ribosomal proteins, an average increase in expression was found in rCHOP, except mitochondrial ribosomal proteins, which were found to be slightly higher in rituximab alone treatment. While many biological factors can contribute to ribosomal changes, mitochondrial ribosome gene expression has been linked to resistance in cancer cells. Ribosomal RNA expression has been shown to have a range of consequences in cancer and other diseases, correlating both to cancer resistance and NK cells activity. In addition to ribosome upregulation, protein trafficking and enzymatic activity, such as NADPH and oxidoreductase expression, were found upregulated in rCHOP treated cells. Overall, this insinuates that metabolic activity in the rCHOP treated cells may be elevated, potentially through DLBCL resistance mechanisms to CHOP treatment.
- FABPs fatty acid binding proteins
- PPARs Peroxisome Activator Receptors
- TNFSF10 TNFSF10
- RNKL TNFSF1 1
- TWEAK TNFSF12
- TRAIL is a well known ligand expressed on NK cells and other immune cells that can directly induce apoptosis in target cells.
- transcriptomic data When comparing the secretomic data (Fig. 5) to the transcriptomic data (Figs. 6A-6F), there appears to be an increase in cellular activity.
- the transcriptomic data reveals a more active NK phenotype when treated with rituximab alone, while the transcriptomic data shows increased metabolic activity with rCHOP. Given these results, it seems that the increased metabolic activity can be primarily attributed to the DLBCL response to CHOP. These metabolic changes may sensitize the DLBCL to NK cell ADCC mediated cytotoxicity, without the need of a high inflammatory response.
- rCHOP has been shown to have much higher rates of patients with complete responses than patients with rituximab alone.
- RNA-seq of F-FADS sorted BiTE activated CD8+ T-cell killers and non-killers from 4 donor samples was performed.
- a total of 15389 ⁇ 1492 genes for CD8+ killers and 13209 ⁇ 2632 genes for CD8+ non-killers were identified in these samples, with a threshold of fragments per kilobase of exon model per million reads mapped (FPKM) > 0.5.
- Principal component analysis was used to emphasize variation as well as similarity in the dataset.
- BiTE activated CD8+ killers and non-killers A global representation of BiTE activated CD8+ killers and non-killers is shown in the PCA plots (not shown).
- BiTE activated CD8+ T-cell killers and non-killers from all the donors did not form a distinct cluster, suggestive of unique molecular heterogeneity within each donor sample.
- the number of genes uniquely expressed by killers vs. non-killers was evaluated, in addition to the number of genes that were co-expressed (Figs. 7A, 7B-1 , 7B-2, 7C).
- DEG differentially expressed gene
- Cytotoxic T- cells mediate lysis of target cells by exocytosis of specialized cytoplasmic granules containing cytotoxins perforin (PRF1), granzyme B (GZMB) and granulysin (GNLY), and receptor-ligand binding of Fas ligand (FASL) and TNF-related apoptosis-inducing ligand (TRAIL) molecules (ref: www.ncbi.nlm.nih.gov/books/NBK27101/).
- PRF1 cytotoxins perforin
- GZMB granzyme B
- GNLY granulysin
- FASL Fas ligand
- TRAIL TNF-related apoptosis-inducing ligand
- GZMK granzyme
- GZMH granzyme
- GZMM granzyme
- GZMA granzyme-dependent form of cell death
- BiTE activated CD8+ T-cell killers from some donors had low GZMB expression levels and no difference was observed between killers and non-killers. These same donors, however, had statistically significant upregulation in other granzymes (GZMH, GZMA), FASL and TNFSF10. Other donors (specifically donors 2 and 4) had statistically significant upregulation of GZMB in BiTE activated CD8+ T-cell killers compared to non-killers, while there was no upregulation in FASL and TNSF10.
- Immune checkpoint molecules are inhibitory receptors on the surface of immune cells that ensure appropriate regulation of the immune response.
- Prominent checkpoint molecules include programmed cell death receptor 1 (PD-1) and CTLA-4 expressed by T cells and other immune cells.
- Other checkpoint molecules include lymphocyte-activation gene 3 (LAG-3), T- cell immunoglobulin and mucin domain-3 (TIM-3), T-cell immunoreceptor with Ig and ITIM domains (TIGIT) and inducible T-cell co-stimulatory receptor (ICOS).
- LAG-3 lymphocyte-activation gene 3
- TIM-3 T- cell immunoglobulin and mucin domain-3
- T-cell immunoreceptor with Ig and ITIM domains T-cell immunoreceptor with Ig and ITIM domains
- IAGIT T-cell immunoreceptor with Ig and ITIM domains
- ICOS inducible T-cell co-stimulatory receptor
- CD244 and TIGIT were upregulated in 3 of the 4 donors.
- CD244 expression has been shown to be a mediator of CD8+ T-cell exhaustion in the setting of persistent antigen exposure, but it has also been shown to be co-expressed on a subset of antigen-experienced effector and effector memory CD8+ T-cells along with other immunoregulatory receptors such as PD-1 but not TIGIT.
- CD244 has also been shown to be a prominent regulator involved in T cell aging.
- T-cell exhaustion a panel of cytokine and chemokine genes was examined. Exhaustion of T-cells is typically accompanied by loss of effector cytokines such as interferon-y (IFNG), tumor necrosis (TNF) and interleukin-2 (IL2) as well as chemokine receptors CCL2 and CCL5. Increases were observed after CD3/CD19 bispecific antibody treatment of CD8+ T cell killers in IL2, IL7R, IL12RB, IL15, IL18R1 , IL18BP, IFNG, and TNF. but not in all donors.
- IFNG interferon-y
- TNF tumor necrosis
- IL2 interleukin-2
- Increases were observed after CD3/CD19 bispecific antibody treatment of CD8+ T cell killers in IL2, IL7R, IL12RB, IL15, IL18R1 , IL18BP, IFNG, and TNF. but not in all donors.
- Donors 2 and 4 appear to have an overall dampened cytokine response compared to donors 1 and 3.
- BiTE activated CD8+ T-cell killers from donor 2 and 4 also had increased GZMB, EOMES, CTLA4 and PDCD1 compared to the other donors. Since GZMB is typically lost in advanced stages of exhaustion, these results may reflect an early exhaustion phenotype or a highly activated state.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Biotechnology (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Microbiology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Hematology (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Cell Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Urology & Nephrology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Food Science & Technology (AREA)
- Analytical Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Physics & Mathematics (AREA)
- Epidemiology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Toxicology (AREA)
- Mycology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
La présente technologie propose une cellule immunitaire génétiquement modifiée exprimant un récepteur antigénique chimérique (CAR) et exprimant également un niveau élevé d'un ligand pour un récepteur du facteur de nécrose tumorale (TNF). La cellule génétiquement modifiée tire profit de voies biologiques doubles pour tuer des cellules cancéreuses, des pathogènes cellulaires et des cellules infectées par des virus. La voie médiée par CAR favorise l'action destructrice par la perforine et les granzymes libérés par des lymphocytes T cytotoxiques et des cellules tueuses naturelles, ainsi que la cellule cible médiée par le récepteur du TNF (c'est-à-dire, le domaine de la mort) par ces mêmes cellules. En outre, la présente technologie, en particulier lorsqu'elle est combinée à une analyse d'image de cellule unique, à un tri de cellules basé sur la microfluidique et à une analyse transcriptomique permet d'adapter des modifications génétiques aux cellules d'un patient, en réponse à un patient significatif de la variation du patient dans l'expression génique de lymphocytes T et de cellules NK.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363470820P | 2023-06-02 | 2023-06-02 | |
| US63/470,820 | 2023-06-02 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024250037A1 true WO2024250037A1 (fr) | 2024-12-05 |
Family
ID=93658569
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/032321 Pending WO2024250037A1 (fr) | 2023-06-02 | 2024-06-03 | Thérapie cellulaire immunitaire ciblée à l'aide de trail et d'apoptose médiée par le tnf |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024250037A1 (fr) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20170369550A1 (en) * | 2014-12-24 | 2017-12-28 | Ucl Business Plc | Cell |
| US20200400669A1 (en) * | 2016-01-15 | 2020-12-24 | Berkeley Lights, Inc. | Methods of Producing Patient-Specific Anti-Cancer Therapeutics and Methods of Treatment Therefor |
| US20220212192A1 (en) * | 2019-06-01 | 2022-07-07 | Northeastern University | Microfluidic Device for High-Throughput Screening of Tumor Cell Adhesion and Motility |
| US20220281950A1 (en) * | 2021-03-04 | 2022-09-08 | Allogene Therapeutics, Inc. | Fasl expression and fasr gene knockout to protect therapeutic cells from allogeneic rejection and activation-induced cell death |
-
2024
- 2024-06-03 WO PCT/US2024/032321 patent/WO2024250037A1/fr active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20170369550A1 (en) * | 2014-12-24 | 2017-12-28 | Ucl Business Plc | Cell |
| US20200400669A1 (en) * | 2016-01-15 | 2020-12-24 | Berkeley Lights, Inc. | Methods of Producing Patient-Specific Anti-Cancer Therapeutics and Methods of Treatment Therefor |
| US20220212192A1 (en) * | 2019-06-01 | 2022-07-07 | Northeastern University | Microfluidic Device for High-Throughput Screening of Tumor Cell Adhesion and Motility |
| US20220281950A1 (en) * | 2021-03-04 | 2022-09-08 | Allogene Therapeutics, Inc. | Fasl expression and fasr gene knockout to protect therapeutic cells from allogeneic rejection and activation-induced cell death |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20250197471A1 (en) | Methods for the treatment of b cell malignancies using adoptive cell therapy | |
| Li et al. | KIR-based inhibitory CARs overcome CAR-NK cell trogocytosis-mediated fratricide and tumor escape | |
| US12247982B2 (en) | Markers selectively deregulated in tumor-infiltrating regulatory T cells | |
| Feucht et al. | T-cell responses against CD19+ pediatric acute lymphoblastic leukemia mediated by bispecific T-cell engager (BiTE) are regulated contrarily by PD-L1 and CD80/CD86 on leukemic blasts | |
| Freeman et al. | A conserved intratumoral regulatory T cell signature identifies 4-1BB as a pan-cancer target | |
| Sharma et al. | Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells | |
| US10738278B2 (en) | Engineered cells for adoptive cell therapy | |
| Roussel et al. | Functional characterization of PD1+ TIM3+ tumor-infiltrating T cells in DLBCL and effects of PD1 or TIM3 blockade | |
| Parry et al. | NK cell function is markedly impaired in patients with chronic lymphocytic leukaemia but is preserved in patients with small lymphocytic lymphoma | |
| US20210190763A1 (en) | Quality of immunological synapse predicts effectiveness of chimeric antigen receptor (car) t cells | |
| JP2020516248A (ja) | 前立腺特異的膜抗原(psma)またはその改変型を発現する操作された細胞および関連方法 | |
| BR112020011223A2 (pt) | marcadores fenotípicos para terapia celular e mé-todos relacionados | |
| Li et al. | Single‐cell transcriptome profiling reveals the key role of ZNF683 in natural killer cell exhaustion in multiple myeloma | |
| Navabi et al. | Natural killer cell functional activity after 4-1BB costimulation | |
| Shapiro et al. | First-in-human evaluation of memory-like NK cells with an IL-15 super-agonist and CTLA-4 blockade in advanced head and neck cancer | |
| Zhu et al. | CAR-NK cells for gastrointestinal cancer immunotherapy: from bench to bedside | |
| Sullivan et al. | An innovative single-cell approach for phenotyping and functional genotyping of CAR NK cells | |
| CN108778312A (zh) | 使用基于细胞的疗法治疗癌症和传染病的方法 | |
| WO2024250037A1 (fr) | Thérapie cellulaire immunitaire ciblée à l'aide de trail et d'apoptose médiée par le tnf | |
| WO2020233503A1 (fr) | Procédé de détection | |
| Zhu et al. | CD2 augmentation enhances CAR-T-cell efficacy via immunological synapse remodeling and T-cell exhaustion mitigation | |
| Sullivan | Development of integrated platform of lymphocyte phenotyping, and immunotherapy validation in single cell and 3D droplet microfluidic systems | |
| RU2346279C2 (ru) | СПОСОБ ОПРЕДЕЛЕНИЯ УРОВНЯ мРНК КАТАЛИТИЧЕСКОЙ СУБЪЕДИНИЦЫ ТЕЛОМЕРАЗЫ (hTERT) | |
| Fan et al. | Bispecific killer cell engager-secreting CAR-T cells redirect natural killer specificity to enhance antitumour responses | |
| Rodin | Unconventional T cells in colon adenocarcinomas |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24816687 Country of ref document: EP Kind code of ref document: A1 |
|
| DPE1 | Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101) |