WO2024102777A2 - Compositions and method for expansion of embryonic stem cells - Google Patents

Compositions and method for expansion of embryonic stem cells Download PDF

Info

Publication number
WO2024102777A2
WO2024102777A2 PCT/US2023/079019 US2023079019W WO2024102777A2 WO 2024102777 A2 WO2024102777 A2 WO 2024102777A2 US 2023079019 W US2023079019 W US 2023079019W WO 2024102777 A2 WO2024102777 A2 WO 2024102777A2
Authority
WO
WIPO (PCT)
Prior art keywords
days
cells
mdcs
cell
cancer
Prior art date
Application number
PCT/US2023/079019
Other languages
French (fr)
Inventor
Dana Hayoun NEEMAN
Rami Skaliter
Original Assignee
Lineage Cell Therapeutics, Inc.
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by Lineage Cell Therapeutics, Inc. filed Critical Lineage Cell Therapeutics, Inc.
Publication of WO2024102777A2 publication Critical patent/WO2024102777A2/en

Links

Definitions

  • the method includes: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce a cultured suspendable expansion complex comprising cultured cells; (c) removing the cultured cells from the cultured suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs); and (e) contacting the population of iDCs with a maturation cocktail for a third period of time to differentiate the population of iDCs into a population of mature dendritic cells (mDCs).
  • ECM extracellular matrix
  • mDCs maturation cocktail
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier.
  • a method of manufacturing a cell vaccine for treating cancer including: (a) obtaining one or more mature dendritic cells (mDCs) according to the method provided herein including embodiments thereof; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer.
  • mDCs mature dendritic cells
  • a method for treating or delaying progression of a disease or disorder in a subject including administering to the subject a cell vaccine provided herein including embodiments thereof.
  • FIG.2 shows the H1 Master Cell Bank (MCB) production process.
  • FIG.3 shows the H1 Working Cell Bank (WCB) production process.
  • FIG.4 shows the LCT-VAC2 and LCT-mDCs production process.
  • FIG.5 shows results from the H1 post irradiation performance test.
  • FIG.6 shows the detailed IPC plan specific for monitoring LCT-mDCs differentiation process.
  • FIGS.7A-7B show results from a migration assay. The assay test migratory activity of shows a schematic of the transwell cell migration assay. FIG.7B shows the quantified migratory activity of LCT-mDCs compared to PBMC-mDCs.
  • FIGS.8A-8C show results from a mixed lymphocyte reaction (MLR) assay. The MLR assay tests ton-specific activation of PMBCs by LCT-mDCs by measuring gated CD4+ T cell proliferation using fluorescent dye labeling (CFSE) after 6 days of co-culture.
  • MLR mixed lymphocyte reaction
  • FIGS.9A-9B show a schematic of (FIG.9A) and results (FIG.9B) from a PC antigen presentation assay.
  • the PC antigen presentation assay tests mDCs ability to successfully process PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • FIGS.10A-10B show a schematic of (FIG.10) and results (FIG.10B) from an hTERT antigen presentation assay.
  • the hTERT antigen presentation assay tests mDCs ability to successfully process and present hTERT antigen (by peptide or mRNA EP) to antigen specific T DETAILED DESCRIPTION
  • Nucleic acid refers to nucleotides (e.g., deoxyribonucleotides or ribonucleotides) and polymers thereof in either single-, double- or multiple-stranded form, or complements thereof; or nucleosides (e.g., deoxyribonucleosides or ribonucleosides). In embodiments, “nucleic acid” does not include nucleosides.
  • nucleoside refers, in the usual and customary sense, to a glycosylamine including a nucleobase and a five- carbon sugar (ribose or deoxyribose).
  • nucleosides include, cytidine, uridine, adenosine, guanosine, thymidine and inosine.
  • nucleotide refers, in the usual and customary sense, to a single unit of a polynucleotide, i.e., a monomer. Nucleotides can be ribonucleotides, deoxyribonucleotides, or modified versions thereof. Examples of polynucleotides contemplated herein include single and double stranded DNA, single and double stranded RNA, and hybrid molecules having mixtures of single and double stranded DNA and RNA. Examples of nucleic acid, e.g. polynucleotides contemplated herein include any types of RNA, e.g.
  • RNA means the segment of DNA involved in producing a protein; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
  • the leader, the trailer as well as the introns include regulatory elements that are necessary during the transcription and the translation of a gene.
  • a "protein gene product” is a protein expressed from a particular gene.
  • plasmid refers to a nucleic acid molecule that encodes for genes and/or regulatory elements necessary for the expression of genes. Expression of a gene from a plasmid can occur in cis or in trans. If a gene is expressed in cis, the gene and the regulatory elements are encoded by the same plasmid. Expression in trans refers to the instance where the gene and the regulatory elements are encoded by separate plasmids. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • transfection can be used interchangeably and are defined as a process of introducing a nucleic acid molecule or a protein to a cell.
  • Nucleic acids are introduced to a cell using non-viral or viral-based methods.
  • the nucleic acid molecules may be gene sequences encoding complete proteins or functional portions thereof.
  • Non-viral methods of transfection include any appropriate transfection method that does not use viral DNA or viral particles as a delivery system to introduce the nucleic acid molecule into the cell.
  • Exemplary non-viral transfection methods include calcium phosphate transfection, liposomal transfection, nucleofection, sonoporation, transfection through heat shock, magnetifection and electroporation.
  • the nucleic acid molecules are introduced into a cell using electroporation following standard procedures well known in the art.
  • any useful viral vector may be used in the methods described herein. Examples for viral vectors include, but are not limited to retroviral, adenoviral, lentiviral and adeno-associated viral vectors.
  • the nucleic acid molecules are introduced into a cell using a Typically, transduction or transfection of a protein relies on attachment of a peptide or protein capable of crossing the cell membrane to the protein of interest. See, e.g., Ford et al. (2001) Gene Therapy 8:1-4 and Prochiantz (2007) Nat. Methods 4:119-20.
  • Contacting is used in accordance with its plain ordinary meaning and refers to the process of allowing at least two distinct species (e.g. cells and an agent) to become sufficiently proximal to react, interact, or physically touch.
  • the term “cell culture,” as used herein, refers to a plurality of cells grown in vitro over time.
  • the cell culture my originate from a plurality of human pluripotent stem (hPS) cells or from a single hPS cell and may include all of the progeny of the originating cell or cells, regardless of 1) the number of passages or divisions the cell culture undergoes over the lifetime of the culture; and PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCTX-CC-600PCT 2 any changes in phenotype to one or more cells within the culture over the lifetime of the culture (e.g. resulting from differentiation of one or more hPS cells in the culture).
  • a cell culture begins with the initial seeding of one or more suitable vessels with at least one hPS cell and ends when the last surviving progeny of the original founder(s) is harvested or dies. Seeding of one or more additional culture vessels with progeny of the original founder cells is also considered to be a part of the original cell culture.
  • dendritic cells or “DCs,” as used herein, refers to professional antigen- presenting cells (also known as accessory cells) of the mammalian immune system.
  • the main function of DCs is to process antigen material and present it on the cell surface to the T cells of the immune system.
  • DCs act as messengers between the innate and the adaptive immune systems.
  • isolated when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It can be, for example, in a homogeneous state and may be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
  • exogenous refers to a molecule or substance (e.g., a compound, nucleic acid or protein) that originates from outside a given cell or organism.
  • an "exogenous cytokine” as referred to herein is a cytokine that does not originate from the cell or cell population.
  • endogenous refers to a molecule or substance that is native to, or originates within, a given cell or organism.
  • the term “inhibition”, “inhibit”, “inhibiting” and the like in reference to cell proliferation means negatively affecting (e.g., decreasing proliferation) or killing the cell.
  • inhibition refers to reduction of a disease or symptoms of disease (e.g., cancer, cancer cell proliferation).
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0035]
  • expression includes any step involved in the production of the polypeptide including, but not limited to, transcription, post-transcriptional modification, translation, post- translational modification, and secretion. Expression can be detected using conventional techniques for detecting protein (e.g., ELISA, Western blotting, flow cytometry, immunofluorescence, immunohistochemistry, etc.).
  • a “control” or “standard control” refers to a sample, measurement, or value that serves as a reference, usually a known reference, for comparison to a test sample, measurement, or value.
  • a test sample can be taken from a patient suspected of having a given disease (e.g. cancer) and compared to a known normal (non-diseased) individual (e.g. a standard control subject).
  • a standard control can also represent an average measurement or value gathered from a population of similar individuals (e.g. standard control subjects) that do not have a given disease (i.e. standard control population), e.g., healthy individuals with a similar medical background, same age, weight, etc.
  • a standard control value can also be obtained from the same individual, e.g. from an earlier-obtained sample from the patient prior to disease onset.
  • a control can be devised to compare therapeutic benefit based on pharmacological data (e.g., half- life) or therapeutic measures (e.g., comparison of side effects). Controls are also valuable for determining the significance of data. For example, if values for a given parameter are widely variant in controls, variation in test samples will not be considered as significant.
  • standard controls can be designed for assessment of any number of parameters (e.g. RNA levels, protein levels, specific cell types, specific bodily fluids, specific tissues, etc).
  • Standard controls are also valuable for determining the significance (e.g. statistical significance) of data. For example, if values for a given parameter are widely variant in standard controls, variation in test samples will not be considered as significant.
  • “Patient” or “subject in need thereof” refers to a living organism suffering from or prone to a disease or condition that can be treated by administration of a composition or pharmaceutical composition as provided herein. Non-limiting examples include humans, other mammals, PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • a patient is human.
  • the terms “disease” or “condition” refer to a state of being or health status of a patient or subject capable of being treated with the compositions or methods provided herein.
  • the disease may be a cancer.
  • the cancer may refer to a solid tumor malignancy. Solid tumor malignancies include malignant tumors that may be devoid of fluids or cysts.
  • the solid tumor malignancy may include breast cancer, ovarian cancer, pancreatic cancer, cervical cancer, gastric cancer, renal cancer, head and neck cancer, bone cancer, skin cancer or prostate cancer.
  • cancer refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, including solid and lymphoid cancers, kidney, breast, lung, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, glioma, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including B-acute lymphoblastic lymphoma, non-Hodgkin’s lymphomas (e.g., Burkitt’s, Small Cell, and Large Cell lymphomas), Hodgkin’s lymphoma, leukemia (including acute myeloid leukemia (AML), ALL, and CML), or multiple myeloid leukemia (AML
  • cancer refers to all types of cancer, neoplasm or malignant tumors found in mammals (e.g., humans), including leukemia, carcinomas and sarcomas.
  • exemplary cancers that may be treated with a composition or method provided herein include breast cancer, colon cancer, kidney cancer, leukemia, lung cancer, melanoma, ovarian cancer,
  • associated or “associated with” in the context of a substance or substance activity or function associated with a disease (e.g. a protein associated disease, a cancer (e.g., breast cancer, lung cancer)) means that the disease (e.g.
  • a "therapeutic agent” as referred to herein is a composition useful in treating a disease such as cancer (e.g., leukemia).
  • the therpaeutic agent is an anti-cancer agent.
  • Anti-cancer agent is used in accordance with its plain ordinary meaning and refers to a PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCTX-CC-600PCT composition e.g. compound, drug, antagonist, inhibitor, modulator having antineoplastic properties or the ability to inhibit the growth or proliferation of cells.
  • an anti- cancer agent is a chemotherapeutic.
  • an anti-cancer agent is an agent identified herein having utility in methods of treating cancer.
  • an anti-cancer agent is an agent approved by the FDA or similar regulatory agency of a country other than the USA, for treating cancer.
  • This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder.
  • this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. It is understood that treatment, while intended to cure, ameliorate, or stabilize a disease, pathological condition, or disorder, need not actually result in the cure, amelioration, or stabilization.
  • the effects of treatment can be measured or assessed as described herein and as known in the art as is suitable for the disease, pathological condition, or disorder involved. Such measurements and assessments can be made in qualitative and/or quantitative terms. Thus, for example, characteristics or features of a disease, pathological condition, or disorder and/or symptoms of a disease, pathological condition, or disorder can be reduced to any effect or to any amount.
  • the term “in need of treatment” as used herein refers to a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals) that a subject requires or will benefit from treatment.
  • feeder cells refers to non-hPS cells that are co-cultured with hPS cells and provide support for the hPS cells. Support may include facilitating the growth and maintenance of the hPS cell culture by providing the hPS cell culture with one or more cell factors such that the hPS cells are maintained in a substantially undifferentiated state.
  • Feeder cells may either have a different genome than the hPS cells or the same genome as the hPS cells and may originate from a non-primate species, such as mouse, or may be of primate origin, e.g., human.
  • Examples of feeder cells may include cells having the phenotype of connective tissue such as murine fibroblast cells, human fibroblasts.
  • feeder-free refers to a condition where the referenced composition contains no added feeder cells. To clarify, the term feeder-free encompasses, inter alia, situations where primate pluripotent stem cells are passaged from a culture which may comprise some feeders into a culture without added feeders even if some of the feeders from the first culture are present in the second culture.
  • Serum free refers tissue culture growth conditions that have no added animal serum such fetal bovine serum, calf serum, horse serum, and no added commercially available serum replacement supplements such as B-27. Serum free includes, for example, media which may comprise human albumin, human transferrin and recombinant human insulin.
  • electroporation refers to a method for permeabilizing cell membranes by generating membrane pores with electrical stimulation.
  • the applications of electroporation include, but are not limited to, the delivery of DNA, RNA, siRNA, peptides, proteins, antibodies, drugs or other substances to a variety of cells such as mammalian cells, plant cells, yeasts, other eukaryotic cells, bacteria, other microorganisms, and cells from human patients.
  • dose refers to the amount of active ingredient given to an individual at each administration. The dose will vary depending on a number of factors, including the range of normal doses for a given therapy, PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • a dosage form refers to the particular format of the pharmaceutical or pharmaceutical composition, and depends on the route of administration.
  • a dosage form can be in a liquid form for nebulization, e.g., for inhalants, in a tablet or liquid, e.g., for oral delivery, or a saline solution, e.g., for injection.
  • a therapeutically effective dose or amount as used herein is meant a dose that produces effects for which it is administered (e.g. treating a disease).
  • the exact dose and formulation will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques.
  • a therapeutically effective amount will show an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100%.
  • Therapeutic efficacy can also be expressed as “- fold” increase or decrease.
  • a therapeutically effective amount can have at least a 1.2- fold, 1.5-fold, 2-fold, 5-fold, or more effect over a standard control.
  • a therapeutically effective dose or amount may ameliorate one or more symptoms of a disease.
  • a therapeutically effective dose or amount may prevent or delay the onset of a disease or one or more symptoms of a disease when the effect for which it is being administered is to treat a person who is at risk of developing the disease.
  • administering means any suitable method for administering cells to a subject, including parenteral, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial.
  • compositions described herein are administered at the same time, just prior to, or just after the administration of one or more additional therapies, for example cancer therapies such as chemotherapy, hormonal therapy, radiotherapy, or immunotherapy.
  • additional therapies such as chemotherapy, hormonal therapy, radiotherapy, or immunotherapy.
  • the compositions of the invention can be administered alone or can be coadministered to the patient.
  • Coadministration is meant to include simultaneous or sequential administration of the compositions individually or in PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT combination (more than one composition).
  • the preparations can also be combined, when desired, with other active substances (e.g. to reduce metabolic degradation).
  • compositions will generally comprise agents for buffering and preservation in storage, and can include buffers and carriers for appropriate delivery, depending on the route of administration.
  • “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present invention without causing a significant adverse toxicological effect on the patient.
  • Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidines, and the like.
  • Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention.
  • Pluripotent stem cells have the ability to both proliferate continuously in culture and, under appropriate growth conditions, differentiate into lineage restricted cell types representative of all three primary germ layers: endoderm, mesoderm and ectoderm (U.S. Patent Nos.5,843,780; 6,200,806; 7,029,913; Shamblott et al., (1998) Proc. Natl. Acad. Sci. USA 95:13726; Takahashi et al., (2007) Cell 131(5):861; Yu et al., (2007) Science 318:5858).
  • a pluripotent stem cell will, under appropriate growth conditions, be able to form at least one cell type from each of the three primary germ layers: mesoderm, endoderm and ectoderm.
  • the PS cells may originate from pre-embryonic, embryonic or fetal tissue or mature differentiated cells.
  • an established PS cell line may be a suitable source of cells for practicing the invention.
  • the hPS cells are not derived from a malignant source.
  • hPS cells will form teratomas when implanted in an immuno-deficient mouse, e.g. a SCID mouse.
  • Prototype “human Pluripotent Stem cells” are pluripotent cells derived from pre-embryonic, embryonic, or fetal tissue at any time after fertilization, and have the characteristic of being capable under appropriate conditions of producing progeny of several different cell types that are derivatives of all of the three germinal layers (endoderm, mesoderm, and ectoderm), according to a standard art-accepted test, such as the ability to form a teratoma in 8-12 week old SCID mice.
  • hPS cells are not derived from a cancer cell or other malignant source. It is desirable (but not always necessary) that the cells be euploid.
  • Exemplary are embryonic stem cells and embryonic germ cells used as existing cell lines or established from primary embryonic tissue of human origin. This invention can also be practiced using pluripotent cells obtained from primary embryonic tissue, without first establishing an undifferentiated cell line.
  • hPS cells can be propagated continuously in culture, using culture conditions that promote proliferation while inhibiting differentiation. Traditionally, ES cells are cultured on a PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT layer of feeder cells, typically fibroblasts derived from embryonic or fetal tissue (Thomson et al., Science 282: 1145, 1998).
  • hPS cells can be maintained in an undifferentiated state even without feeder cells.
  • the environment for feeder-free cultures includes a suitable culture substrate, such as ECM- based hydrogel, such as solubilized basement membrane preparation extracted from the Engelbreth-Holm- Swarm (EHS) mouse sarcoma (e.g., MATRIGEL®), or laminin.
  • EHS Engelbreth-Holm- Swarm
  • MATRIGEL® solubilized basement membrane preparation extracted from the Engelbreth-Holm- Swarm
  • laminin e.g., solubilized basement membrane preparation extracted from the Engelbreth-Holm- Swarm (EHS) mouse sarcoma (e.g., MATRIGEL®), or laminin.
  • EHS Engelbreth-Holm- Swarm
  • laminin laminin
  • Such factors may be introduced into the medium by culturing the medium with cells secreting such factors, such as irradiated primary mouse embryonic fibroblasts, telomerized mouse fibroblasts, or fibroblast-like cells derived from hPS cells (U.S. Patent 6,642,048).
  • Medium can be conditioned by plating the feeders in a serum free medium such as Knock-Out DMEM (Gibco), supplemented with serum replacement ranging from 10-30%, preferably 20% (US 2002/0076747 Al, Life Technologies Inc.) and 4 ng/mL bFGF.
  • a serum free medium such as Knock-Out DMEM (Gibco)
  • serum replacement ranging from 10-30%, preferably 20% (US 2002/0076747 Al, Life Technologies Inc.) and 4 ng/mL bFGF.
  • the culturing in step (b) is dynamic. In embodiments, the culturing in step (b) is static.
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0063]
  • the removing of step (c) includes an enzyme-free dissociation reagent. In embodiments, the removing of step (c) does not include manual dissection or manual scraping. In embodiments, the removing of step (c) does not include manual dissection. In embodiments, the removing of step (c) does not include manual scraping. [0064] In embodiments, the first period of time is between about 1 day to about 9 days.
  • the first period of time is between about 2 days to about 9 days. In embodiments, the first period of time is between about 3 days to about 9 days. In embodiments, the first period of time is between about 4 days to about 9 days. In embodiments, the first period of time is between about 5 days to about 9 days. In embodiments, the first period of time is between about 6 days to about 9 days. In embodiments, the first period of time is between about 7 days to about 9 days. In embodiments, the first period of time is between about 8 days to about 9 days. [0065] In embodiments, the first period of time is between 1 day to 8 days. In embodiments, the first period of time is between 1 day to 7 days. In embodiments, the first period of time is between 1 day to 6 days.
  • the first period of time is between 1 day to 5 days. In embodiments, the first period of time is between 1 day to 4 days. In embodiments, the first period of time is between about 1 day to 3 days. In embodiments, the first period of time is between 1 day to 2 days. In embodiments, the first period of time is between 1 day to 9 days. In embodiments, the first period of time is between 2 days to 9 days. In embodiments, the first period of time is between 3 days to 9 days. In embodiments, the first period of time is between 4 days to 9 days. In embodiments, the first period of time is between 5 days to 9 days. In embodiments, the first period of time is between 6 days to 9 days. In embodiments, the first period of time is between 7 days to 9 days.
  • the first period of time is between 8 days to 9 days. [0066] In embodiments, the first period of time is between about 1 day to about 8 days. In embodiments, the first period of time is between 1 day to 7 days. In embodiments, the first period of time is between 1 day to 6 days. In embodiments, the first period of time is between 1 day to 5 days. In embodiments, the first period of time is between 1 day to 4 days. In embodiments, the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT first period of time is between 1 day to 3 days. In embodiments, the first period of time is between 1 day to 2 days.
  • the differentiation cocktail includes at least one exogenous cytokine, wherein the at least one exogenous cytokine includes granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3).
  • the at least one exogenous cytokine includes granulocyte- macrophage colony stimulating factor (GM-CSF).
  • the at least one exogenous cytokine includes bond morphogenic protein 4 (BMP-4).
  • the at least one exogenous cytokine includes vascular endothelial growth factor (VEGF). In embodiments, the at least one exogenous cytokine includes stem cell factor (SCF). In embodiments, the at least one exogenous cytokine includes fetal liver kinase ligand (FLT3L). In embodiments, the at least one exogenous cytokine includes thrombopoietin (TPO). In embodiments, the at least one exogenous cytokine includes interleukin 3 (IL-3). In embodiments, one or more of the recited cytokines may be expressly excluded. [0068] In embodiments, the differentiation cocktail includes GM-CSF.
  • the differentiation cocktail includes GM-CSF and SCF. In embodiments, the differentiation cocktail includes GM-CSF, SCF, and VEGF. In embodiments, the differentiation cocktail includes GM- CSF, SCF, VEGF, and BMP-4. In embodiments, the BMP-4 is an animal-free Good Manufacturing Process (GMP) grade BMP-4.
  • GMP animal-free Good Manufacturing Process
  • an iDC of the population of iDCs in step (d) expresses CD45 and CD86. In embodiments, an iDC of the population of iDCs in step (d) does not express CD38.
  • the second period of time is between about 1 day to about 32 days.
  • the second period of time is between about 2 days to about 32 days. In embodiments, the second period of time is between about 3 days to about 32 days. In embodiments, the second period of time is between about 4 days to about 32 days. In embodiments, the second period of time is between about 5 days to about 32 days. In embodiments, the second period of time is between about 6 days to about 32 days.
  • the second period of time is between about 7 days to about 32 days. In embodiments, the second period of time is between about 8 days to about 32 days. In embodiments, the second period of time is between about 9 days to about 32 days.
  • the second period of time is between about 10 days to about 32 days. In embodiments, the second period of time is between about 11 days to about 32 days. In embodiments, the second period of time is between about 12 days to about 32 days. In embodiments, the second period of time is between about 13 days to about 32 days. In embodiments, the second period of time is between about 14 days to about 32 days. In embodiments, the second period of time is between about 15 days to about 32 days. In embodiments, the second period of time is between about 16 days to about 32 days. In embodiments, the second period of time is between about 17 days to about 32 days. In embodiments, the second period of time is between about 18 days to about 32 days. In embodiments, the second period of time is between about 19 days to about 32 days.
  • the second period of time is between about 20 days to about 32 days. In embodiments, the second period of time is between about 21 days to about 32 days. In embodiments, the second period of time is between about 22 days to about 32 days. In embodiments, the second period of time is between about 23 days to about 32 days. In embodiments, the second period of time is between about 24 days to about 32 days. In embodiments, the second period of time is between about 25 days to about 32 days. In embodiments, the second period of time is between about 26 days to about 32 days. In embodiments, the second period of time is between about 27 days to about 32 days. In embodiments, the second period of time is between about 28 days to about 32 days. In embodiments, the second period of time is between about 29 days to about 32 days.
  • the second period of time is between about 30 days to about 32 days. In embodiments, the second period of time is between about 31 days to about 32 days. [0071] In embodiments, the second period of time is between about 1 day to about 31 days. In embodiments, the second period of time is between about 1 day to about 30 days. In embodiments, the second period of time is between about 1 day to about 29 days. In embodiments, the second period of time is between about 1 day to about 28 days. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the second period of time is between about 1 day to about 27 days. In embodiments, the second period of time is between about 1 day to about 26 days.
  • the second period of time is between about 1 day to about 25 days. In embodiments, the second period of time is between about 1 day to about 24 days. In embodiments, the second period of time is between about 1 day to about 23 days. In embodiments, the second period of time is between about 1 day to about 22 days. In embodiments, the second period of time is between about 1 day to about 21 days. In embodiments, the second period of time is between about 1 day to about 20 days. In embodiments, the second period of time is between about 1 day to about 19 days. In embodiments, the second period of time is between about 1 day to about 18 days. In embodiments, the second period of time is between about 1 day to about 17 days. In embodiments, the second period of time is between about 1 day to about 16 days.
  • the second period of time is between about 1 day to about 15 days. In embodiments, the second period of time is between about 1 day to about 14 days. In embodiments, the second period of time is between about 1 day to about 13 days. In embodiments, the second period of time is between about 1 day to about 12 days. In embodiments, the second period of time is between about 1 day to about 11 days. In embodiments, the second period of time is between about 1 day to about 10 days. In embodiments, the second period of time is between about 1 day to about 9 days. In embodiments, the second period of time is between about 1 day to about 8 days. In embodiments, the second period of time is between about 1 day to about 7 days. In embodiments, the second period of time is between about 1 day to about 6 days.
  • the second period of time is between about 1 day to about 5 days. In embodiments, the second period of time is between about 1 day to about 4 days. In embodiments, the second period of time is between about 1 day to about 3 days. In embodiments, the second period of time is between about 1 day to about 2 days. [0072] In embodiments, the second period of time is between 1 day to 32 days. In embodiments, the second period of time is between 2 days to 32 days. In embodiments, the second period of time is between 3 days to 32 days. In embodiments, the second period of time is between 4 days to 32 days. In embodiments, the second period of time is between 5 days to 32 PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • the second period of time is between 6 days to 32 days. In embodiments, the second period of time is between 7 days to 32 days. In embodiments, the second period of time is between 8 days to 32 days. In embodiments, the second period of time is between 9 days to 32 days. In embodiments, the second period of time is between 10 days to 32 days. In embodiments, the second period of time is between 11 days to 32 days. In embodiments, the second period of time is between 12 days to 32 days. In embodiments, the second period of time is between 13 days to 32 days. In embodiments, the second period of time is between 14 days to 32 days. In embodiments, the second period of time is between 15 days to 32 days.
  • the second period of time is between 16 days to 32 days. In embodiments, the second period of time is between 17 days to 32 days. In embodiments, the second period of time is between 18 days to 32 days. In embodiments, the second period of time is between 19 days to 32 days. In embodiments, the second period of time is between 20 days to 32 days. In embodiments, the second period of time is between 21 days to 32 days. In embodiments, the second period of time is between 22 days to 32 days. In embodiments, the second period of time is between 23 days to 32 days. In embodiments, the second period of time is between 24 days to 32 days. In embodiments, the second period of time is between 25 days to 32 days. In embodiments, the second period of time is between 26 days to 32 days.
  • the second period of time is between 27 days to 32 days. In embodiments, the second period of time is between 28 days to 32 days. In embodiments, the second period of time is between 29 days to 32 days. In embodiments, the second period of time is between 30 days to 32 days. In embodiments, the second period of time is between 31 days to 32 days. [0073] In embodiments, the second period of time is between 1 day to 31 days. In embodiments, the second period of time is between 1 day to 30 days. In embodiments, the second period of time is between 1 day to 29 days. In embodiments, the second period of time is between 1 day to 28 days. In embodiments, the second period of time is between 1 day to 27 days. In embodiments, the second period of time is between 1 day to 26 days.
  • the second period of time is between 1 day to 25 days. In embodiments, the second period of time is between 1 day to 24 days. In embodiments, the second period of time is between 1 day to 23 days. In embodiments, the second period of time is between 1 day to 22 days. In embodiments, the second PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT period of time is between 1 day to 21 days. In embodiments, the second period of time is between 1 day to 20 days. In embodiments, the second period of time is between 1 day to 19 days. In embodiments, the second period of time is between 1 day to 18 days. In embodiments, the second period of time is between 1 day to 17 days.
  • the second period of time is between 1 day to 16 days. In embodiments, the second period of time is between 1 day to 15 days. In embodiments, the second period of time is between 1 day to 14 days. In embodiments, the second period of time is between 1 day to 13 days. In embodiments, the second period of time is between 1 day to 12 days. In embodiments, the second period of time is between 1 day to 11 days. In embodiments, the second period of time is between 1 day to 10 days. In embodiments, the second period of time is between 1 day to 9 days. In embodiments, the second period of time is between 1 day to 8 days. In embodiments, the second period of time is between 1 day to 7 days. In embodiments, the second period of time is between 1 day to 6 days.
  • step (d) includes dynamic culturing of the cultured cells followed by static culturing of the cultured cells.
  • the dynamic culturing is for about 1 day to about 5 days.
  • the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days.
  • the dynamic culturing is for about 1 day.
  • the dynamic culturing is for about 2 days.
  • the dynamic culturing is for about 3 days. In embodiments, the dynamic culturing is for about 4 days. In embodiments, the dynamic culturing is for about 5 days. In embodiments, the dynamic culturing is for 1 day, 2 days, 3 days, 4 days, or 5 days. In embodiments, the dynamic culturing is for 1 day. In embodiments, the dynamic culturing is for 2 days. In embodiments, the dynamic culturing is for 3 days. In embodiments, the dynamic culturing is for 4 days. In embodiments, the dynamic culturing is for 5 days. [0075] In embodiments, the static culturing is for about 3 days to about 27 days.
  • the static culturing is for about 4 days to about 27 days.
  • the static PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT culturing is for about 5 days to about 27 days.
  • the static culturing is for about 6 days to about 27 days.
  • the static culturing is for about 7 days to about 27 days.
  • the static culturing is for about 8 days to about 27 days.
  • the static culturing is for about 9 days to about 27 days.
  • the static culturing is for about 10 days to about 27 days.
  • the static culturing is for about 11 days to about 27 days. In embodiments, the static culturing is for about 12 days to about 27 days. In embodiments, the static culturing is for about 13 days to about 27 days. In embodiments, the static culturing is for about 14 days to about 27 days. In embodiments, the static culturing is for about 15 days to about 27 days. In embodiments, the static culturing is for about 16 days to about 27 days. In embodiments, the static culturing is for about 17 days to about 27 days. In embodiments, the static culturing is for about 18 days to about 27 days. In embodiments, the static culturing is for about 19 days to about 27 days.
  • the static culturing is for about 20 days to about 27 days. In embodiments, the static culturing is for about 21 days to about 27 days. In embodiments, the static culturing is for about 22 days to about 27 days. In embodiments, the static culturing is for about 23 days to about 27 days. In embodiments, the static culturing is for about 24 days to about 27 days. In embodiments, the static culturing is for about 25 days to about 27 days. In embodiments, the static culturing is for about 26 days to about 27 days. [0076] In embodiments, the static culturing is for about 3 days to about 26 days. In embodiments, the static culturing is for about 3 days to about 25 days.
  • the static culturing is for about 3 days to about 24 days. In embodiments, the static culturing is for about 3 days to about 23 days. In embodiments, the static culturing is for about 3 days to about 22 days. In embodiments, the static culturing is for about 3 days to about 21 days. In embodiments, the static culturing is for about 3 days to about 20 days. In embodiments, the static culturing is for about 3 days to about 19 days. In embodiments, the static culturing is for about 3 days to about 18 days. In embodiments, the static culturing is for about 3 days to about 17 days. In embodiments, the static culturing is for about 3 days to about 16 days.
  • the static culturing is for about 3 days to about 15 days. In embodiments, the static culturing is for about 3 days to about 14 days. In embodiments, the static culturing is for about 3 days to about 13 days. In embodiments, PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT the static culturing is for about 3 days to about 12 days. In embodiments, the static culturing is for about 3 days to about 11 days. In embodiments, the static culturing is for about 3 days to about 10 days. In embodiments, the static culturing is for about 3 days to about 9 days.
  • the static culturing is for about 3 days to about 8 days. In embodiments, the static culturing is for about 3 days to about 7 days. In embodiments, the static culturing is for about 3 days to about 6 days. In embodiments, the static culturing is for about 3 days to about 5 days. In embodiments, the static culturing is for about 3 days to about 4 days. [0077] In embodiments, the static culturing is for 3 days to 27 days. In embodiments, the static culturing is for 4 days to 27 days. In embodiments, the static culturing is for 5 days to 27 days. In embodiments, the static culturing is for 6 days to 27 days.
  • the static culturing is for 7 days to 27 days. In embodiments, the static culturing is for 8 days to 27 days. In embodiments, the static culturing is for 9 days to 27 days. In embodiments, the static culturing is for 10 days to 27 days. In embodiments, the static culturing is for 11 days to 27 days. In embodiments, the static culturing is for 12 days to 27 days. In embodiments, the static culturing is for 13 days to 27 days. In embodiments, the static culturing is for 14 days to 27 days. In embodiments, the static culturing is for 15 days to 27 days. In embodiments, the static culturing is for 16 days to 27 days.
  • the static culturing is for 17 days to 27 days. In embodiments, the static culturing is for 18 days to 27 days. In embodiments, the static culturing is for 19 days to 27 days. In embodiments, the static culturing is for 20 days to 27 days. In embodiments, the static culturing is for 21 days to 27 days. In embodiments, the static culturing is for 22 days to 27 days. In embodiments, the static culturing is for 23 days to 27 days. In embodiments, the static culturing is for 24 days to 27 days. In embodiments, the static culturing is for 25 days to 27 days. In embodiments, the static culturing is for 26 days to 27 days.
  • the static culturing is for 3 days to 26 days. In embodiments, the static culturing is for 3 days to 25 days. In embodiments, the static culturing is for 3 days to 24 days. In embodiments, the static culturing is for 3 days to 23 days. In embodiments, the static culturing is for 3 days to 22 days. In embodiments, the static culturing is for 3 days to 21 days. In embodiments, the static culturing is for 3 days to 20 days. In embodiments, the static culturing is PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT for 3 days to 19 days.
  • the static culturing is for 3 days to 18 days. In embodiments, the static culturing is for 3 days to 17 days. In embodiments, the static culturing is for 3 days to 16 days. In embodiments, the static culturing is for 3 days to 15 days. In embodiments, the static culturing is for 3 days to 14 days. In embodiments, the static culturing is for 3 days to 13 days. In embodiments, the static culturing is for 3 days to 12 days. In embodiments, the static culturing is for 3 days to 11 days. In embodiments, the static culturing is for 3 days to 10 days. In embodiments, the static culturing is for 3 days to 9 days.
  • the static culturing is for 3 days to 8 days. In embodiments, the static culturing is for 3 days to 7 days. In embodiments, the static culturing is for 3 days to 6 days. In embodiments, the static culturing is for 3 days to 5 days. In embodiments, the static culturing is for 3 days to 4 days.
  • the maturation cocktail includes at least one exogenous cytokine, wherein the at least one exogenous cytokine includes GM-CSF, interleukin 4 (IL-4), interferon embodiments, the at least one exogenous cytokine includes GM-CSF. In embodiments, the at least one exogenous cytokine includes interleukin 4 (IL-4).
  • the at least one one exogenous cytokine includes prostaglandin E2 (PGE-2). In embodiments, the at least one exogenous cytokine includes polyinosinic-polycytidylic acid (poly(I:C)). In embodiments, the at exogenous cytokine includes CD40L. In embodiments, one or more of the recited cytokines may be expressly excluded. [0080] In embodiments, the maturation cocktail includes GM-CSF and IL-4. In embodiments, PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • an mDC of the population of mDCs in step (e) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express one or more of CD14, TRA160, and SSEA-5.
  • an mDC of the population of mDCs in step (e) expresses CD40.
  • an mDC of the population of mDCs in step (e) expresses CD45.
  • an mDC of the population of mDCs in step (e) expresses CD83.
  • an mDC of the population of mDCs in step (e) expresses CD86. In embodiments, an mDC of the population of mDCs in step (e) expresses HLA-DR. In embodiments, an mDC of the population of mDCs in step (e) expresses HLA-A. In embodiments, an mDC of the population of mDCs in step (e) expresses CCR7. In embodiments, an mDC of the population of mDCs in step (e) expresses CD141. In embodiments, an mDC of the population of mDCs in step (e) expresses CD11c.
  • an mDC of the population of mDCs in step (e) expresses CD45 and CD83. [0082] In embodiments, an mDC of the population of mDCs in step (e) does not express CD14. In embodiments, an mDC of the population of mDCs in step (e) does not express TRA160. In embodiments, an mDC of the population of mDCs in step (e) does not express SSEA-5. In embodiments, an mDC of the population of mDCs in step (e) does not express TRA160 and SSEA-5.
  • an mDC of the population of mDCs in step (e) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express D14, CD45, TRA160, and SSEA-5.
  • an mDC of the population of mDCs in step (e) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express D14, TRA160, and SSEA-5.
  • at least 70% of the population of mDCs in step (e) express CD45.
  • At least 75% of the population of mDCs in step (e) express CD45. In embodiments, at least 80% of the population of mDCs in step (e) express CD45. In embodiments, at least 85% of the population of mDCs in step (e) express CD45. In embodiments, at least 90% of the population of mDCs in step (e) express CD45. In embodiments, at least 95% of the population of mDCs in step (e) express CD45. In embodiments, at least 96% of the population of mDCs in step PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT (e) express CD45.
  • At least 97% of the population of mDCs in step (e) express CD45. In embodiments, at least 98% of the population of mDCs in step (e) express CD45. In embodiments, at least 99% of the population of mDCs in step (e) express CD45. [0085] In embodiments, at least 70% of the population of mDCs in step (e) express CD83. In embodiments, at least 75% of the population of mDCs in step (e) express CD83. In embodiments, at least 80% of the population of mDCs in step (e) express CD83. In embodiments, at least 85% of the population of mDCs in step (e) express CD83.
  • At least 90% of the population of mDCs in step (e) express CD83. In embodiments, at least 95% of the population of mDCs in step (e) express CD83. In embodiments, at least 96% of the population of mDCs in step (e) express CD83. In embodiments, at least 97% of the population of mDCs in step (e) express CD83. In embodiments, at least 98% of the population of mDCs in step (e) express CD83. In embodiments, at least 99% of the population of mDCs in step (e) express CD83. [0086] In embodiments, at least 70% of the population of mDCs in step (e) express CD86, CD40, or HLA-A.
  • At least 75% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 80% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 85% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 90% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 95% of the population of mDCs in step (e) express CD86, CD40, or HLA-A.
  • At least 96% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 97% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 98% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 99% of the population of mDCs in step (e) express CD86, CD40, or HLA- A. [0087] In embodiments, at least 70% of the population of mDCs in step (e) express CD86. In embodiments, at least 75% of the population of mDCs in step (e) express CD86.
  • At least 80% of the population of mDCs in step (e) express CD86. In embodiments, at least 85% of the population of mDCs in step (e) express CD86. In embodiments, at least 90% of the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT population of mDCs in step (e) express CD86. In embodiments, at least 95% of the population of mDCs in step (e) express CD86. In embodiments, at least 96% of the population of mDCs in step (e) express CD86. In embodiments, at least 97% of the population of mDCs in step (e) express CD86.
  • At least 98% of the population of mDCs in step (e) express CD86. In embodiments, at least 99% of the population of mDCs in step (e) express CD86.
  • at least 70% of the population of mDCs in step (e) express CD40. In embodiments, at least 75% of the population of mDCs in step (e) express CD40. In embodiments, at least 80% of the population of mDCs in step (e) express CD40. In embodiments, at least 85% of the population of mDCs in step (e) express CD40. In embodiments, at least 90% of the population of mDCs in step (e) express CD40.
  • At least 95% of the population of mDCs in step (e) express CD40. In embodiments, at least 96% of the population of mDCs in step (e) express CD40. In embodiments, at least 97% of the population of mDCs in step (e) express CD40. In embodiments, at least 98% of the population of mDCs in step (e) express CD40. In embodiments, at least 99% of the population of mDCs in step (e) express CD40. [0089] In embodiments, at least 70% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 75% of the population of mDCs in step (e) express HLA-A.
  • At least 80% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 85% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 90% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 95% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 96% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 97% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 98% of the population of mDCs in step (e) express HLA-A.
  • At least 99% of the population of mDCs in step (e) express HLA-A.
  • at least 20% of the population of mDCs in step (e) express HLA-DR.
  • at least 25% of the population of mDCs in step (e) express HLA-DR.
  • at least 30% of the population of mDCs in step (e) express HLA-DR.
  • at least 35% of the population of mDCs in step (e) express HLA-DR.
  • At least 40% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 45% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 50% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 55% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 60% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 65% of the population of mDCs in step (e) express HLA-DR.
  • At least 70% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 75% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 80% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 85% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 90% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 95% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 96% of the population of mDCs in step (e) express HLA-DR.
  • At least 97% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 98% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 99% of the population of mDCs in step (e) express HLA-DR. [0091] In embodiments, less than 20% of the population of mDCs in step (e) express CD14. In embodiments, less than 19% of the population of mDCs in step (e) express CD14. In embodiments, less than 18% of the population of mDCs in step (e) express CD14. In embodiments, less than 17% of the population of mDCs in step (e) express CD14.
  • less than 16% of the population of mDCs in step (e) express CD14. In embodiments, less than 15% of the population of mDCs in step (e) express CD14. In embodiments, less than 14% of the population of mDCs in step (e) express CD14. In embodiments, less than 13% of the population of mDCs in step (e) express CD14. In embodiments, less than 12% of the population of mDCs in step (e) express CD14. In embodiments, less than 11% of the population of mDCs in step (e) express CD14. In embodiments, less than 10% of the population of mDCs in step (e) express CD14.
  • less than 9% of the population of mDCs in step (e) express CD14. In embodiments, less than 8% of the population of mDCs in step (e) express CD14. In embodiments, less than 7% PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT of the population of mDCs in step (e) express CD14. In embodiments, less than 6% of the population of mDCs in step (e) express CD14. In embodiments, less than 5% of the population of mDCs in step (e) express CD14. In embodiments, less than 4% of the population of mDCs in step (e) express CD14.
  • less than 3% of the population of mDCs in step (e) express CD14. In embodiments, less than 2% of the population of mDCs in step (e) express CD14. In embodiments, less than 1% of the population of mDCs in step (e) express CD14. [0092] In embodiments, less than 20% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 19% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 18% of the population of mDCs in step (e) are CD45+ and CD83-.
  • less than 17% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 16% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 15% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 14% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 13% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 12% of the population of mDCs in step (e) are CD45+ and CD83-.
  • less than 11% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 10% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 9% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 8% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 7% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 6% of the population of mDCs in step (e) are CD45+ and CD83-.
  • less than 5% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 4% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 3% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 2% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 1% of the population of mDCs in step (e) are CD45+ and CD83-. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • the third period of time is between about 1 day to about 10 days. In embodiments, the third period of time is between about 2 days to about 10 days. In embodiments, the third period of time is between about 3 days to about 10 days. In embodiments, the third period of time is between about 4 days to about 10 days. In embodiments, the third period of time is between about 5 days to about 10 days. In embodiments, the third period of time is between about 6 days to about 10 days. In embodiments, the third period of time is between about 7 days to about 10 days. In embodiments, the third period of time is between about 8 days to about 10 days. In embodiments, the third period of time is between about 9 days to about 10 days.
  • the third period of time is between about 1 day to about 9 days. In embodiments, the third period of time is between about 1 day to about 8 days. In embodiments, the third period of time is between about 1 day to about 7 days. In embodiments, the third period of time is between about 1 day to about 6 days. In embodiments, the third period of time is between about 1 day to about 5 days. In embodiments, the third period of time is between about 1 day to about 4 days. In embodiments, the third period of time is between about 1 day to about 3 days. In embodiments, the third period of time is between about 1 day to about 2 days. [0095] In embodiments, the third period of time is between 1 day to 10 days.
  • the third period of time is between 2 days to 10 days. In embodiments, the third period of time is between 3 days to 10 days. In embodiments, the third period of time is between 4 days to 10 days. In embodiments, the third period of time is between 5 days to 10 days. In embodiments, the third period of time is between about 6 days to about 10 days. In embodiments, the third period of time is between 7 days to 10 days. In embodiments, the third period of time is between 8 days to 10 days. In embodiments, the third period of time is between 9 days to 10 days. [0096] In embodiments, the third period of time is between 1 day to 9 days. In embodiments, the third period of time is between 1 day to 8 days.
  • the third period of time is between 1 day to 7 days. In embodiments, the third period of time is between 1 day to 6 days. In embodiments, the third period of time is between 1 day to 5 days. In embodiments, the third period of time is between 1 day to 4 days. In embodiments, the third period of time is between 1 day to 3 days. In embodiments, the third period of time is between 1 day to 2 days.
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0097]
  • step (e) includes static culturing of the population of iDCs.
  • the static culturing is for about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days. In embodiments, the static culturing is for about 4 days. In embodiments, the static culturing is for about 5 days. In embodiments, the static culturing is for about 6 days. In embodiments, the static culturing is for about 7 days. In embodiments, the static culturing is for about 8 days. In embodiments, the static culturing is for about 9 days. In embodiments, the static culturing is for about 10 days. In embodiments, the static culturing is for 4 days. In embodiments, the static culturing is for 5 days.
  • the static culturing is for 6 days. In embodiments, the static culturing is for 7 days. In embodiments, the static culturing is for 8 days. In embodiments, the static culturing is for 9 days. In embodiments, the static culturing is for 10 days. [0098] In embodiments, any one of or both of steps (d) and (e) are performed under serum free conditions. In embodiments, step (d) is performed under serum free conditions. In embodiments, step (e) is performed under serum free conditions. In embodiments, steps (d) and (e) are performed under serum free conditions. In embodiments, any one of or both of steps (d) and (e) are performed in the absence of feeder cells.
  • step (d) is performed in the absence of feeder cells.
  • step (e) is performed in the absence of feeder cells.
  • steps (d) and (e) are performed in the absence of feeder cells.
  • any one of or both of steps (d) and (e) are performed in the absence of stromal cells.
  • step (d) is performed in the absence of stromal cells.
  • step (e) is performed in the absence of stromal cells.
  • steps (d) and (e) are performed in the absence of stromal cells. [0099] In embodiments, steps (a), (b), and (c) are repeated after step (c), prior to step (d).
  • steps (a), (b) and (c) are each repeated three times prior to step (d).
  • steps (a), (b) and (c) combined are about three days to about five days.
  • steps (a), (b) and (c) combined are about three days.
  • steps (a), (b) and (c) combined are about four days.
  • steps (a), (b) and (c) combined are about five days.
  • steps (a), (b) and (c) combined are three days.
  • steps PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT (a), (b) and (c) combined are four days.
  • steps (a), (b) and (c) combined are five days.
  • the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in steps (a) through (c).
  • the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in step (a).
  • the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in step (b).
  • the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in step (c).
  • At least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. In embodiments, at least about 80% of the undifferentiated and pluripotent cells express SSEA-5. In embodiments, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. In embodiments, at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog. In embodiments, at least about 70% of the undifferentiated and pluripotent cells express Oct-4.
  • step (b) further includes changing the growth medium to remove accumulated lactate.
  • the growth medium includes Y-27632.
  • the culturing in step (b) is dynamic.
  • the cultured cells in step (b) include hematopoietic cell intermediates.
  • the hematopoietic cell intermediates express CD34, CD90.
  • the hematopoietic cell intermediates express CD34.
  • the hematopoietic cell intermediates express CD90.
  • the hematopoietic cell intermediates do not express CD38.
  • the microcarrier includes one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof.
  • the microcarrier includes polystyrene.
  • the microcarrier includes cross-linked dextran.
  • the microcarrier includes magnetic particles.
  • the microcarrier includes microchips.
  • the microcarrier includes cellulose.
  • the microcarrier includes hydroxylated methacrylate.
  • the microcarrier includes collagen.
  • the microcarrier includes gelatin. In embodiments, the microcarrier includes polystyrene, plastic. In embodiments, the microcarrier includes glass. In embodiments, the microcarrier includes ceramic. In embodiments, the microcarrier includes silicon. In embodiments, one or more of the recited microcarriers may be expressly excluded. [0103] In embodiments, the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof. In embodiments, the microcarrier is spherical. In embodiments, the microcarrier is smooth. In embodiments, the microcarrier is macroporous. In embodiments, the microcarrier is rod-shaped. In embodiments, the microcarrier is not coated.
  • the microcarrier is coupled with protamine or polylysine. In embodiments, the microcarrier is coupled with protamine. In embodiments, the microcarrier is coupled with polylysine. In embodiments, the microcarrier has a neutral charge. In embodiments, the microcarrier is negatively charged. In embodiments, the microcarrier is hydrophilic. In embodiments, one or more of the recited microcarriers may be expressly excluded.
  • the ECM component comprises a matrigel, laminin, vitronectin, collagen, a derivative thereof, or any combination thereof.
  • the ECM component includes a matrigel, a laminin, a vitronectin, a collagen, a derivative thereof, or any combination thereof. In embodiments, the ECM component includes a matrigel. In embodiments, the ECM component includes a laminin. In embodiments, the ECM component includes a vitronectin. In embodiments, the ECM component includes a collagen. In embodiments, the laminin is human PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT laminin. In embodiments, the human laminin is 511 E8 fragment. In embodiments, one or more of the recited ECM components may be expressly excluded.
  • the mDCs are characterized by: (i) expression of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c, but not expressing CD14, CD45, TRA160, and SSEA-5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii).
  • the mDCs are characterized by: (i) expression of at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c, but not expressing one or more of CD14, TRA160, and SSEA-5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii).
  • the mDCs are characterized by expression of at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c.
  • the mDCs are characterized by expression of CD40.
  • the mDCs are characterized by expression of CD45.
  • the mDCs are characterized by expression of CD83.
  • the mDCs are characterized by expression of CD86.
  • the mDCs are characterized by expression of HLA-DR.
  • the mDCs are characterized by expression of HLA-A.
  • the mDCs are characterized by expression of CD40.
  • the mDCs are characterized by expression of CCR7. In embodiments, the mDCs are characterized by expression of CD141. In embodiments, the mDCs are characterized by expression of CD11c. In embodiments, the mDCs are characterized by expression of CD45 and CD83. [0108] In embodiments, the mDCs are characterized by not expressing one or more of CD14, TRA160, and SSEA-5. In embodiments, the mDCs are characterized by not expressing CD14. In embodiments, the mDCs are characterized by not expressing TRA160. In embodiments, the mDCs are characterized by not expressing SSEA-5.
  • the mDCs are characterized by not expressing TRA160 and SSEA-5. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0109] In embodiments, the mDCs are characterized by exhibiting CCL19-induced migration. In embodiments, the mDCs are characterized by an ability to present an antigen at the cell surface. [0110] In embodiments, the mDCs are unable to replicate. In embodiments, the mDCs are irradiated to prevent replication. [0111] In embodiments, further including (f) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen.
  • the population of mDCs is contacted with two or more antigens.
  • the antigen is a tumor antigen.
  • the tumor antigen includes a tumor associated antigen (TAA).
  • TAA tumor associated antigen
  • the antigen is a neoantigen.
  • the antigen is a bacteria associated antigen.
  • the antigen is a viral antigen.
  • the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof.
  • the tumor antigen includes a tumor-specific antigen (TSA).
  • the nucleic acid includes an RNA.
  • the RNA includes an mRNA encoding hTERT or fragment thereof.
  • the fragment includes one or more immunogenic epitopes of hTERT.
  • the RNA includes an mRNA encoding lysosomal-associated membrane protein 1 (LAMP1) or a fragment thereof.
  • the RNA includes an mRNA encoding heat shock protein 96 (HSP96).
  • the population of mDCs including the one or more antigens is cryopreserved.
  • the cryopreserved population of mDCs including the one or more antigens is irradiated. In embodiments, the cryopreserved population of mDCs is irradiated to prevent replication.
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT CELL COMPOSITIONS [0113] Antigen presenting cells of this invention are often referred to in this disclosure as “dendritic cells”. However, this is not meant to imply any morphological, phenotypic, or functional feature beyond what is explicitly required.
  • the term is used to refer to cells that are phagocytic or can present antigen to T lymphocytes, falling within the general class of monocytes, macrophages, dendritic cells and the like, such as may be found circulating in the blood or lymph, or fixed in tissue sites.
  • Phagocytic properties of a cell can be determined according to their ability to take up labeled antigen or small particulates.
  • the ability of a cell to present antigen can be determined in a mixed lymphocyte reaction as described.
  • Certain types of dendritic cells and antigen-presenting cells in the body are first identified in tissue sites such as the skin or the liver; but regardless of their origin, location, and developmental pathway, they are considered in the art to fall within the general category of hematopoietic cells.
  • dendritic cells used in this disclosure also fall in the broad category of hematopoietic cells, whether produced through the hematopoietic or direct paradigm framed earlier, or through a related or combined pathway.
  • the putative role of hPS derived cells as antigen-presenting cells is provided in this disclosure as an explanation to facilitate the understanding of the reader. However, the theories expostulated here are not intended to limit the invention beyond what is explicitly required.
  • the hPS derived cells of this invention may be used therapeutically regardless of their mode of action, as long as they achieve a desirable clinical benefit in a substantial proportion of patients treated.
  • antigen-presenting cells may include, but are not necessarily limited to, dendritic cells, macrophages, and B cells.
  • the antigen-presenting cells are dendritic cells.
  • the cell expresses CD40.
  • the cell expresses CD45.
  • the cell expresses CD83.
  • the cell expresses CD86.
  • the cell expresses HLA-DR. In embodiments, the cell expresses HLA-A. In embodiments, the cell expresses CD40. In embodiments, the cell expresses CCR7. In embodiments, the cell expresses CD45 and CD83. In embodiments, the cell does not express CD14. In embodiments, the cell does not express TRA160. In embodiments, the cell does not express SSEA-5. In embodiments, the cell does not express TRA160 and SSEA-5.
  • the cell expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA- A, CD40 and CCR7, but does not express one or more of CD14, TRA160, and SSEA-5; and (ii) includes the one or more antigens.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier.
  • the one or more antigens include a tumor associated antigen (TAA), a tumor specific antigen (TSA), a neoantigen, or an antigen from an infectious organism.
  • the one or more antigens include a tumor associated antigen (TAA).
  • the one or more antigens include a tumor specific antigen (TSA).
  • the one or more antigens include a neoantigen.
  • the one or more antigens include an antigen from an infectious organism.
  • the TAA includes a human telomerase reverse transcriptase (hTERT) or fragment thereof.
  • the TAA includes a lysosomal-associated membrane protein 1 (LAMP1). In embodiments, the TAA includes a fusion protein. In embodiments, the TAA is an hTERT-LAMP1. In embodiments, the TAA includes HSP96. In embodiments, the TAA includes hTERT-LAMP1 and HSP96. In embodiments, the one or more antigens are not endogenous. In embodiments, the one or more antigens are encoded by a nucleic acid. [0124] In embodiments, the nucleic acid is introduced into the cell via electroporation, transfection, transduction. In embodiments, the nucleic acid is introduced into the cell via electroporation.
  • the electroporation includes high throughput flow electroporation.
  • the nucleic acid is introduced into the cell via transfection.
  • the transfection includes chemical-based transfection.
  • the chemical-based transfection includes a lipid-based transfection agent.
  • the lipid- based transfection agent is a positively charged (e.g., cationic) lipid-based transfection agent.
  • the chemical-based transfection includes calcium phosphate (e.g., Ca3(PO4)2).
  • the nucleic acid is introduced into the cell via transduction.
  • the transduction includes a viral vector.
  • the viral vector is an Adenoviral vector, an Adeno-associated virus (AAV) vector, or a lentiviral vector.
  • the viral vector is PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT an Adenoviral vector.
  • the viral vector is an Adeno-associated virus (AAV) vector.
  • the viral vector is a lentiviral vector.
  • the nucleic acid is mRNA or DNA. In embodiments, the nucleic acid is mRNA. In embodiments, the nucleic acid is DNA.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier.
  • the nucleic acid comprises a DNA.
  • the DNA is under the control of a promoter.
  • the promoter is an inducible promoter.
  • the inducing agent is desferrioxamine. In embodiments, the inducing agent is tetracycline. In embodiments, the agent is picolinic acid. In embodiments, the inducing agent is desferrioxamine. In embodiments, the nucleic acid includes an RNA. In embodiments, the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent. [0128] In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between about 0.6 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.7 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.8 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the cell vaccine includes between about 0.9 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between about 1.0 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.1 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.2 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.3 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.4 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between about 1.5 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.6 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.7 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.8 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.9 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between about 2.0 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 2.1 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 2.2 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 2.3 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 2.4 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 2.4 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 2.3 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 2.2 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 2.1 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells.
  • the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.9 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.8 x 10 7 dendritic cells. In embodiments, the cell vaccine includes PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT between about 0.5 x 10 7 dendritic cells to about 1.7 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.6 x 10 7 dendritic cells.
  • the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.4 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.3 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.2 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.1 x 10 7 dendritic cells.
  • the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 1.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 0.9 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 0.8 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 0.7 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 10 7 dendritic cells to about 0.6 x 10 7 dendritic cells.
  • the cell vaccine includes between 0.5 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.6 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.7 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.8 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.9 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between 1.0 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.1 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.2 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.3 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.4 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.5 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between 1.6 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.7 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.8 x 10 7 dendritic cells PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.9 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between 2.0 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 2.1 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 2.2 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 2.3 x 10 7 dendritic cells to abot 2.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 2.4 x 10 7 dendritic cells to 2.5 x 10 7 dendritic cells.
  • the cell vaccine includes between 0.5 x 10 7 dendritic cells to 2.4 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to about 2.3 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 2.2 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 2.1 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells.
  • the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.9 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.8 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.7 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.6 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.5 x 10 7 dendritic cells.
  • the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.4 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.3 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.2 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.1 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 1.0 x 10 7 dendritic cells.
  • the cell vaccine includes between 0.5 x 10 7 dendritic cells to 0.9 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 0.8 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 0.7 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 10 7 dendritic cells to 0.6 x 10 7 dendritic cells. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • the cell vaccine preferably includes between about 1,0 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.1 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.2 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.3 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells.
  • the cell vaccine includes between about 1.4 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.5 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.6 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.7 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1.8 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells.
  • the cell vaccine includes between about 1.9 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells. [0133] In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.9 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.8 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.7 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.6 x 10 7 dendritic cells.
  • the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.4 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.3 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.2 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 10 7 dendritic cells to about 1.1 x 10 7 dendritic cells.
  • the cell vaccine preferably includes between 1,0 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.1 x 10 7 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.2 x 10 7 dendritic cells to about 2.0 x 10 7 dendritic cells.
  • the cell vaccine includes between 1.3 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.4 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.5 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.6 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.7 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1.8 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells.
  • the cell vaccine includes between 1.9 x 10 7 dendritic cells to 2.0 x 10 7 dendritic cells. [0135] In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.9 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.8 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.7 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.6 x 10 7 dendritic cells.
  • the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.5 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.4 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.3 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.2 x 10 7 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 10 7 dendritic cells to 1.1 x 10 7 dendritic cells. [0136] In embodiments, the cell vaccine more preferably includes about 1.0 x 10 7 dendritic cells.
  • the cell vaccine includes about 1.0 x 10 7 dendritic cells. In embodiments, the cell vaccine more preferably includes 1.0 x 10 7 dendritic cells. In embodiments, the cell vaccine includes 1.0 x 10 7 dendritic cells.
  • the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder. In embodiments, the disease or disorder is cancer. In embodiments, the disease or disorder is an immune disease or disorder. In embodiments, the disease or disorder is an infectious disease or disorder. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer.
  • the cancer is breast cancer.
  • the cancer is ovarian cancer.
  • the cancer is colon cancer.
  • the cancer is prostate cancer.
  • the cancer is bone cancer. In embodiments, the cancer is colorectal cancer. In embodiments, the cancer is gastric cancer. In embodiments, the cancer is lymphoma. In embodiments, the cancer is malignant melanoma. In embodiments, the cancer is liver cancer. In embodiments, the cancer is small cell lung cancer. In embodiments, the cancer is non-small cell lung cancer. In embodiments, the cancer is pancreatic cancer. In embodiments, the cancer is thyroid cancers. In embodiments, the cancer is kidney cancer. In embodiments, the cancer is cancer of the bile duct. In embodiments, the cancer is brain cancer. In embodiments, the cancer is cervical cancer. In embodiments, the cancer is maxillary sinus cancer. In embodiments, the cancer is bladder cancer.
  • the cancer is esophageal cancer. In embodiments, the cancer is Hodgkin's disease. In embodiments, the cancer is adrenocortical cancer. [0139] In embodiments, further comprising one or more additional therapeutic agents.
  • the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents.
  • the one or more additional therapeutic agents is chemotherapy.
  • the one or more additional therapeutic agents is adjuvants.
  • the one or more additional therapeutic agents is radiation.
  • the one or more additional therapeutic agents is gene therapy. In embodiments, the one or more additional therapeutic agents is surgery.
  • the one or more additional therapeutic agents is one or more anti-inflammatory agents. In embodiments, the one or more additional therapeutic agents is one or more immunomodulatory agents. In embodiments, the one or more antigens is identified from the subject. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0140]
  • the cell vaccine is formulated to be administered to the subject directly after thawing. In embodiments, directly administering includes that dead cells are not removed from the composition prior to administration to the subject. In embodiments, the cell vaccine is formulated to be administered without washing or reconstitution. In embodiments, the cell vaccine is formulated to be administered without washing.
  • the cell vaccine is formulated to be administered without reconstitution. [0141] In embodiments, the cell vaccine is formulated for administration within about 4 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 3.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 3 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 2.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 2 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 1.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 1 hour of thawing.
  • the cell vaccine is formulated for administration within about 30 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within about 20 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within about 10 minutes of thawing. [0142] In embodiments, the cell vaccine is formulated for administration within 4 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 3.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 3 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 2.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 2 hours of thawing.
  • the cell vaccine is formulated for administration within 1.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within o1 hour of thawing. In embodiments, the cell vaccine is formulated for administration within 30 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within 20 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within 10 minutes of thawing.
  • the cell vaccine is formulated for administration within about 24 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 23 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 22 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 21 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 20 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 19 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 18 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 17 hours after thawing.
  • the cell vaccine is formulated for administration within about 16 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 15 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 14 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 13 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 12 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 11 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 10 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 9 hours after thawing.
  • the cell vaccine is formulated for administration within about 8 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 7 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 6 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 5 hours after thawing. [0145] In embodiments, the cell vaccine is formulated for administration within 24 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 23 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 22 hours after PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT thawing.
  • the cell vaccine is formulated for administration within 21 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 20 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 19 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 18 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 17 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 16 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 15 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 14 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 13 hours after thawing.
  • the cell vaccine is formulated for administration within 12 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 11 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 10 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 9 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 8 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 7 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 6 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 5 hours after thawing.
  • a method of manufacturing a cell vaccine for treating cancer including: (a) obtaining one or more mature dendritic cells (mDCs) according to the method provided herein including embodiments thereof; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer.
  • mDCs mature dendritic cells
  • the one or more mDCs are cryopreserved and thawed before step (b).
  • the one or more antigens is one or more tumor antigens.
  • the one or more tumor antigens includes a TAA.
  • the TAA PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT includes hTERT.
  • the TAA is hTERT.
  • the TAA includes LAMP1.
  • the TAA is a fusion protein.
  • the TAA is hTERT- LAMP1.
  • the TAA includes HSP96.
  • the TAA includes hTERT-LAMP1 and HSP96.
  • the one or more tumor antigens includes a TSA.
  • the one or more tumor antigens includes a neoantigen.
  • the one or more nucleic acid molecules includes one or more RNA molecules. In embodiments, the one or more RNA molecules include one or more mRNA molecules. In embodiments, the one or more nucleic acid molecules includes on ore more DNA molecules. In embodiments, the one or more RNA molecules includes hTERT mRNA. In embodiments, the one or more RNA molecules includes hTERT-LAMP1 mRNA. In embodiments, the one or more RNA molecules includes HSP96 mRNA. [0148] In embodiments, the one or more nucleic acid molecules is introduced into the cell via electroporation, transfection, transduction. In embodiments, the one or more nucleic acid molecules is introduced into the cell via electroporation.
  • the electroporation includes high throughput flow electroporation.
  • the one or more nucleic acid molecules is introduced into the cell via transfection.
  • the transfection includes chemical-based transfection.
  • the chemical-based transfection includes a lipid- based transfection agent.
  • the lipid-based transfection agent is a positively charged (e.g., cationic) lipid-based transfection agent.
  • the chemical-based transfection includes calcium phosphate (e.g., Ca 3 (PO 4 ) 2 ).
  • the one or more nucleic acid molecules is introduced into the cell via transduction.
  • the transduction includes a viral vector.
  • the viral vector is an Adenoviral vector, an Adeno-associated virus (AAV) vector, or a lentiviral vector.
  • the viral vector is an Adenoviral vector.
  • the viral vector is an Adeno-associated virus (AAV) vector.
  • the viral vector is a lentiviral vector.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCTX-CC-600PCT is between about 0.6 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.7 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.8 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.9 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 1 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 2 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 3 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 4 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 5 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 6 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 7 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 8 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 9 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 10 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 11 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 12 ⁇ g per million cells to about 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT molecules used for electroporation is between about 13 ⁇ g per million cells to about 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 14 ⁇ g per million cells to about 15 ⁇ g per million cells. [0150] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 14 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 13 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 12 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 11 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 10 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 9 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 8 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 7 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 6 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 5 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 4 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 3 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 2 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 1 ⁇ g per million cells. In embodiments, the concentration of the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 0.9 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 0.8 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 0.7 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 ⁇ g per million cells to about 0.6 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is preferably about 6 ⁇ g per million cells. [0151] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 0.6 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.7 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.8 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.9 ⁇ g per million cells to 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 1 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 2 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 3 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 4 ⁇ g per million cells to 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 5 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 6 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 7 ⁇ g per million cells PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT to 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 8 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 9 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 10 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 11 ⁇ g per million cells to 15 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 12 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 13 ⁇ g per million cells to 15 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 14 ⁇ g per million cells to 15 ⁇ g per million cells. [0152] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 14 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 13 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 12 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 11 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 10 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 9 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 8 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 7 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 6 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT molecules used for electroporation is between 0.5 ⁇ g per million cells to 5 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 4 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 3 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 2 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 1 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 0.9 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 0.8 ⁇ g per million cells.
  • the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 0.7 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 ⁇ g per million cells to 0.6 ⁇ g per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is preferably 6 ⁇ g per million cells. [0153] In embodiments, the manufacturing takes about 30 days to about 50 days. In embodiments, the manufacturing takes about 31 days to about 50 days. In embodiments, the manufacturing takes about 32 days to about 50 days. In embodiments, the manufacturing takes about 33 days to about 50 days.
  • the manufacturing takes about 34 days to about 50 days. In embodiments, the manufacturing takes about 35 days to about 50 days. In embodiments, the manufacturing takes about 36 days to about 50 days. In embodiments, the manufacturing takes about 37 days to about 50 days. In embodiments, the manufacturing takes about 38 days to about 50 days. In embodiments, the manufacturing takes about 29 days to about 50 days. In embodiments, the manufacturing takes about 40 days to about 50 days. In embodiments, the manufacturing takes about 41 days to about 50 days. In embodiments, the manufacturing takes about 42 days to about 50 days. In embodiments, the manufacturing takes about 43 days to about 50 days. In embodiments, the manufacturing takes about 44 days to about 50 days. In embodiments, the manufacturing takes about 45 days to about 50 days.
  • the manufacturing takes about 46 days to about 50 days. In embodiments, the manufacturing takes about 47 days to about 50 days. In embodiments, the manufacturing takes about 48 days to about 50 days. In embodiments, the manufacturing takes about 49 days to about 50 days. [0154] In embodiments, the manufacturing takes about 30 days to about 49 days. In embodiments, the manufacturing takes about 30 days to about 48 days. In embodiments, the manufacturing takes about 30 days to about 47 days. In embodiments, the manufacturing takes about 30 days to about 46 days. In embodiments, the manufacturing takes about 30 days to about 45 days. In embodiments, the manufacturing takes about 30 days to about 44 days.
  • the manufacturing takes about 30 days to about 43 days. In embodiments, the manufacturing takes about 30 days to about 42 days. In embodiments, the manufacturing takes about 30 days to about 41 days. In embodiments, the manufacturing takes about 30 days to about 40 days. In embodiments, the manufacturing takes about 30 days to about 39 days. In embodiments, the manufacturing takes about 30 days to about 38 days. In embodiments, the manufacturing takes about 30 days to about 37 days. In embodiments, the manufacturing takes about 30 days to about 36 days. In embodiments, the manufacturing takes about 30 days to about 35 days. In embodiments, the manufacturing takes about 30 days to about 34 days. In embodiments, the manufacturing takes about 30 days to about 33 days. In embodiments, the manufacturing takes about 30 days to about 33 days.
  • the manufacturing takes about 30 days to about 31 days. [0155] In embodiments, the manufacturing takes 30 days to 50 days. In embodiments, the manufacturing takes 31 days to 50 days. In embodiments, the manufacturing takes 32 days to 50 days. In embodiments, the manufacturing takes 33 days to 50 days. In embodiments, the manufacturing takes 34 days to 50 days. In embodiments, the manufacturing takes 35 days to 50 days. In embodiments, the manufacturing takes 36 days to 50 days. In embodiments, the manufacturing takes 37 days to 50 days. In embodiments, the manufacturing takes about 38 days to 50 days. In embodiments, the manufacturing takes 29 days to 50 days. In embodiments, the manufacturing takes 40 days to 50 days.
  • the manufacturing takes 41 days to 50 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT days.
  • the manufacturing takes 42 days to 50 days.
  • the manufacturing takes 43 days to 50 days.
  • the manufacturing takes 44 days to 50 days.
  • the manufacturing takes 45 days to 50 days.
  • the manufacturing takes 46 days to 50 days.
  • the manufacturing takes 47 days to 50 days.
  • the manufacturing takes about 48 days to about 50 days.
  • the manufacturing takes 49 days to 50 days. [0156]
  • the manufacturing takes 30 days to 49 days. In embodiments, the manufacturing takes 30 days to 48 days.
  • the manufacturing takes 30 days to 47 days. In embodiments, the manufacturing takes 30 days to 46 days. In embodiments, the manufacturing takes 30 days to 45 days. In embodiments, the manufacturing takes 30 days to 44 days. In embodiments, the manufacturing takes 30 days to 43 days. In embodiments, the manufacturing takes 30 days to 42 days. In embodiments, the manufacturing takes 30 days to 41 days. In embodiments, the manufacturing takes 30 days to 40 days. In embodiments, the manufacturing takes 30 days to 39 days. In embodiments, the manufacturing takes 30 days to 38 days. In embodiments, the manufacturing takes 30 days to 37 days. In embodiments, the manufacturing takes 30 days to 36 days. In embodiments, the manufacturing takes 30 days to 35 days. In embodiments, the manufacturing takes 30 days to 34 days.
  • the manufacturing takes 30 days to 33 days. In embodiments, the manufacturing takes 30 days to 33 days. In embodiments, the manufacturing takes 30 days to 31 days.
  • the one or more antigens is an antigen expressed by the cancer.
  • the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. In embodiments, the cancer is non-small cell lung cancer (NSCLC) . In embodiments, the cancer is prostate cancer. In embodiments, the cancer is acute myelogenous leukemia.
  • NSCLC non-small cell lung cancer
  • the cancer is prostate cancer.
  • the cancer is acute myelogenous leukemia.
  • a method for treating or delaying progression of a disease or disorder in a subject including administering to the subject a cell vaccine provided herein including embodiments thereof.
  • the dendritic cells are allogeneic to the subject.
  • a method for treating cancer including administering to a subject having cancer a cell vaccine made by the method provided herein including embodiments thereof.
  • the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia.
  • the cancer is non-small cell lung cancer (NSCLC).
  • the cancer is prostate cancer.
  • the cancer is acute myelogenous leukemia.
  • EXAMPLES [0161] LIST OF ABBREVIATIONS Abbreviation Definition A AE Adverse Event PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT o r or PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT
  • Example 1 [0162] LCT-VAC2 consists of mature dendritic cells (mDCs) derived from the H1 human embryonic stem cells (hESCs) line that have been transfected with mRNA coding for tumor- associated antigen hTERT/LAMP-1.
  • mDCs mature dendritic cells
  • hESCs human embryonic stem cells
  • LCT-VAC2 is a novel allogeneic dendritic cell (DC) vaccine derived from H1 undifferentiated human embryonic stem cells (hESCs).
  • LCT-VAC2 was produced by the stepwise differentiation of H1 hESCs into mature DCs (mDCs) which were then pulsed (electroporated) with messenger ribonucleic acid (mRNA) encoding the tumor associated antigen human telomerase reverse transcriptase (hTERT) fused to the cytoplasmic tail of lysosomal associated membrane protein 1 (LAMP-1), which facilitated human leukocyte antigen (HLA) II, as well as HLA I loading of hTERT. Both elements (hTERT and LAMP-1) were fused to the coding sequence for heat shock protein 96 (HSP96) to facilitate efficient shuttling of the translated protein to the endoplasmic reticulum.
  • HSP96 heat shock protein 96
  • the electroporated mDCs were then cryopreserved and irradiated to eliminate the replication of any remaining cells with proliferative capability.
  • the mDCs were engineered to present hTERT, a tumor-associated antigen found in most cancer cells, and LCT- VAC2 stimulated an immunogenic anti-tumor CD4+/CD8+ T cell response toward hTERT expressed on tumor cells.
  • LCT-VAC2 was developed to target the telomerase enzyme since telomerase is expressed in >85% of human cancers and the safety and efficacy of this approach have been previously demonstrated.
  • Telomerase is an established target to inhibit cancer growth since it is essential for maintaining the proliferative capacity and survival of tumor cells by preventing the shorting of the telomeres (Ouellette, Wright et al.2011). It is not usually expressed in normal tissues. Where it is observed (e.g., gastrointestinal tract and CD34+ hematopoietic cells), it is only transiently expressed at low levels.
  • the LCT-VAC2 manufacturing process is an optimized differentiation process originated from the H1 hESCs line into mDCs, which are then electroporated with hTERT mRNA fused to the cytoplasmic tail (LAMP-1). Improvement of the process used to generate the original VAC2 utilized in the CRUK trial included: a scaled-up process for H1 expansion, enhanced in-process controls (IPC) during the differentiation process, and development of specific in-process and release tests.
  • IPC enhanced in-process controls
  • a specific IPC plan to monitor cell characteristics along the differentiation process was created based on hESCs-mDCs differentiation steps: hESCs pluripotent cells, hematopoietic progenitors, common myeloid precursors (CMP), monocytes, immature DC (iDCs), and finally, mature DCs (mDCs).
  • CMP common myeloid precursors
  • iDCs immature DC
  • mDCs mature DCs
  • mDCs Mature DCs
  • LCT-mDCs were produced as part of LCT-VAC2 manufacturing from the hESCs by a differentiation process as an intermediate material. Characterization release tests of the intermediate material included evaluation of LCT-mDCs’ identity and functionality. Thus, LCT-mDCs were used as a source starting material in future applications by introduction of alternative mRNAs to induce different, specific immune responses.
  • Preclinical Development To support use of VAC-2 in the CRUK study, a series of in vitro characterization assays was completed to demonstrate that VAC-2 had the characteristics of, and behaved biologically as, activated mDCs.
  • LCT-VAC2 The Phase 2a clinical trial with LCT-VAC2 was designed to evaluate the safety and tolerability of escalating target doses of LCT-VAC2 utilizing four (4) dose groups.
  • LCT- VAC2 was given as six weekly intradermal (ID) injections in subjects with refractory Stage IIlB or IV NSCLC, who previously failed at least one approved immunotherapy regimen (e.g., PD-1 or PD-L1 checkpoint inhibitors or CTLA-4 receptor antibodies) and one platinum-based chemotherapy cycle, or a combination of both.
  • ID intradermal
  • LCT- VAC2 may address an unmet medical need via the use of defined HLA Class I and Class II potentiating activities.
  • Dendritic cells are naturally able to process and present antigens, activating both CD4+ and CD8+ T cell responses, and thus functionally promote an inflammatory response against specific antigens, including tumor antigens.
  • hESCs-derived mature DCs migrate from the site of administration to the regional lymph nodes and are capable of stimulating T cell proliferation in response to viral and other antigens including the tumor antigen hTERT. These cells have phagocytic activity and release pro- inflammatory cytokines.
  • hTERT The antigen (hTERT) is a known tumor antigen and cancer target, which has been well- characterized as being overexpressed almost exclusively in a variety of tumors, including NSCLC. To date, dozens of studies have either targeted hTERT directly, or have used hTERT expression levels as an endpoint to determine efficacy of treatment and there have been no unexpected safety events of note.
  • the therapeutic vaccine was administered via two, equally split intradermal (ID) injections at a target dose of 1 x 10 7 cells on each vaccination day, administered weekly for 6 consecutive weeks.
  • the target dose was 1 x 10 7 viable cells/day.
  • SAEs serious adverse events
  • Adverse events (AEs) reported at sites of VAC2 injection were mild, self-limiting and had no clinical sequelae. AEs reported by some of the subjects were suggestive of provocation or initiation of an adaptive immune response and included injection site PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCTX-CC-600PCT immunogenicity data in this study suggest the desired immune effects were generated in some VAC2 treated subjects. These data support further clinical investigation of VAC2.
  • Clinical Rationale [0191] Lung cancer is the leading cause of cancer mortality among men and women and accounts for about one-fifth of all cancer-related deaths (Spira and Ettinger 2004). Lung cancer is broadly split into non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), with 80-85% classified as NSCLC. Even with remarkable recent advances in treatment, long-term survival rates for NSCLC and SCLC remain poor and new treatments are necessary. LCT-VAC2 may provide an additional treatment option for these subjects.
  • NSCLC non-small cell lung cancer
  • SCLC small cell lung cancer
  • the hTERT sequence (amino acids 168-1132) used to create VAC2, both in the completed CRUK study and the proposed trial, contains epitopes that are known to be presented by HLA A*02:01 (Lin, Guarnieri et al.1996).
  • the HLA A*02:01 locus is expressed at high frequencies in the general population, including in approximately 46% of individuals of North European Caucasian ancestry (Morse, Coleman et al.1999, Eskelinen 2006).
  • H1 Cell Banking System [0195] H1 Master Cell Bank (MCB) Production [0196] The origin of the new H1 Master Cell Bank (MCB) was the H1 Bank Lot. No. MCBH101.
  • MCBH101 was manufactured by Geron directly from the H1 Original Cell Bank (OCB) in 2009 (manufacturing of Geron’s MCBH101 Seed Bank is detailed in GR10-014 report).
  • the production of H1 MCB (LCT-H1-001) was performed according to FIG.2 and involved the following steps - one vial of MCBH101 Passage 30 is thawed and seeded on rh- Laminin 521 (BioLamina, LN521) coated flasks in mTeSR plus (STEMCELL Technologies, 100- 0276).
  • the cells were passaged using non-enzymatic dissociation reagent ‘ReLeSR’ (STEMCELL Technologies, 05872).
  • the culture was expanded for 6 passages and evaluated for morphology, % confluency, and lactate concentration.
  • the MCB was cryopreserved in CryoStor 10 cryopreservation solution (CS10, Biolife solutions, 210102) at 10 x 10 6 cells per 1 mL per 1 vial, using CryoMed controlled rate freezer.
  • CS10 CryoStor 10 cryopreservation solution
  • Table 2 The characterization of Pilot LCT-H1-001 Master Cell Bank is summarized in Table 2. Table 2.
  • the culture was expanded for 4 passages and evaluated for morphology, % confluency, and lactate concentration.
  • the WCB was cryopreserved in CryoStor 10 cryopreservation solution (CS10, Biolife solutions, 2010102) at 7 x 10 6 cells per 1 mL per 1 vial, using CryoMed controlled rate freezer.
  • CryoStor 10 cryopreservation solution CS10, Biolife solutions, 2010102
  • CryoMed controlled rate freezer CryoMed controlled rate freezer.
  • LCT VAC2 and LCT mDCs LCTVAC2 and LCT-FnDCr Process Overview
  • LCT-VAC2 and LCT mDCs production process is described in FIG.4 and involved the following steps: hESCs thawing (WCB) and expansion prior to differentiation hESCs differentiation into monocytes and further into iDCs iDCs maturation into mDCs Harvesting mDCs The mDCs were then be processed by one of two options: o LCT-m
  • the cells were passaged using non-enzymatic dissociation reagent ‘ReLeSR’ (STEMCELL Technologies, 05872).
  • the culture was expanded for 2 passages (P30+6+4+3) and evaluated for morphology, % confluency, and lactate concentration.
  • the hESCs were seeded on enhanced attachment microcarriers (Corning, 3779) in mTeSR plus media for 4 days in dynamic 0.5 L PBS wheels vessels (PBS Biotech, FA-0.5-D-001) (P30+6+4+4).
  • the cells were PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • hESCs Differentiation into Monocytes and iDCs Differentiation [0212] On day 0, to start the differentiation process, the harvested hESCs were seeded for aggregate formation in X-VIVO15 medium (Lonza, BEBP02-061Q) supplemented with 50 ng/ml rh-GM-CSF (Cellgenix, 001012-1000), 20 ng/ml rh-SCF (R&D systems, 255B-GMP-01M), 50 ng/ml rh-VEGF165 (R&D systems, 293-GMP-01M), 50 ng/ml rh-BMP4 (R&D systems, 314E- GMP-050), and 10 ⁇ M of Y27632 (Tocris, 1254), from day 0 to day 3 of the differentiation.
  • the aggregates were formed in 0.5L PBS wheel dynamic vessels. At the end of aggregate formation (day 3), lactate was measured, and the medium was replaced with fresh X-VIVO15 supplemented with 50 ng/ml rh-GM-CSF, 20 ng/ml rh-SCF, 50 ng/ml rh-VEGF165, and 50 ng/ml rh-BMP4. Then, the aggregates were transferred into static culture according to the lactate concentration and seeded in ULA CellSTACKs (Corning, 3303) (Lactate concentration was measured in the PBS wheel and the relevant volume according to Lactate was transferred per CellSTACK).
  • the medium was replaced three times a week with X-VIVO15 supplemented with the following according to the relevant day of differentiation: Days 3-5 - 50 ng/ml rh-GM-CSF, 20 ng/ml rh-SCF, 50 ng/ml rh-VEGF165, and 50 ng/ml rh-BMP4 Days 6-10 - 50 ng/ml rh-GM-CSF, 20 ng/ml rh-SCF, and 50 ng/ml rh-VEGF165 Days 11-15 - 50 ng/ml rh-GM-CSF, and 20 ng/ml rh-SCF Days 15-27 - 50 ng/ml rh-GM-CSF [0214] Lactate concentration and culture morphology were evaluated during every media replacement.
  • the aggregates were removed from the culture using customized 60 ⁇ m closed system filtration system (Meissner) and the single cells were cultured in CellGenix® GMP DC Medium (CG-DC, CellGenix, 20901-0500) supplemented with 50 ng/ml rh-GM-CSF and 50 ng/ml rh-IL-4 (R&D systems, 204-GMP-050) for 5 days in the CellSTACK.
  • the medium was replaced on day 31 with fresh CG-DC media supplemented with 50 ng/ml rh- GM-CSF and 50 ng/ml rh-IL-4.
  • lactate concentration measurement and morphology evaluation were performed.
  • Harvested mDCs can be further processed to LCT-VAC2 by electroporation, cryopreservation, and X-Ray irradiation to yield the final LCT-VAC2.
  • the harvested mDCs can be cryopreserved and processed to the final product as a separate stage.
  • LCT-mDCs - Cryopreserved non-EP mDCs [0222] mDCs were cryopreserved at 25 x 10 6 cells per 1 ml per vial in CryoStor 5 cryopreservation solution (CS5, Biolife solutions, 205102) - LCT-mDCs intermediate product. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCTX-CC-600PCT LCTX-CC-600PCT
  • LCT-mDCs were thawed and electroporated with desired mRNA into the final product. The characterization of LCT-mDCs is detailed below.
  • Electroporation of LCT-mDCs with hTERT-LAMP1 mRNA [0226] For LCT-VAC2 mDCs, following harvest on day 35, cells in the quenching solution (1:3, 1mM EDTA quenched by CG-DC medium), as detailed above, were counted, diluted (1:10, v:v) with PBS minus (Sartorius, 02-023-1A) and centrifuged.
  • the cells were resuspended and pooled at a cell concentration of 250 x 10 6 per mL Belzer UW Solution (Bridge to Life, BUWC).
  • the cell suspension was adjusted to 100 x 10 6 cells per mL by adding Belzer UW solution, after which the cell suspension was divided into 0.3-1 x 10 9 aliquots each assigned for consecutive EP cycles, and kept at 2-8 °C.
  • GMP grade hTERT-LAMP1 mRNA was added to the cell suspension in Belzer UW, to reach a final cell/mRNA mixture of 50 x 10 6 cells with 300 ⁇ g mRNA per mL.
  • Cell/mRNA mixture was then transferred into the 4D Nucleofector inlet reservoir (Lonza, V4LR-1001W) via a Luer port.
  • the inlet reservoir was placed on a stirrer at 300 RPM, keeping the mixture homogenous during EP.
  • the EP cycles were performed using the 4D Nucleofector LV unit (Lonza) under vendor’s optimal electroporation program for introducing mRNA into human mature dendritic cells (CB150).
  • Cell/mRNA mixture was pumped from the reservoir, through the LV cartridge (Lonza, V4LN-7020), electroporated, and finally transferred into the CG-DC medium - filled outlet bag, at fixed increments of 1 mL.
  • Post EP the cells were allowed to sit for 10 minutes at RT to recover from the EP process, at a cell concentration of 1 x 10 6 cells per mL.
  • Post-EP cells were allowed to recover and are then filtered using a customized 60 ⁇ m closed-system filtration kit (Meissner), and then were counted using the NC-200 cell counter (DS counts). The cells were centrifuged and re-suspended in CryoStor 5 cryopreservation solution (CS5, Biolife solutions, 205102,).
  • the cells were counted directly in CS5 while CS5 wasa PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT incrementally added followed by additional cell count until the required final cell concentration was achieved, prior to cryopreservation. After confirmation of target cell concentration, the cells were vialed at 6.25 x 10 6 cells per 500 ⁇ l per vial (Nunc Cryovial, Thermo-Fisher Scientific, 374086). The vials were then cryopreserved using the CryoMed controlled rate freezer.
  • cryopreserved electroporated mDCs were X-ray irradiated, to eliminate cell proliferation capabilities. Irradiation was carried out using Precision Xray’s XRAD 320 irradiation system. [0232] Determination of the irradiation dose parameters was performed with pure cryopreserved H1 hESCs, serving as a ‘worst case’ scenario to determine the irradiation dose parameters that are required to achieve mitotic arrest of these highly proliferating cells.
  • harvested single-cell H1 hESCs were cryopreserved using CS10 at 6.25 x 10 6 cells per 500 ⁇ l in NUNC cryovials (Thermo-Fisher Scientific, 374086), simulating the LCT-VAC2 final product vial type and cell concentration.
  • Post irradiation evaluation of the proliferation capacity was done via a performance test, in which irradiated and non-irradiated H1 hESCs vials are thawed and cultured, for a 4-day culturing session, during which cell proliferation is evaluated by lactate measurements and morphological assessment from day 1 to 4.
  • Lactate served as a direct indicator to the number of H1 hESCs in the culture, and the daily lactate accumulation as an indication of H1 hESCs proliferation capacity. Both lactate concentration and morphological appearance of irradiated hESCs are markedly distinguished from those of non-irradiated ESCs culture, as can be seen in Table 7, on day 4 post thawing non irradiated H1 cells reach above than 10mM lactate compared to 0 mM lactate of the irradiated H1 cells.
  • the irradiation parameters that were selected and shown to be effective in arresting proliferation were: Dose: 1200 rad (12Gy), 130 kV, 12.5 mA, SSD (‘Source-to-Sample Distance’): 45/5 cm. Irradiation cycle time is in the range of approx.730-750 seconds. This dose was confirmed as having no effect on other attributes of LCT-VAC2 (viability, recovery, identity, and functional activity). [0233] Irradiation dose verification was done prior to each irradiation cycle by conducting a dry simulation (without vials) using a calibrated dose meter to verify the system’s operation. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • hESCs-mDCs differentiation was based on the following hematopoietic cell types: hematopoietic progenitors, common myeloid precursors (CMP), monocytes, immature DCs (iDCs) and finally mDCs.
  • CMP common myeloid precursors
  • iDCs immature DCs
  • FIG.6 A detailed IPC plan according to the different differentiation steps is depicted in FIG.6. A list of markers tested at IPC sample points are listed in Table 8 below.
  • CD34 is a transmembrane phosphor-glycoprotein expressed on hematopoietic stem cells and progenitor cells which is tested on day 6 (Patente, Pinho et al.2018). Generally, sufficient fraction of CD34+ cells among the differentiating cells is needed to make the differentiation process robust.
  • CD90 is a known progenitor marker expressed by pluripotent cells including hESCs. As the differentiation process progresses CD90 expression was expected to be downregulated. ii.
  • CD45 leukocyte common antigen
  • IPC3-day 20 CD45 (leukocyte common antigen) is a unique and ubiquitous membrane glycoprotein that is expressed on all hematopoietic cells except for mature erythrocytes (Nakano, Harada et al.1990). CD45 expression was known to be elevated around day 20 of differentiation process while cells were still grown in aggregates, indicating cellular differentiation towards leukocyte lineage.
  • IPC4-Day 27 Enables evaluation of cells before filtration of aggregates on day 28 of the differentiation process.
  • CD45 expression is expected to increase compared to expression on day 20 as most of the single cells are leucocytes.
  • CD14 expression was tested to ensure cells undergo monocytic step as part of their differentiation (Landmann, Muller et al.2000).
  • CD86 a DC marker, was tested as well as part of cellular differentiation towards DCs (Mir 2015).
  • iv. 1PC5-Day 32 Time point for evaluation of cells post aggregate filtration and prior to maturation toward mDCs to ensure that CD86+ expression continues to rise compared to day 27 and that high CD45+ expression is maintained.
  • CD83 is expressed by mDCs (Zhou, Schwarting et al.1992) and was tested as part of kinetics profile, and CD83 expression increased until the end of the process on day 35, indicating that cells acquired the desired mDCs fate.
  • LCTVAC2 Final Drug Product Characterization [0240] LCT-VAC2 was subjected to a panel of characterization assays developed to evaluate cells’ identity and functionality. These final product characterizations included cells’ viability, identity, and functional activity, and are summarized in Table 9 below. LCT-VAC2 cryopreserved vials were thawed and evaluated according to established assay protocols. Table 9. LCT-VAC2 Final Product Characterization Quality attribute Method Viability (% Viable cells) NC-200 PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCT-mDCs Characterization LCT-mDCs was subjected to a panel of characterization assays developed to evaluate cells’ identity and functionality. These characterizations are summarized in Table 10 below. LCT- mDCs cryopreserved vials were thawed and evaluated according to established assay protocols. Table 10. LCT-mDCs Intermediate Product Characterization Quality attribute Method % CD45+ Leukocyte marker PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • CD45 antigen leukocyte common antigen
  • mDCs were expected to express it at high levels at the end of differentiation process.
  • CD83 is an identity marker for mDCs at the end of mDCs differentiation process.
  • iii. CD86 is a co-receptor expressed on antigen presenting cells in general and DCs, in particular (Mir 2015).
  • iv. HLA-A is a common isotype of MHC-I molecule. It was used to evaluate MHC class I molecule that is expressed on all nucleated cells and it’s involved in antigen presenting processes (Bray 2001).
  • v. HLA-DR is a common isotype of MHC-II molecule. It is expressed on antigen presenting cells (Neefjes, Jongsma et al.2011).
  • CD40 is a co-stimulatory protein found on antigen-presenting cells and is required for their activation. Binding of CD154 (CD40-L) leads to a cascade of downstream effects result in cell activation (Grewal and Flavell 1998).
  • mDCs cells express it at high levels upon maturation.
  • CCR7 is a chemokine receptor which serves to direct DCs to lymph nodes (Dieu, Vanbervliet et al.1998). Its expression is known to be significantly elevated upon cell maturation to mature DCs.
  • Non-Targeted Cell Population Evaluation of non-targeted cell population was also performed by flow cytometry, a quantitative method capable of detecting and identifying cellular markers on a single-cell basis. This assay was based on the assessment of cells expressing a selected panel of mDCs characteristic markers as listed in Table 10 and specified below: PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT i.
  • CD14 is a monocyte/macrophage differentiation antigen presented on the surface of cells of myeloid lineage (Landmann, Muller et al.2000).
  • CD14 was tested as part of final product characterization to examine the final product for non-targeted cell populations with monocyte characteristics. ii. CD45(+) and CD83(-) staining of cells was used to determine percentage of non- DC cells at the final product.
  • Residual hESCs [0251] LCT-VAC2/LCT-mDCs was manufactured by differentiating hESCs to mDCs cells. The level of residual hESCs in the final product was measured using two-dimensional flow cytometry analysis for two markers of undifferentiated hESCs: TRA-1-60 and SSEA-5.
  • LCT-VAC2 cells mode of action in the patient was based on the premise that these matured DC cells EP with hTERT antigen can migrate to adjacent lymph node and successfully activate T cells by presenting the hTERT antigen.
  • a set of functional activity assays was designed for characterization of LCT-VAC2 final product. These tests were based on mimicking the biological activity of LCT-VAC2 cells in the patient’s immune system and lymph nodes. i. using in-vitro Transwell system 6.5 mm with 8.0 ⁇ m Pore Polycarbonate Membrane Insert, Corning, #3422) based assay. Briefly, DC media supplemented cells were added to the top chambers.
  • the number of cells that were migrated to the lower chamber was determined by counting in a NC-200 cell counter 4 hours post incubation. Percentage of migrated cells was calculated based on dividing the number of migrated cells by the number of cells initially cultured.
  • Mixed Leukocyte Reaction assay (MLR) The ability of LCT-VAC2 cells to promote allogeneic T cell proliferation was assessed using Mixed Leukocyte Reaction assay (MLR). Proliferation of fluorescently pre-labeled peripheral blood mononuclear cells (PBMC) following co-culture with LCT-VAC2 was measured using flow cytometry.
  • hTERT Antigen presentation assay (for LCT-VAC2 only): The ability of LCT- VAC2 cells to present and activate hTERT specific T cells was evaluated by co- incubation of LCT-VAC 2 cells with hTERT specific T cells generated using co culture of HLA-A-02:01 PBMC derived mDCs differentiated from isolated monocytes with autologous T cells in the presence of hTERT peptide (Miltenyi following co culture of cells with allogeneic mDCs in the presence of hTERT peptide (Miltenyi Biotec #130-097-277).
  • CMV Antigen presentation was used to assess the potential of mDCs to activate T cells via general antigen presentation via peptide uptake.
  • T cells co-cultured with LCT-mDCs at a ratio of 1:10 (Effector:Target), with 1 ⁇ g of pp65 peptide (PepTivatorCMV pp65, Miltenyi Biotec, #130-093-438) in 24-well plate (Corning, #3524) for 20-24 hours.100 IU/ml IL-2 (Miltenyi Biotec, #130-097-742) was added to the media.
  • T cells co- cultured with mDCs but without peptide, or T cells alone were used as negative PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • LCTX-CC-600PCT controls for A INFy secretion calculation.
  • INFy Interferon gamma
  • secretion into culture media was evaluated using a commercially available ELISA assay (R&D Systems,# DIF50C). To evaluate the extent of antigen specific T cell activation, A secretion by negative control group without peptide.
  • v. Expression of hTERT-LAMP-1 mRNA in the final LCT-VAC 2 product was confirmed by Western blot (protein) and RT-PCR (mRNA presence) methods.
  • qPCR was performed to confirm the presence of hTERT-LAMP-1 mRNA in the final product after electroporation. Briefly, mRNA was extracted from cells using a Qiagen RNeasy Mini kit (Qiagen # 74106) and transcribed into cDNA. cDNA amplification was performed with hTERT-LAMP-1 construct specific primers (Tagman). Assay was performed on thawed LCT-VAC2 final product. Non-electroporated cells are used as a negative control. [0254] Materials Used in Manufacturing of LCT-VAC2
  • LCTX-CC-600PCT g n n Stability of LCT-VAC2 Drug Product
  • the LCT-VAC2 drug product is planned to undergo a 5-year stability program. Each produced batch will be tested at 6- and 12-months post-manufacturing and annually for an additional 4 years thereafter.
  • Shipment, Formulation and Administration [0258] LCT-VAC2 is cryopreserved as a TAI product in CS5 and maintained in the vapor phase in LN2. It will be shipped to clinical sites under the same conditions and will be thawed immediately prior to administration at the clinical site.
  • LCT-VAC2 has an immunologically complex mechanism of action. Consequently, there are no relevant animal models in which to evaluate the safety of LCT-VAC2.
  • PBMC peripheral blood mononuclear cells
  • CD34+ hematopoietic progenitor cells PBMC transfer leads to graft versus host disease (GVHD) in the humanized mice in 3-6 weeks, and thus would complicate any safety or efficacy signal observed from a series of LCT-VAC2 injections.
  • GVHD graft versus host disease
  • human T cells that arise from CD34+ transfer are not educated by a human thymus.
  • hESCs-derived DCs exhibit a phenotype comparable with immature and of stimulating T cell proliferation in response to viral antigens and the tumor antigen hTERT. These cells have phagocytic activity and release pro-inflammatory cytokines.
  • Cell surface markers on mature hESCs-derived DCs include CD83 and CD86 HLA I and II as well as CCR7. All these features are common attributes of mature DCs and suggest that hESCs-derived DCs are functionally and phenotypically comparable with endogenous DCs.
  • hESCs-derived DCs transfected with hTERT/LAMP-1 mRNA were reactive towards hTERT-loaded PBMCs obtained PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT from HLA-A2+ individuals as assessed by ELISPOT analysis in vitro.
  • LCT-VAC2 cells transfected with hTERT/LAMP-1 mRNA were capable of priming na ⁇ ve T cells isolated from peripheral blood matched on 1, 2, or 4 HLA alleles.
  • LCT-VAC2 has been shown to be capable of stimulating both memory and na ⁇ ve semi-allogeneic T cells in an hTERT-dependent manner.
  • Safety pharmacodynamic as well as pharmacodynamic interaction studies have not been undertaken with LCT-VAC2. This is partly due to the absence of a relevant non-clinical model but is also principally due to existing clinical data already demonstrating that hTERT/LAMP-1 mRNA transfected into autologous DCs (VAC1) does not demonstrate any evidence of secondary or adverse safety pharmacology (Khoury, Collins et al.2010, Khoury, Collins et al.2017).
  • LCT-VAC2 The potential for LCT-VAC2 to undergo classical pharmacokinetic interactions with other drugs is unlikely. Accordingly, no studies of the pharmacokinetic interactions of LCT-VAC2 have been performed.
  • Toxicology Studies [0267] In the absence of suitable animal models, no animal toxicology studies have been undertaken. However, LCT-VAC2 has been shown to be phenotypically equivalent to autologous DCs which have previously demonstrated tolerability in numerous clinical trials involving many hundreds of subjects (Brunsvig, Kyte et al.2011, Dorrie, Schaft et al.2020).
  • LCT-VAC2 differs from autologous DC therapy in one notable respect in that subjects treated with LCT-VAC2 are only partially haplotype matched to the potential subject. There have been no indications of unexpected SAEs or AEs of increased severity in the completed VAC2 CRUK study and no GVHD or cytokine release syndrome following administration of VAC2 or other allogeneic DC vaccines trials in both solid and hematological malignancies at similar doses as those proposed for the US study in NSCLC. [0268] LCT-VAC2 has been manufactured to produce terminally differentiated DCs, which have limited proliferative potential, minimizing a theoretical risk of ectopic growth or tumorigenicity.
  • LCT-VAC2 presents hTERT epitopes on HLA I expressed upon their cell surfaces (and, by inference, hTERT expressed on HLA II as well since LAMP-1 epitopes are expressed on both HLA I and II). Accordingly, LCT-VAC2 would be expected to elicit an immune response towards cells that express hTERT and/or LAMP- 1. In adults, hTERT expression is restricted to the testes and gastrointestinal tract and LAMP-1 predominantly to exhausted T cells.
  • each subject will initially receive a maximum of six immunizations (in Weeks 1, 2, 3, 4, 5 and 6) based on previous clinical experience with both the autologous VAC1 and the allogeneic VAC2, where six immunizations were found to induce the highest levels of anti-hTERT immune responses.
  • the product has been shown to be a clinically well-tolerated experimental treatment for cancer. Acceptable safety events reported to date include mild, self-limiting, localized injections site reactions (ISR) as well as fatigue, mild-fever, chills etc.
  • ISR localized injections site reactions
  • the partial HLA mismatch of LCT-VAC2 to subjects may contribute to any localized rejection reaction over and above what might be seen with an autologous product.
  • CLINICAL DEVELOPMENT [0273] Overview of Clinical Development Plan [0274] Phase 1 Study [0275] A Phase 1 trial (CRUKD/17/003) of VAC2 (AST VAC2) was conducted at two centers in the UK by Cancer Research UK administered the vaccines weekly via intradermal injection in subjects with advanced NSCLC. The study has completed enrollment in February 2022 and all subjects have either withdrawn or died due to the progressive and advanced stage of their disease.In this study, two intradermal injections with a total target dose of 1 x 10 7 cells of VAC2 were administered at six time points over a period of six consecutive weeks.
  • TG4010 immunotherapy and first-line chemotherapy for advanced non-small-cell lung cancer results from the phase 2b part of a randomised, double-blind, placebo-controlled, phase 2b/3 trial.” Lancet Oncol 17(2): 212-223. [0301] 24. Quoix, E., et al. (2011). "Therapeutic vaccination with TG4010 and first-line chemotherapy in advanced non-small-cell lung cancer: a controlled phase 2B trial.” Lancet Oncol 12(12): 1125-1133. [0302] 25. Reck, M., et al. (2013).
  • ECM extracellular matrix
  • P Embodiment 2. The method of P Embodiment 1, wherein the culturing in step (b) is dynamic.
  • P Embodiment 3. The method of P Embodiment 1, wherein the culturing in step (b) is static.
  • the differentiation cocktail comprises at least one exogenous cytokine
  • the at least one exogenous cytokine comprises granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3).
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • BMP-4 bond morphogenic protein 4
  • VEGF vascular endothelial growth factor
  • SCF stem cell factor
  • FLT3L fetal liver kinase ligand
  • TPO thrombopoietin
  • IL-3 interleukin 3
  • P Embodiment 7 wherein the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days.
  • P Embodiment 9 The method of P Embodiment 7 or 8, wherein the static culturing is for about 3 days to about 27 days.
  • P Embodiment 10 The method of any one of P Embodiments 1-9, wherein the maturation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous [0319] P Embodiment 11.
  • step (e) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express CD14, CD45, TRA160, and SSEA-5.
  • step (e) comprises static culturing of the population of iDCs.
  • step (e) comprises static culturing of the population of iDCs.
  • P Embodiment 13 The method of P Embodiment 12, wherein the static culturing is for about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days.
  • P Embodiment 14 The method of any one of P Embodiments 1-13, wherein any one of or both of steps (d) and (e) are performed under serum free conditions.
  • P Embodiment 15 The method of any one of P Embodiments 1-13, wherein any one of or both of steps (d) and (e) are performed in the absence of feeder cells.
  • P Embodiment 16 The method of any one of P Embodiments 1-13, wherein any one of or both of steps (d) and (e) are performed in the absence of stromal cells.
  • P Embodiment 21 The method of P Embodiment 20, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60.
  • P Embodiment 22 The method of P Embodiment 20, wherein at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog.
  • P Embodiment 23 The method of P Embodiment 20, wherein at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog.
  • P Embodiment 20 wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60, at least about 70% of the undifferentiated and pluripotent cells express Oct-4, and at least about 70% of the undifferentiated and pluripotent cells express Nanog.
  • step (b) further comprises changing the growth medium to remove accumulated lactate.
  • P Embodiment 26 The method of any one of P Embodiments 17-25, wherein the culturing in step (b) is dynamic.
  • P Embodiment 27 The method of any one of P Embodiments 1-26, wherein the cultured cells in step (b) comprise hematopoietic cell intermediates.
  • P Embodiment 28 The method of P Embodiment 27, wherein the hematopoietic cell intermediates express CD34, CD90.
  • P Embodiment 29 The method of any one of P Embodiments 1-24, wherein the growth medium comprises Y-27632.
  • P Embodiment 27 or 28 wherein the hematopoietic cell intermediates do not express CD38.
  • P Embodiment 30 The method of P Embodiment 27, wherein the hematopoietic cell intermediates express CD45.
  • P Embodiment 31 The method of P Embodiment 30, wherein the hematopoietic cell intermediates do not express CD38.
  • P Embodiment 32 The method of P Embodiment 27 or 28, wherein the hematopoietic cell intermediates do not express CD38.
  • the microcarrier comprises one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof.
  • P Embodiment 33 The method of any one of P Embodiments 1-32, the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof.
  • P Embodiment 34 The method of any one of P Embodiments 1-33, wherein the microcarrier is not coated.
  • P Embodiment 35 The method of any one of P Embodiments 1-33, wherein the microcarrier is not coated.
  • P Embodiment 36 The method of any one of P Embodiments 1-35, wherein the microcarrier has a neutral charge.
  • P Embodiment 37 The method of any one of P Embodiments 1-36, wherein the microcarrier is negatively charged.
  • P Embodiment 38 The method of any one of P Embodiments 1-37, wherein the microcarrier is hydrophilic. [0347] P Embodiment 39.
  • P Embodiment 40 The method of P Embodiment 39, wherein the laminin is human laminin.
  • P Embodiment 41 The method of P Embodiment 40, wherein the human laminin is 511 E8 fragment.
  • P Embodiment 42 The method of P Embodiment 42.
  • P Embodiment 44 The method of P Embodiment 38, any one of P Embodiments 1-37, further comprising (f) cryopreserving the population of mDCs.
  • P Embodiment 44a The method of P Embodiment 38, any one of P Embodiments 1-43, further comprising (f) cryopreserving the population of mDCs.
  • P Embodiment 44 further comprising (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen.
  • P Embodiment 46 The method of P Embodiment 43 or 45, wherein the population of mDCs is contacted with two or more antigens.
  • P Embodiment 47 The method of any one of P Embodiments 43-45, wherein the antigen is a tumor antigen.
  • P Embodiment 48 The method of P Embodiment 46, wherein the tumor antigen comprises a tumor associated antigen (TAA).
  • TAA tumor associated antigen
  • P Embodiment 47 wherein the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof.
  • P Embodiment 50 The method of P Embodiment 46, wherein the tumor antigen comprises a tumor-specific antigen (TSA).
  • TSA tumor-specific antigen
  • P Embodiment 51 The method of P Embodiment 46, wherein the tumor antigen comprises a neoantigen.
  • P Embodiment 52 The method of P Embodiment 43 or 45, wherein the nucleic acid comprises an RNA.
  • P Embodiment 53 The method of P Embodiment 52, wherein the RNA comprises an mRNA encoding hTERT or fragment thereof.
  • P Embodiment 54 The method of P Embodiment 54, the wherein the fragment comprises one or more immunogenic epitopes of hTERT.
  • P Embodiment 55 The method of P Embodiment 44 or 45, wherein the population of mDCs comprising the one or more antigens is cryopreserved.
  • P Embodiment 56 The method of P Embodiment 54, wherein the cryopreserved population of mDCs comprising the one or more antigens is irradiated.
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0366] P Embodiment 57.
  • P Embodiment 58. A cell produced by any one of P Embodiments 43-55, wherein the cell (i) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express CD14, CD83, TRA160, and SSEA-5; and (ii) comprises the one or more antigens.
  • P Embodiment 59 A cell produced by any one of P Embodiments 1-41, wherein the cell expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express CD14, CD83, TRA160, and SSEA-5; and (ii) comprises the one or
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject comprising:(a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject comprising: (a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) comprising a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier.
  • P Embodiment 61 The cell vaccine of P Embodiment 60, wherein the nucleic acid comprises a DNA.
  • P Embodiment 62 The cell vaccine of P Embodiment 60, wherein the nucleic acid comprises a DNA.
  • P Embodiment 61 The cell vaccine of P Embodiment 61, wherein the DNA is under the control of a promoter.
  • P Embodiment 63 The cell vaccine of P Embodiment 62, wherein the promoter is an inducible promoter.
  • P Embodiment 64 The cell vaccine of P Embodiment 63, further comprising (c) an inducing agent.
  • P Embodiment 65 The cell vaccine of P Embodiment 64, wherein the inducing agent is desferrioxamine.
  • P Embodiment 66 The cell vaccine of P Embodiment 64, wherein the inducing agent is desferrioxamine.
  • P Embodiment 60 wherein the nucleic acid comprises an RNA.
  • P Embodiment 67 The cell vaccine of any one of P Embodiments 60-66, wherein the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent.
  • P Embodiment 68 The cell vaccine of any one of P Embodiments 60-67, wherein the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder.
  • P Embodiment 69 The cell vaccine of any one of P Embodiments 60-67, wherein the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder.
  • the cell vaccine of P Embodiment 68 wherein the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer.
  • P Embodiment 70 The cell vaccine of any one of P Embodiments 60-69, further comprising one or more additional therapeutic agents.
  • P Embodiment 71 The cell vaccine of any one of P Embodiments 60-69, further comprising one or more additional therapeutic agents.
  • the cell vaccine of P Embodiment 70 wherein the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents.
  • P Embodiment 72 The cell vaccine of any one of P Embodiments 60-71, wherein the one or more antigens is identified from the subject.
  • P Embodiment 73 The cell vaccine of any one of P Embodiments 60-71, wherein the one or more antigens is identified from the subject.
  • a method of manufacturing a cell vaccine for treating cancer comprising: (a) obtaining one or more mature dendritic cells (mDCs) according to the method of any one of P Embodiments 1-42; and (b) contacting the one or more mDCs with one or PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer.
  • P Embodiment 74 The method of P Embodiment 73, further comprising (c) providing one or more additional therapeutic agents.
  • P Embodiment 75 The method of P Embodiment 73, further comprising (c) providing one or more additional therapeutic agents.
  • P Embodiment 73 wherein optionally the one or more mDCs are cryopreserved and thawed before step (b).
  • P Embodiment 76 The method of any one of P Embodiments 73-75, wherein the one or more antigens is one or more tumor antigens.
  • P Embodiment 77 The method of P Embodiment 76, wherein the one or more tumor antigens comprises a TAA.
  • P Embodiment 78 The method of P Embodiment 77, wherein the TAA is hTERT.
  • P Embodiment 79 The method of P Embodiment 77, wherein the TAA is hTERT.
  • P Embodiment 76 wherein the one or more tumor antigens comprises a TSA.
  • P Embodiment 80 The method of P Embodiment 76, wherein the one or more tumor antigens comprises a neoantigen.
  • P Embodiment 81 The method of any one of P Embodiments 73-80, wherein the one or more nucleic acid molecules comprises one or more RNA molecules.
  • P Embodiment 83 The method of P Embodiment 82, wherein the one or more RNA molecules comprises hTERT mRNA.
  • P Embodiment 85 A method for treating or delaying progression of a disease or disorder in a subject, comprising administering to the subject a cell vaccine of any one of P Embodiments 59 to 72. [0395] P Embodiment 86.
  • Embodiment 1 A method of differentiating human embryonic stem cells (hESCs) into mature dendritic cells (mDCs), the method comprising: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce a cultured suspendable expansion complex comprising cultured cells; (c) removing the cultured cells from the cultured suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs
  • Embodiment 2 The method of embodiment 1, wherein the culturing in step (b) is dynamic.
  • Embodiment 3. The method of embodiment 1, wherein the culturing in step (b) is static.
  • Embodiment 4. The method of any one of embodiments 1-3, wherein the removing of step (c) comprises an enzyme-free dissociation reagent.
  • Embodiment 5. The method of any one of embodiments 1-4, wherein the removing of step (c) does not comprise manual dissection or manual scraping.
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0401] Embodiment 6.
  • Embodiment 7 The method of any one of embodiments 1-6, wherein the differentiation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous cytokine comprises granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3).
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • BMP-4 bond morphogenic protein 4
  • VEGF vascular endothelial growth factor
  • SCF stem cell factor
  • FLT3L fetal liver kinase ligand
  • TPO thrombopoietin
  • IL-3 interleukin 3
  • Embodiment 9 The method of any one of embodiments 1-7, wherein the differentiation cocktail comprises GM-CSF.
  • Embodiment 9. The method of any one of embodiments 1-8, wherein the differentiation cocktail comprises GM-CSF and SCF.
  • Embodiment 10. The method of any one of embodiments 1-9, wherein the differentiation cocktail comprises GM-CSF, SCF, and VEGF.
  • Embodiment 11. The method of any one of embodiments 1-10, wherein the differentiation cocktail comprises GM-CSF, SCF, VEGF, and BMP-4.
  • Embodiment 12 The method of any one of embodiments 1-11, wherein an iDC of the population of iDCs in step (d) expresses CD45 and CD86.
  • Embodiment 13 The method of any one of embodiments 1-12, wherein an iDC of the population of iDCs in step (d) does not express CD38.
  • Embodiment 14 The method of any one of embodiments 1-13, wherein the second period of time is between about 1 day to about 32 days.
  • Embodiment 15 The method of any one of embodiments 1-14, wherein step (d) comprises dynamic culturing of the cultured cells followed by static culturing of the cultured cells.
  • Embodiment 16 The method of embodiment 15, wherein the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days.
  • Embodiment 17 The method of embodiment 15 or 16, wherein the static culturing is for about 3 days to about 27 days.
  • Embodiment 18 The method of any one of embodiments 1-17, wherein the maturation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous
  • Embodiment 19 The method of any one of embodiments 1-18, wherein the maturation cocktail comprises GM-CSF and IL-4.
  • Embodiment 20 The method of any one of embodiments 1-19, wherein the [0416] Embodiment 21.
  • Embodiment 22 The method of embodiment 1-21, wherein an mDC of the population of mDCs in step (e) co-expresses CD45 and CD83.
  • Embodiment 23 The method of any one of embodiments 1-22, wherein at least 70% of the population of mDCs in step (e) express CD45.
  • Embodiment 24 The method of any one of embodiments 1-23, wherein at least 70% of the population of mDCs in step (e) express CD83.
  • Embodiment 25 The method of any one of embodiments 1-24, wherein at least 70% of the population of mDCs in step (e) express CD86, CD40, or HLA-A.
  • Embodiment 26 The method of any one of embodiments 1-25, wherein at least 20% of the population of mDCs in step (e) express HLA-DR.
  • PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0422] Embodiment 27.
  • Embodiment 28 The method of any one of embodiments 1-27, wherein less than 20% of the population of mDCs in step (e) are CD45+ and CD83-.
  • Embodiment 29 The method of any one of embodiments 1-22, wherein the third period of time is between about 1 day to about 10 days.
  • Embodiment 30 The method of any one of embodiments 1 to 29, wherein step (e) comprises static culturing of the population of iDCs.
  • Embodiment 31 Embodiment 31.
  • Embodiment 32 The method of any one of embodiments 1-31, wherein any one of or both of steps (d) and (e) are performed under serum free conditions.
  • Embodiment 34 The method of any one of embodiments 1-31, wherein any one of or both of steps (d) and (e) are performed in the absence of stromal cells.
  • Embodiment 35 The method of any one of embodiments 1-31, wherein any one of or both of steps (d) and (e) are performed in the absence of stromal cells.
  • Embodiment 36 The method of embodiment 35, wherein steps (a), (b) and (c) are each repeated three times prior to step (d).
  • Embodiment 37 The method of any one of embodiments 1-36, wherein steps (a), (b) and (c) combined are about three days to about five days.
  • Embodiment 38 The method of any one of embodiments 1-37, wherein the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in steps (a) through (c).
  • Embodiment 39 The method of embodiment 38, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60.
  • Embodiment 40 The method of embodiment 38, wherein at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog. [0436] Embodiment 41.
  • Embodiment 42 The method of any one of embodiments 1-41, wherein step (b) further comprises changing the growth medium to remove accumulated lactate.
  • Embodiment 43 The method of any one of embodiments 1-42, wherein the growth medium comprises Y-27632. [0439] Embodiment 44.
  • Embodiment 45 The method of any one of embodiments 35-43, wherein the culturing in step (b) is dynamic.
  • Embodiment 45 The method of any one of embodiments 1-44, wherein the cultured cells in step (b) comprise hematopoietic cell intermediates.
  • Embodiment 46 The method of embodiment 45, wherein the hematopoietic cell intermediates express CD34, CD90.
  • Embodiment 47 The method of embodiment 45 or 46, wherein the hematopoietic cell intermediates do not express CD38.
  • Embodiment 48 The method of embodiment 45, wherein the hematopoietic cell intermediates express CD45.
  • Embodiment 49 The method of embodiment 48, wherein the hematopoietic cell intermediates do not express CD38.
  • Embodiment 50 The method of any one of embodiments 1-49, wherein the microcarrier comprises one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof.
  • Embodiment 51 Embodiment 51.
  • the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof.
  • Embodiment 52 The method of any one of embodiments 1-51, wherein the microcarrier is not coated.
  • Embodiment 53 The method of any one of embodiments 1-52, wherein the microcarrier is coupled with protamine or polylysine.
  • Embodiment 54 The method of any one of embodiments 1-53, wherein the microcarrier has a neutral charge.
  • Embodiment 55 The method of any one of embodiments 1-54, wherein the microcarrier is negatively charged.
  • Embodiment 56 The method of any one of embodiments 1-54, wherein the microcarrier is negatively charged.
  • Embodiment 60 The method of any one of embodiments 1-55, wherein the microcarrier is hydrophilic.
  • Embodiment 57 The method of any one of embodiments 1-56, wherein the ECM component comprises a matrigel, laminin, vitronectin, collagen, a derivative thereof, or any combination thereof.
  • Embodiment 58 The method of embodiment 57, wherein the laminin is human laminin.
  • Embodiment 59 The method of embodiment 58, wherein the human laminin is 511 E8 fragment. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0455] Embodiment 60.
  • Embodiment 61 The method of any one of embodiments 1-60, wherein the mDCs are unable to replicate. [0457] Embodiment 62.
  • Embodiment 63 The method of any one of embodiments 1-62, further comprising (f) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen.
  • Embodiment 64 The method of any one of embodiments 1-62, further comprising (f) cryopreserving the population of mDCs.
  • Embodiment 65 The method of embodiment 64, further comprising (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen.
  • Embodiment 66 The method of embodiment 63 or 65, wherein the population of mDCs is contacted with two or more antigens.
  • Embodiment 67 The method of any one of embodiments 63-65, wherein the antigen is a tumor antigen.
  • Embodiment 68 The method of embodiment 67, wherein the tumor antigen comprises a tumor associated antigen (TAA).
  • TAA tumor associated antigen
  • Embodiment 69 The method of embodiment 68, wherein the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • Embodiment 70 The method of embodiment 67, wherein the tumor antigen comprises a tumor-specific antigen (TSA).
  • TSA tumor-specific antigen
  • Embodiment 71 The method of embodiment 67, wherein the tumor antigen comprises a neoantigen.
  • Embodiment 72 The method of embodiment 63 or 65, wherein the nucleic acid comprises an RNA.
  • Embodiment 73 The method of embodiment 72, wherein the RNA comprises an mRNA encoding hTERT or fragment thereof.
  • Embodiment 74 The method of embodiment 7 , the wherein the fragment comprises one or more immunogenic epitopes of hTERT.
  • Embodiment 75 The method of any one of embodiments 72-74, wherein the RNA comprises an mRNA encoding lysosomal-associated membrane protein 1 (LAMP1) or a fragment thereof.
  • Embodiment 76 The method of any one of embodiments 72-75, wherein the RNA comprises an mRNA encoding heat shock protein 96 (HSP96).
  • Embodiment 77 The method of any one of embodiments 63-76, wherein the population of mDCs comprising the one or more antigens is cryopreserved.
  • Embodiment 78 Embodiment 78.
  • Embodiment 80 A cell produced by any one of embodiments 1-59, wherein the cell expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160 and SSEA-5. PATENT Attorney Docket No.: 058125-649001WO Client Ref.
  • Embodiment 81 The cell of embodiment 80, wherein the cell co-expresses CD45 and CD83.
  • Embodiment 82 A cell produced by any one of embodiments 63-77, wherein the cell (i) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160, and SSEA-5; and (ii) comprises the one or more antigens.
  • Embodiment 83 The cell of embodiment 82, wherein the cell co-expresses CD45 and CD83.
  • Embodiment 84 Embodiment 84.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject comprising: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier.
  • Embodiment 85 The cell vaccine of embodiment 84, wherein the one or more dendritic cells co-express CD45 and CD83.
  • a cell vaccine for treating or delaying progression of a disease or disorder in a subject comprising: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) comprising a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier.
  • Embodiment 87 The cell vaccine of embodiment 86, wherein the one or more dendritic cells co-express CD45 and CD83.
  • Embodiment 88 Embodiment 88.
  • the cell vaccine of embodiment 86 wherein the nucleic acid comprises a DNA.
  • Embodiment 89 The cell vaccine of embodiment 88, wherein the DNA is under the control of a promoter. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT
  • Embodiment 90 The cell vaccine of embodiment 89, wherein the promoter is an inducible promoter.
  • Embodiment 91 The cell vaccine of embodiment 90, further comprising (c) an inducing agent.
  • Embodiment 92 The cell vaccine of embodiment 91, wherein the inducing agent is desferrioxamine.
  • Embodiment 93 The cell vaccine of embodiment 86, wherein the nucleic acid comprises an RNA.
  • Embodiment 94 The cell vaccine of any one of embodiments 86-93, wherein the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent.
  • Embodiment 95 The cell vaccine of any one of embodiments 86-94, wherein the cell vaccine comprises between about 0.5 x 10 7 dendritic cells to about 2.5 x 10 7 dendritic cells.
  • Embodiment 96 Embodiment 96.
  • Embodiment 97 The cell vaccine of any one of embodiments 86-96, wherein the cell vaccine comprises about 1.0 x 10 7 dendritic cells.
  • Embodiment 98 The cell vaccine of any one of embodiments 86-97, wherein the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder.
  • Embodiment 99 Embodiment 99.
  • the cell vaccine of embodiment 98 wherein the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer.
  • Embodiment 101 The cell vaccine of embodiment 100, wherein the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents.
  • Embodiment 102 The cell vaccine of any one of embodiments 86-101, wherein the one or more antigens is identified from the subject.
  • Embodiment 103 The cell vaccine of any one of embodiments 86-102, wherein the cell vaccine is formulated to be administered to the subject directly after thawing.
  • Embodiment 104 Embodiment 101.
  • Embodiment 101 The cell vaccine of embodiment 100, wherein the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents.
  • Embodiment 102 The cell vaccine of any one of embodiments 86-101, wherein the one or more antigens is identified from the subject.
  • Embodiment 103 The cell vaccine
  • Embodiment 105 The cell vaccine of embodiment 103 or 104, wherein the cell vaccine is formulated to be administered without washing or reconstitution.
  • Embodiment 106 The cell vaccine of any one of embodiments 103-105, wherein the cell vaccine is formulated for administration within about 4 hours of thawing.
  • Embodiment 107 The cell vaccine of any one of embodiments 103-105, wherein the cell vaccine is formulated for administration more than 4 hours after thawing.
  • Embodiment 108 Embodiment 108.
  • Embodiment 109 A method of manufacturing a cell vaccine for treating cancer, the method comprising: (a) obtaining one or more mature dendritic cells (mDCs) according to the method of any one of embodiments 1-60; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer.
  • mDCs mature dendritic cells
  • Embodiment 110 Embodiment 110.
  • Embodiment 111 The method of embodiment 109, wherein optionally the one or more mDCs are cryopreserved and thawed before step (b).
  • Embodiment 112. The method of any one of embodiments 109-111, wherein the one or more antigens is one or more tumor antigens.
  • Embodiment 113 The method of embodiment 112, wherein the one or more tumor antigens comprises a TAA.
  • Embodiment 114 The method of embodiment 113, wherein the TAA is hTERT.
  • Embodiment 115 Embodiment 115.
  • Embodiment 112 wherein the one or more tumor antigens comprises a TSA.
  • Embodiment 116 The method of embodiment 112, wherein the one or more tumor antigens comprises a neoantigen.
  • Embodiment 117 The method of any one of embodiments 109-116, wherein the one or more nucleic acid molecules comprises one or more RNA molecules.
  • Embodiment 118 The method of embodiment 117, wherein the one or more RNA molecules comprises hTERT mRNA.
  • Embodiment 119 The method of any one of embodiments 109-118, wherein the manufacturing takes about 30 days to about 50 days.
  • Embodiment 120 The method of any one of embodiments 109-118, wherein the manufacturing takes about 30 days to about 50 days.
  • Embodiment 121 The method of any one of embodiments 109-120, wherein the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia.
  • NSCLC non-small cell lung cancer
  • Embodiment 122 A method for treating or delaying progression of a disease or disorder in a subject, comprising administering to the subject a cell vaccine of any one of embodiments 84 to 102.
  • Embodiment 123 Embodiment 123.
  • Embodiment 124 A method for treating cancer, comprising administering to a subject having cancer a cell vaccine made by the method of any one of embodiments 109-120.
  • Embodiment 125 The method of embodiment 124, wherein the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia.
  • Embodiment 126 The method of embodiment 124 or 125, wherein the mDCs are allogeneic to the subject.

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Provided herein, inter alia, are methods for differentiating human embryonic stem cells into dendritic cells. Inter alia, are provided methods and compositions for manufacturing cell vaccines. The cell vaccines provided herein can be used in the treatment of cancer.

Description

PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT COMPOSITIONS AND METHOD FOR EXPANSION OF EMBRYONIC STEM CELLS CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of priority under 35 U.S.C. § 119(e) of the U.S. Patent Application No.63/423,501, filed on November 8, 2022, which is hereby incorporated by reference in its entirety and for all purposes. BACKGROUND [0002] Novel immunotherapy agents, such as anti-CTLA4 and anti-PD-1 antibodies, as well as vaccines such as TG4010, have demonstrated that some cancers are amenable to immunotherapy. While these agents appear to improve outcomes, they are only able to offer durable disease control to a subset of subjects. The compositions and methods provided herein, inter alia, address these and other problems in the art. BRIEF SUMMARY OF THE INVENTION [0003] In an aspect is provided a method of differentiating human embryonic stem cells (hESCs) into mature dendritic cells (mDCs). In embodiments, the method includes: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce a cultured suspendable expansion complex comprising cultured cells; (c) removing the cultured cells from the cultured suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs); and (e) contacting the population of iDCs with a maturation cocktail for a third period of time to differentiate the population of iDCs into a population of mature dendritic cells (mDCs). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0004] In another aspect is provided a cell produced as provided herein including embodiments thereof, wherein the cell expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160 and SSEA-5. [0005] In another aspect is provided a cell produced as provided herein including embodiments thereof, wherein the cell (i) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA- A, CD40 and CCR7, but does not express one or more of CD14, TRA160, and SSEA-5; and (ii) includes the one or more antigens. [0006] In another aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier. [0007] In another aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. [0008] In another aspect is provided a method of manufacturing a cell vaccine for treating cancer, the method including: (a) obtaining one or more mature dendritic cells (mDCs) according to the method provided herein including embodiments thereof; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer. [0009] In another aspect is provided a method for treating or delaying progression of a disease or disorder in a subject, including administering to the subject a cell vaccine provided herein including embodiments thereof. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0010] In another aspect is provided a method for treating cancer, including administering to a subject having cancer a cell vaccine made by the method provided herein including embodiments thereof. BRIEF DESCRIPTION OF THE DRAWINGS [0011] FIG.1 shows relapse free survival at 52 Months in AML subjects receiving autologous VAC1 (n=19). [0012] FIG.2 shows the H1 Master Cell Bank (MCB) production process. [0013] FIG.3 shows the H1 Working Cell Bank (WCB) production process. [0014] FIG.4 shows the LCT-VAC2 and LCT-mDCs production process. [0015] FIG.5 shows results from the H1 post irradiation performance test. The morphology assessment occurred on Day 4 of culturing. [0016] FIG.6 shows the detailed IPC plan specific for monitoring LCT-mDCs differentiation process. [0017] FIGS.7A-7B show results from a migration assay. The assay test migratory activity of shows a schematic of the transwell cell migration assay. FIG.7B shows the quantified migratory activity of LCT-mDCs compared to PBMC-mDCs. [0018] FIGS.8A-8C show results from a mixed lymphocyte reaction (MLR) assay. The MLR assay tests ton-specific activation of PMBCs by LCT-mDCs by measuring gated CD4+ T cell proliferation using fluorescent dye labeling (CFSE) after 6 days of co-culture. LCT-mDCs evoked non-specific T cell proliferation of both CD4+ and CD8+ populations after co-incubation with PBMCs. [0019] FIGS.9A-9B show a schematic of (FIG.9A) and results (FIG.9B) from a PC antigen presentation assay. The PC antigen presentation assay tests mDCs ability to successfully process PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT and present PC antigen (by peptide or mRNA EP) to antigen specific T cells, which results in T [0020] FIGS.10A-10B show a schematic of (FIG.10) and results (FIG.10B) from an hTERT antigen presentation assay. The hTERT antigen presentation assay tests mDCs ability to successfully process and present hTERT antigen (by peptide or mRNA EP) to antigen specific T DETAILED DESCRIPTION [0021] While various embodiments and aspects of the present invention are shown and described herein, it will be obvious to those skilled in the art that such embodiments and aspects are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. [0022] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in the application including, without limitation, patents, patent applications, articles, books, manuals, and treatises are hereby expressly incorporated by reference in their entirety for any purpose. [0023] The abbreviations used herein have their conventional meaning within the chemical and biological arts. The chemical structures and formulae set forth herein are constructed according to the standard rules of chemical valency known in the chemical arts. [0024] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art. See, e.g., Singleton et al., DICTIONARY OF MICROBIOLOGY AND MOLECULAR BIOLOGY 2nd ed., J. Wiley & Sons (New York, NY 1994); Sambrook et al., MOLECULAR CLONING, A LABORATORY MANUAL, Cold Springs Harbor Press (Cold Springs Harbor, NY 1989). Any methods, devices and materials similar or equivalent to those described herein can be used in the practice of this PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT invention. The following definitions are provided to facilitate understanding of certain terms used frequently herein and are not meant to limit the scope of the present disclosure. [0025] "Nucleic acid" refers to nucleotides (e.g., deoxyribonucleotides or ribonucleotides) and polymers thereof in either single-, double- or multiple-stranded form, or complements thereof; or nucleosides (e.g., deoxyribonucleosides or ribonucleosides). In embodiments, “nucleic acid” does not include nucleosides. The terms “polynucleotide,” “oligonucleotide,” “oligo” or the like refer, in the usual and customary sense, to a linear sequence of nucleotides. The term “nucleoside” refers, in the usual and customary sense, to a glycosylamine including a nucleobase and a five- carbon sugar (ribose or deoxyribose). Non limiting examples, of nucleosides include, cytidine, uridine, adenosine, guanosine, thymidine and inosine. The term “nucleotide” refers, in the usual and customary sense, to a single unit of a polynucleotide, i.e., a monomer. Nucleotides can be ribonucleotides, deoxyribonucleotides, or modified versions thereof. Examples of polynucleotides contemplated herein include single and double stranded DNA, single and double stranded RNA, and hybrid molecules having mixtures of single and double stranded DNA and RNA. Examples of nucleic acid, e.g. polynucleotides contemplated herein include any types of RNA, e.g. mRNA, siRNA, miRNA, and guide RNA and any types of DNA, genomic DNA, plasmid DNA, and minicircle DNA, and any fragments thereof. [0026] The term "gene" means the segment of DNA involved in producing a protein; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons). The leader, the trailer as well as the introns include regulatory elements that are necessary during the transcription and the translation of a gene. Further, a "protein gene product" is a protein expressed from a particular gene. [0027] The terms "plasmid", "vector" or "expression vector" refer to a nucleic acid molecule that encodes for genes and/or regulatory elements necessary for the expression of genes. Expression of a gene from a plasmid can occur in cis or in trans. If a gene is expressed in cis, the gene and the regulatory elements are encoded by the same plasmid. Expression in trans refers to the instance where the gene and the regulatory elements are encoded by separate plasmids. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0028] The terms "transfection", "transduction", "transfecting" or "transducing" can be used interchangeably and are defined as a process of introducing a nucleic acid molecule or a protein to a cell. Nucleic acids are introduced to a cell using non-viral or viral-based methods. The nucleic acid molecules may be gene sequences encoding complete proteins or functional portions thereof. Non-viral methods of transfection include any appropriate transfection method that does not use viral DNA or viral particles as a delivery system to introduce the nucleic acid molecule into the cell. Exemplary non-viral transfection methods include calcium phosphate transfection, liposomal transfection, nucleofection, sonoporation, transfection through heat shock, magnetifection and electroporation. In some embodiments, the nucleic acid molecules are introduced into a cell using electroporation following standard procedures well known in the art. For viral-based methods of transfection any useful viral vector may be used in the methods described herein. Examples for viral vectors include, but are not limited to retroviral, adenoviral, lentiviral and adeno-associated viral vectors. In some embodiments, the nucleic acid molecules are introduced into a cell using a Typically, transduction or transfection of a protein relies on attachment of a peptide or protein capable of crossing the cell membrane to the protein of interest. See, e.g., Ford et al. (2001) Gene Therapy 8:1-4 and Prochiantz (2007) Nat. Methods 4:119-20. [0029] "Contacting" is used in accordance with its plain ordinary meaning and refers to the process of allowing at least two distinct species (e.g. cells and an agent) to become sufficiently proximal to react, interact, or physically touch. It should be appreciated; however, that the resulting reaction product can be produced directly from a reaction between the added reagents or from an intermediate from one or more of the added reagents which can be produced in the reaction mixture. [0030] The term “cell culture,” as used herein, refers to a plurality of cells grown in vitro over time. The cell culture my originate from a plurality of human pluripotent stem (hPS) cells or from a single hPS cell and may include all of the progeny of the originating cell or cells, regardless of 1) the number of passages or divisions the cell culture undergoes over the lifetime of the culture; and PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 2) any changes in phenotype to one or more cells within the culture over the lifetime of the culture (e.g. resulting from differentiation of one or more hPS cells in the culture). Thus, as used herein, a cell culture begins with the initial seeding of one or more suitable vessels with at least one hPS cell and ends when the last surviving progeny of the original founder(s) is harvested or dies. Seeding of one or more additional culture vessels with progeny of the original founder cells is also considered to be a part of the original cell culture. [0031] The term “dendritic cells” or “DCs,” as used herein, refers to professional antigen- presenting cells (also known as accessory cells) of the mammalian immune system. In embodiments, the main function of DCs is to process antigen material and present it on the cell surface to the T cells of the immune system. In embodiments, DCs act as messengers between the innate and the adaptive immune systems. [0032] The term "isolated", when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It can be, for example, in a homogeneous state and may be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. [0033] The term "exogenous" refers to a molecule or substance (e.g., a compound, nucleic acid or protein) that originates from outside a given cell or organism. For example, an "exogenous cytokine" as referred to herein is a cytokine that does not originate from the cell or cell population. Conversely, the term "endogenous" refers to a molecule or substance that is native to, or originates within, a given cell or organism. [0034] As defined herein, the term "inhibition", "inhibit", "inhibiting" and the like in reference to cell proliferation (e.g., cancer cell proliferation) means negatively affecting (e.g., decreasing proliferation) or killing the cell. In some embodiments, inhibition refers to reduction of a disease or symptoms of disease (e.g., cancer, cancer cell proliferation). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0035] The term "expression" includes any step involved in the production of the polypeptide including, but not limited to, transcription, post-transcriptional modification, translation, post- translational modification, and secretion. Expression can be detected using conventional techniques for detecting protein (e.g., ELISA, Western blotting, flow cytometry, immunofluorescence, immunohistochemistry, etc.). [0036] A “control” or “standard control” refers to a sample, measurement, or value that serves as a reference, usually a known reference, for comparison to a test sample, measurement, or value. For example, a test sample can be taken from a patient suspected of having a given disease (e.g. cancer) and compared to a known normal (non-diseased) individual (e.g. a standard control subject). A standard control can also represent an average measurement or value gathered from a population of similar individuals (e.g. standard control subjects) that do not have a given disease (i.e. standard control population), e.g., healthy individuals with a similar medical background, same age, weight, etc. A standard control value can also be obtained from the same individual, e.g. from an earlier-obtained sample from the patient prior to disease onset. For example, a control can be devised to compare therapeutic benefit based on pharmacological data (e.g., half- life) or therapeutic measures (e.g., comparison of side effects). Controls are also valuable for determining the significance of data. For example, if values for a given parameter are widely variant in controls, variation in test samples will not be considered as significant. One of skill will recognize that standard controls can be designed for assessment of any number of parameters (e.g. RNA levels, protein levels, specific cell types, specific bodily fluids, specific tissues, etc). [0037] One of skill in the art will understand which standard controls are most appropriate in a given situation and be able to analyze data based on comparisons to standard control values. Standard controls are also valuable for determining the significance (e.g. statistical significance) of data. For example, if values for a given parameter are widely variant in standard controls, variation in test samples will not be considered as significant. [0038] “Patient” or “subject in need thereof” refers to a living organism suffering from or prone to a disease or condition that can be treated by administration of a composition or pharmaceutical composition as provided herein. Non-limiting examples include humans, other mammals, PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT bovines, rats, mice, dogs, monkeys, goat, sheep, cows, deer, and other non-mammalian animals. In some embodiments, a patient is human. [0039] The terms “disease” or “condition” refer to a state of being or health status of a patient or subject capable of being treated with the compositions or methods provided herein. The disease may be a cancer. The cancer may refer to a solid tumor malignancy. Solid tumor malignancies include malignant tumors that may be devoid of fluids or cysts. For example, the solid tumor malignancy may include breast cancer, ovarian cancer, pancreatic cancer, cervical cancer, gastric cancer, renal cancer, head and neck cancer, bone cancer, skin cancer or prostate cancer. In some further instances, “cancer” refers to human cancers and carcinomas, sarcomas, adenocarcinomas, lymphomas, leukemias, including solid and lymphoid cancers, kidney, breast, lung, bladder, colon, ovarian, prostate, pancreas, stomach, brain, head and neck, skin, uterine, testicular, glioma, esophagus, and liver cancer, including hepatocarcinoma, lymphoma, including B-acute lymphoblastic lymphoma, non-Hodgkin’s lymphomas (e.g., Burkitt’s, Small Cell, and Large Cell lymphomas), Hodgkin’s lymphoma, leukemia (including acute myeloid leukemia (AML), ALL, and CML), or multiple myeloma. [0040] As used herein, the term “cancer” refers to all types of cancer, neoplasm or malignant tumors found in mammals (e.g., humans), including leukemia, carcinomas and sarcomas. Exemplary cancers that may be treated with a composition or method provided herein include breast cancer, colon cancer, kidney cancer, leukemia, lung cancer, melanoma, ovarian cancer, [0041] The term “associated” or “associated with” in the context of a substance or substance activity or function associated with a disease (e.g. a protein associated disease, a cancer (e.g., breast cancer, lung cancer)) means that the disease (e.g. cancer) is caused by (in whole or in part), or a symptom of the disease is caused by (in whole or in part) the substance or substance activity or function. As used herein, what is described as being associated with a disease, if a causative agent, could be a target for treatment of the disease. [0042] A "therapeutic agent" as referred to herein, is a composition useful in treating a disease such as cancer (e.g., leukemia). In embodiments, the therpaeutic agent is an anti-cancer agent. “Anti-cancer agent” is used in accordance with its plain ordinary meaning and refers to a PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT composition (e.g. compound, drug, antagonist, inhibitor, modulator) having antineoplastic properties or the ability to inhibit the growth or proliferation of cells. In embodiments, an anti- cancer agent is a chemotherapeutic. In embodiments, an anti-cancer agent is an agent identified herein having utility in methods of treating cancer. In embodiments, an anti-cancer agent is an agent approved by the FDA or similar regulatory agency of a country other than the USA, for treating cancer. [0043] The terms “treatment” and “treating” as used herein refer to the medical management of a subject with the intent to cure, ameliorate, or stabilize a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. It is understood that treatment, while intended to cure, ameliorate, or stabilize a disease, pathological condition, or disorder, need not actually result in the cure, amelioration, or stabilization. The effects of treatment can be measured or assessed as described herein and as known in the art as is suitable for the disease, pathological condition, or disorder involved. Such measurements and assessments can be made in qualitative and/or quantitative terms. Thus, for example, characteristics or features of a disease, pathological condition, or disorder and/or symptoms of a disease, pathological condition, or disorder can be reduced to any effect or to any amount. [0044] The term “in need of treatment” as used herein refers to a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, or individual in the case of humans; veterinarian in the case of animals, including non-human mammals) that a subject requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a care PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT giver's expertise, but that include the knowledge that the subject is ill, or will be ill, as the result of a condition that is treatable by the disclosed compositions. [0045] As used herein, "feeder cells" refers to non-hPS cells that are co-cultured with hPS cells and provide support for the hPS cells. Support may include facilitating the growth and maintenance of the hPS cell culture by providing the hPS cell culture with one or more cell factors such that the hPS cells are maintained in a substantially undifferentiated state. Feeder cells may either have a different genome than the hPS cells or the same genome as the hPS cells and may originate from a non-primate species, such as mouse, or may be of primate origin, e.g., human. Examples of feeder cells may include cells having the phenotype of connective tissue such as murine fibroblast cells, human fibroblasts. [0046] As used herein, "feeder-free" refers to a condition where the referenced composition contains no added feeder cells. To clarify, the term feeder-free encompasses, inter alia, situations where primate pluripotent stem cells are passaged from a culture which may comprise some feeders into a culture without added feeders even if some of the feeders from the first culture are present in the second culture. [0047] “Serum free”, as used herein, refers tissue culture growth conditions that have no added animal serum such fetal bovine serum, calf serum, horse serum, and no added commercially available serum replacement supplements such as B-27. Serum free includes, for example, media which may comprise human albumin, human transferrin and recombinant human insulin. [0048] As used herein, “electroporation” refers to a method for permeabilizing cell membranes by generating membrane pores with electrical stimulation. The applications of electroporation include, but are not limited to, the delivery of DNA, RNA, siRNA, peptides, proteins, antibodies, drugs or other substances to a variety of cells such as mammalian cells, plant cells, yeasts, other eukaryotic cells, bacteria, other microorganisms, and cells from human patients. [0049] The terms “dose” and “dosage” are used interchangeably herein. A dose refers to the amount of active ingredient given to an individual at each administration. The dose will vary depending on a number of factors, including the range of normal doses for a given therapy, PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT frequency of administration; size and tolerance of the individual; severity of the condition; risk of side effects; and the route of administration. One of skill will recognize that the dose can be modified depending on the above factors or based on therapeutic progress. The term “dosage form” refers to the particular format of the pharmaceutical or pharmaceutical composition, and depends on the route of administration. For example, a dosage form can be in a liquid form for nebulization, e.g., for inhalants, in a tablet or liquid, e.g., for oral delivery, or a saline solution, e.g., for injection. [0050] By “therapeutically effective dose or amount” as used herein is meant a dose that produces effects for which it is administered (e.g. treating a disease). The exact dose and formulation will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques. For example, for the given parameter, a therapeutically effective amount will show an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100%. Therapeutic efficacy can also be expressed as “- fold” increase or decrease. For example, a therapeutically effective amount can have at least a 1.2- fold, 1.5-fold, 2-fold, 5-fold, or more effect over a standard control. A therapeutically effective dose or amount may ameliorate one or more symptoms of a disease. A therapeutically effective dose or amount may prevent or delay the onset of a disease or one or more symptoms of a disease when the effect for which it is being administered is to treat a person who is at risk of developing the disease. [0051] As used herein, the term "administering" means any suitable method for administering cells to a subject, including parenteral, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial. By "co-administer" it is meant that a composition described herein is administered at the same time, just prior to, or just after the administration of one or more additional therapies, for example cancer therapies such as chemotherapy, hormonal therapy, radiotherapy, or immunotherapy. The compositions of the invention can be administered alone or can be coadministered to the patient. Coadministration is meant to include simultaneous or sequential administration of the compositions individually or in PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT combination (more than one composition). Thus, the preparations can also be combined, when desired, with other active substances (e.g. to reduce metabolic degradation). [0052] As used herein, the term “pharmaceutically acceptable” is used synonymously with “physiologically acceptable” and “pharmacologically acceptable”. A pharmaceutical composition will generally comprise agents for buffering and preservation in storage, and can include buffers and carriers for appropriate delivery, depending on the route of administration. [0053] "Pharmaceutically acceptable excipient" and "pharmaceutically acceptable carrier" refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present invention without causing a significant adverse toxicological effect on the patient. Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidines, and the like. Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the invention. One of skill in the art will recognize that other pharmaceutical excipients are useful in the present invention. [0054] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes. [0055] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes. METHODS OF DIFFERENTIATING [0056] Pluripotent stem cells have the ability to both proliferate continuously in culture and, under appropriate growth conditions, differentiate into lineage restricted cell types representative of all three primary germ layers: endoderm, mesoderm and ectoderm (U.S. Patent Nos.5,843,780; 6,200,806; 7,029,913; Shamblott et al., (1998) Proc. Natl. Acad. Sci. USA 95:13726; Takahashi et al., (2007) Cell 131(5):861; Yu et al., (2007) Science 318:5858). [0057] A pluripotent stem cell (PS) will, under appropriate growth conditions, be able to form at least one cell type from each of the three primary germ layers: mesoderm, endoderm and ectoderm. The PS cells may originate from pre-embryonic, embryonic or fetal tissue or mature differentiated cells. Alternatively, an established PS cell line may be a suitable source of cells for practicing the invention. Typically, the hPS cells are not derived from a malignant source. hPS cells will form teratomas when implanted in an immuno-deficient mouse, e.g. a SCID mouse. [0058] Prototype “human Pluripotent Stem cells” (hPS cells) are pluripotent cells derived from pre-embryonic, embryonic, or fetal tissue at any time after fertilization, and have the characteristic of being capable under appropriate conditions of producing progeny of several different cell types that are derivatives of all of the three germinal layers (endoderm, mesoderm, and ectoderm), according to a standard art-accepted test, such as the ability to form a teratoma in 8-12 week old SCID mice. Unless otherwise specified, hPS cells are not derived from a cancer cell or other malignant source. It is desirable (but not always necessary) that the cells be euploid. [0059] Exemplary are embryonic stem cells and embryonic germ cells used as existing cell lines or established from primary embryonic tissue of human origin. This invention can also be practiced using pluripotent cells obtained from primary embryonic tissue, without first establishing an undifferentiated cell line. [0060] hPS cells can be propagated continuously in culture, using culture conditions that promote proliferation while inhibiting differentiation. Traditionally, ES cells are cultured on a PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT layer of feeder cells, typically fibroblasts derived from embryonic or fetal tissue (Thomson et al., Science 282: 1145, 1998). [0061] However, hPS cells can be maintained in an undifferentiated state even without feeder cells. The environment for feeder-free cultures includes a suitable culture substrate, such as ECM- based hydrogel, such as solubilized basement membrane preparation extracted from the Engelbreth-Holm- Swarm (EHS) mouse sarcoma (e.g., MATRIGEL®), or laminin. The cultures are supported by a nutrient medium containing factors that promote proliferation of the cells without differentiation (WO 99/20741). Such factors may be introduced into the medium by culturing the medium with cells secreting such factors, such as irradiated primary mouse embryonic fibroblasts, telomerized mouse fibroblasts, or fibroblast-like cells derived from hPS cells (U.S. Patent 6,642,048). Medium can be conditioned by plating the feeders in a serum free medium such as Knock-Out DMEM (Gibco), supplemented with serum replacement ranging from 10-30%, preferably 20% (US 2002/0076747 Al, Life Technologies Inc.) and 4 ng/mL bFGF. Medium that has been conditioned for 1-2 days is supplemented with further bFGF, and used to support hPS cell culture for 1-2 days (WO 01/51616; Xu et al., Nat. Biotechnol.19:971, 2001). [0062] In an aspect is provided a method of differentiating human embryonic stem cells (hESCs) into mature dendritic cells (mDCs), the method including: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce a cultured suspendable expansion complex comprising cultured cells; (c) removing the cultured cells from the cultured suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs); and (e) contacting the population of iDCs with a maturation cocktail for a third period of time to differentiate the population of iDCs into a population of mature dendritic cells (mDCs). In embodiments, the culturing in step (b) is dynamic. In embodiments, the culturing in step (b) is static. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0063] In embodiments, the removing of step (c) includes an enzyme-free dissociation reagent. In embodiments, the removing of step (c) does not include manual dissection or manual scraping. In embodiments, the removing of step (c) does not include manual dissection. In embodiments, the removing of step (c) does not include manual scraping. [0064] In embodiments, the first period of time is between about 1 day to about 9 days. In embodiments, the first period of time is between about 2 days to about 9 days. In embodiments, the first period of time is between about 3 days to about 9 days. In embodiments, the first period of time is between about 4 days to about 9 days. In embodiments, the first period of time is between about 5 days to about 9 days. In embodiments, the first period of time is between about 6 days to about 9 days. In embodiments, the first period of time is between about 7 days to about 9 days. In embodiments, the first period of time is between about 8 days to about 9 days. [0065] In embodiments, the first period of time is between 1 day to 8 days. In embodiments, the first period of time is between 1 day to 7 days. In embodiments, the first period of time is between 1 day to 6 days. In embodiments, the first period of time is between 1 day to 5 days. In embodiments, the first period of time is between 1 day to 4 days. In embodiments, the first period of time is between about 1 day to 3 days. In embodiments, the first period of time is between 1 day to 2 days. In embodiments, the first period of time is between 1 day to 9 days. In embodiments, the first period of time is between 2 days to 9 days. In embodiments, the first period of time is between 3 days to 9 days. In embodiments, the first period of time is between 4 days to 9 days. In embodiments, the first period of time is between 5 days to 9 days. In embodiments, the first period of time is between 6 days to 9 days. In embodiments, the first period of time is between 7 days to 9 days. In embodiments, the first period of time is between 8 days to 9 days. [0066] In embodiments, the first period of time is between about 1 day to about 8 days. In embodiments, the first period of time is between 1 day to 7 days. In embodiments, the first period of time is between 1 day to 6 days. In embodiments, the first period of time is between 1 day to 5 days. In embodiments, the first period of time is between 1 day to 4 days. In embodiments, the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT first period of time is between 1 day to 3 days. In embodiments, the first period of time is between 1 day to 2 days. [0067] In embodiments, the differentiation cocktail includes at least one exogenous cytokine, wherein the at least one exogenous cytokine includes granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3). In embodiments, the at least one exogenous cytokine includes granulocyte- macrophage colony stimulating factor (GM-CSF). In embodiments, the at least one exogenous cytokine includes bond morphogenic protein 4 (BMP-4). In embodiments, the at least one exogenous cytokine includes vascular endothelial growth factor (VEGF). In embodiments, the at least one exogenous cytokine includes stem cell factor (SCF). In embodiments, the at least one exogenous cytokine includes fetal liver kinase ligand (FLT3L). In embodiments, the at least one exogenous cytokine includes thrombopoietin (TPO). In embodiments, the at least one exogenous cytokine includes interleukin 3 (IL-3). In embodiments, one or more of the recited cytokines may be expressly excluded. [0068] In embodiments, the differentiation cocktail includes GM-CSF. In embodiments, the differentiation cocktail includes GM-CSF and SCF. In embodiments, the differentiation cocktail includes GM-CSF, SCF, and VEGF. In embodiments, the differentiation cocktail includes GM- CSF, SCF, VEGF, and BMP-4. In embodiments, the BMP-4 is an animal-free Good Manufacturing Process (GMP) grade BMP-4. [0069] In embodiments, an iDC of the population of iDCs in step (d) expresses CD45 and CD86. In embodiments, an iDC of the population of iDCs in step (d) does not express CD38. [0070] In embodiments, the second period of time is between about 1 day to about 32 days. In embodiments, the second period of time is between about 2 days to about 32 days. In embodiments, the second period of time is between about 3 days to about 32 days. In embodiments, the second period of time is between about 4 days to about 32 days. In embodiments, the second period of time is between about 5 days to about 32 days. In embodiments, the second period of time is between about 6 days to about 32 days. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the second period of time is between about 7 days to about 32 days. In embodiments, the second period of time is between about 8 days to about 32 days. In embodiments, the second period of time is between about 9 days to about 32 days. In embodiments, the second period of time is between about 10 days to about 32 days. In embodiments, the second period of time is between about 11 days to about 32 days. In embodiments, the second period of time is between about 12 days to about 32 days. In embodiments, the second period of time is between about 13 days to about 32 days. In embodiments, the second period of time is between about 14 days to about 32 days. In embodiments, the second period of time is between about 15 days to about 32 days. In embodiments, the second period of time is between about 16 days to about 32 days. In embodiments, the second period of time is between about 17 days to about 32 days. In embodiments, the second period of time is between about 18 days to about 32 days. In embodiments, the second period of time is between about 19 days to about 32 days. In embodiments, the second period of time is between about 20 days to about 32 days. In embodiments, the second period of time is between about 21 days to about 32 days. In embodiments, the second period of time is between about 22 days to about 32 days. In embodiments, the second period of time is between about 23 days to about 32 days. In embodiments, the second period of time is between about 24 days to about 32 days. In embodiments, the second period of time is between about 25 days to about 32 days. In embodiments, the second period of time is between about 26 days to about 32 days. In embodiments, the second period of time is between about 27 days to about 32 days. In embodiments, the second period of time is between about 28 days to about 32 days. In embodiments, the second period of time is between about 29 days to about 32 days. In embodiments, the second period of time is between about 30 days to about 32 days. In embodiments, the second period of time is between about 31 days to about 32 days. [0071] In embodiments, the second period of time is between about 1 day to about 31 days. In embodiments, the second period of time is between about 1 day to about 30 days. In embodiments, the second period of time is between about 1 day to about 29 days. In embodiments, the second period of time is between about 1 day to about 28 days. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the second period of time is between about 1 day to about 27 days. In embodiments, the second period of time is between about 1 day to about 26 days. In embodiments, the second period of time is between about 1 day to about 25 days. In embodiments, the second period of time is between about 1 day to about 24 days. In embodiments, the second period of time is between about 1 day to about 23 days. In embodiments, the second period of time is between about 1 day to about 22 days. In embodiments, the second period of time is between about 1 day to about 21 days. In embodiments, the second period of time is between about 1 day to about 20 days. In embodiments, the second period of time is between about 1 day to about 19 days. In embodiments, the second period of time is between about 1 day to about 18 days. In embodiments, the second period of time is between about 1 day to about 17 days. In embodiments, the second period of time is between about 1 day to about 16 days. In embodiments, the second period of time is between about 1 day to about 15 days. In embodiments, the second period of time is between about 1 day to about 14 days. In embodiments, the second period of time is between about 1 day to about 13 days. In embodiments, the second period of time is between about 1 day to about 12 days. In embodiments, the second period of time is between about 1 day to about 11 days. In embodiments, the second period of time is between about 1 day to about 10 days. In embodiments, the second period of time is between about 1 day to about 9 days. In embodiments, the second period of time is between about 1 day to about 8 days. In embodiments, the second period of time is between about 1 day to about 7 days. In embodiments, the second period of time is between about 1 day to about 6 days. In embodiments, the second period of time is between about 1 day to about 5 days. In embodiments, the second period of time is between about 1 day to about 4 days. In embodiments, the second period of time is between about 1 day to about 3 days. In embodiments, the second period of time is between about 1 day to about 2 days. [0072] In embodiments, the second period of time is between 1 day to 32 days. In embodiments, the second period of time is between 2 days to 32 days. In embodiments, the second period of time is between 3 days to 32 days. In embodiments, the second period of time is between 4 days to 32 days. In embodiments, the second period of time is between 5 days to 32 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT days. In embodiments, the second period of time is between 6 days to 32 days. In embodiments, the second period of time is between 7 days to 32 days. In embodiments, the second period of time is between 8 days to 32 days. In embodiments, the second period of time is between 9 days to 32 days. In embodiments, the second period of time is between 10 days to 32 days. In embodiments, the second period of time is between 11 days to 32 days. In embodiments, the second period of time is between 12 days to 32 days. In embodiments, the second period of time is between 13 days to 32 days. In embodiments, the second period of time is between 14 days to 32 days. In embodiments, the second period of time is between 15 days to 32 days. In embodiments, the second period of time is between 16 days to 32 days. In embodiments, the second period of time is between 17 days to 32 days. In embodiments, the second period of time is between 18 days to 32 days. In embodiments, the second period of time is between 19 days to 32 days. In embodiments, the second period of time is between 20 days to 32 days. In embodiments, the second period of time is between 21 days to 32 days. In embodiments, the second period of time is between 22 days to 32 days. In embodiments, the second period of time is between 23 days to 32 days. In embodiments, the second period of time is between 24 days to 32 days. In embodiments, the second period of time is between 25 days to 32 days. In embodiments, the second period of time is between 26 days to 32 days. In embodiments, the second period of time is between 27 days to 32 days. In embodiments, the second period of time is between 28 days to 32 days. In embodiments, the second period of time is between 29 days to 32 days. In embodiments, the second period of time is between 30 days to 32 days. In embodiments, the second period of time is between 31 days to 32 days. [0073] In embodiments, the second period of time is between 1 day to 31 days. In embodiments, the second period of time is between 1 day to 30 days. In embodiments, the second period of time is between 1 day to 29 days. In embodiments, the second period of time is between 1 day to 28 days. In embodiments, the second period of time is between 1 day to 27 days. In embodiments, the second period of time is between 1 day to 26 days. In embodiments, the second period of time is between 1 day to 25 days. In embodiments, the second period of time is between 1 day to 24 days. In embodiments, the second period of time is between 1 day to 23 days. In embodiments, the second period of time is between 1 day to 22 days. In embodiments, the second PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT period of time is between 1 day to 21 days. In embodiments, the second period of time is between 1 day to 20 days. In embodiments, the second period of time is between 1 day to 19 days. In embodiments, the second period of time is between 1 day to 18 days. In embodiments, the second period of time is between 1 day to 17 days. In embodiments, the second period of time is between 1 day to 16 days. In embodiments, the second period of time is between 1 day to 15 days. In embodiments, the second period of time is between 1 day to 14 days. In embodiments, the second period of time is between 1 day to 13 days. In embodiments, the second period of time is between 1 day to 12 days. In embodiments, the second period of time is between 1 day to 11 days. In embodiments, the second period of time is between 1 day to 10 days. In embodiments, the second period of time is between 1 day to 9 days. In embodiments, the second period of time is between 1 day to 8 days. In embodiments, the second period of time is between 1 day to 7 days. In embodiments, the second period of time is between 1 day to 6 days. In embodiments, the second period of time is between 1 day to 5 days. In embodiments, the second period of time is between 1 day to 4 days. In embodiments, the second period of time is between 1 day to 3 days. In embodiments, the second period of time is between 1 day to 2 days. [0074] In embodiments, step (d) includes dynamic culturing of the cultured cells followed by static culturing of the cultured cells. In embodiments, the dynamic culturing is for about 1 day to about 5 days. In embodiments, the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days. In embodiments, the dynamic culturing is for about 1 day. In embodiments, the dynamic culturing is for about 2 days. In embodiments, the dynamic culturing is for about 3 days. In embodiments, the dynamic culturing is for about 4 days. In embodiments, the dynamic culturing is for about 5 days. In embodiments, the dynamic culturing is for 1 day, 2 days, 3 days, 4 days, or 5 days. In embodiments, the dynamic culturing is for 1 day. In embodiments, the dynamic culturing is for 2 days. In embodiments, the dynamic culturing is for 3 days. In embodiments, the dynamic culturing is for 4 days. In embodiments, the dynamic culturing is for 5 days. [0075] In embodiments, the static culturing is for about 3 days to about 27 days. In embodiments, the static culturing is for about 4 days to about 27 days. In embodiments, the static PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT culturing is for about 5 days to about 27 days. In embodiments, the static culturing is for about 6 days to about 27 days. In embodiments, the static culturing is for about 7 days to about 27 days. In embodiments, the static culturing is for about 8 days to about 27 days. In embodiments, the static culturing is for about 9 days to about 27 days. In embodiments, the static culturing is for about 10 days to about 27 days. In embodiments, the static culturing is for about 11 days to about 27 days. In embodiments, the static culturing is for about 12 days to about 27 days. In embodiments, the static culturing is for about 13 days to about 27 days. In embodiments, the static culturing is for about 14 days to about 27 days. In embodiments, the static culturing is for about 15 days to about 27 days. In embodiments, the static culturing is for about 16 days to about 27 days. In embodiments, the static culturing is for about 17 days to about 27 days. In embodiments, the static culturing is for about 18 days to about 27 days. In embodiments, the static culturing is for about 19 days to about 27 days. In embodiments, the static culturing is for about 20 days to about 27 days. In embodiments, the static culturing is for about 21 days to about 27 days. In embodiments, the static culturing is for about 22 days to about 27 days. In embodiments, the static culturing is for about 23 days to about 27 days. In embodiments, the static culturing is for about 24 days to about 27 days. In embodiments, the static culturing is for about 25 days to about 27 days. In embodiments, the static culturing is for about 26 days to about 27 days. [0076] In embodiments, the static culturing is for about 3 days to about 26 days. In embodiments, the static culturing is for about 3 days to about 25 days. In embodiments, the static culturing is for about 3 days to about 24 days. In embodiments, the static culturing is for about 3 days to about 23 days. In embodiments, the static culturing is for about 3 days to about 22 days. In embodiments, the static culturing is for about 3 days to about 21 days. In embodiments, the static culturing is for about 3 days to about 20 days. In embodiments, the static culturing is for about 3 days to about 19 days. In embodiments, the static culturing is for about 3 days to about 18 days. In embodiments, the static culturing is for about 3 days to about 17 days. In embodiments, the static culturing is for about 3 days to about 16 days. In embodiments, the static culturing is for about 3 days to about 15 days. In embodiments, the static culturing is for about 3 days to about 14 days. In embodiments, the static culturing is for about 3 days to about 13 days. In embodiments, PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT the static culturing is for about 3 days to about 12 days. In embodiments, the static culturing is for about 3 days to about 11 days. In embodiments, the static culturing is for about 3 days to about 10 days. In embodiments, the static culturing is for about 3 days to about 9 days. In embodiments, the static culturing is for about 3 days to about 8 days. In embodiments, the static culturing is for about 3 days to about 7 days. In embodiments, the static culturing is for about 3 days to about 6 days. In embodiments, the static culturing is for about 3 days to about 5 days. In embodiments, the static culturing is for about 3 days to about 4 days. [0077] In embodiments, the static culturing is for 3 days to 27 days. In embodiments, the static culturing is for 4 days to 27 days. In embodiments, the static culturing is for 5 days to 27 days. In embodiments, the static culturing is for 6 days to 27 days. In embodiments, the static culturing is for 7 days to 27 days. In embodiments, the static culturing is for 8 days to 27 days. In embodiments, the static culturing is for 9 days to 27 days. In embodiments, the static culturing is for 10 days to 27 days. In embodiments, the static culturing is for 11 days to 27 days. In embodiments, the static culturing is for 12 days to 27 days. In embodiments, the static culturing is for 13 days to 27 days. In embodiments, the static culturing is for 14 days to 27 days. In embodiments, the static culturing is for 15 days to 27 days. In embodiments, the static culturing is for 16 days to 27 days. In embodiments, the static culturing is for 17 days to 27 days. In embodiments, the static culturing is for 18 days to 27 days. In embodiments, the static culturing is for 19 days to 27 days. In embodiments, the static culturing is for 20 days to 27 days. In embodiments, the static culturing is for 21 days to 27 days. In embodiments, the static culturing is for 22 days to 27 days. In embodiments, the static culturing is for 23 days to 27 days. In embodiments, the static culturing is for 24 days to 27 days. In embodiments, the static culturing is for 25 days to 27 days. In embodiments, the static culturing is for 26 days to 27 days. [0078] In embodiments, the static culturing is for 3 days to 26 days. In embodiments, the static culturing is for 3 days to 25 days. In embodiments, the static culturing is for 3 days to 24 days. In embodiments, the static culturing is for 3 days to 23 days. In embodiments, the static culturing is for 3 days to 22 days. In embodiments, the static culturing is for 3 days to 21 days. In embodiments, the static culturing is for 3 days to 20 days. In embodiments, the static culturing is PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT for 3 days to 19 days. In embodiments, the static culturing is for 3 days to 18 days. In embodiments, the static culturing is for 3 days to 17 days. In embodiments, the static culturing is for 3 days to 16 days. In embodiments, the static culturing is for 3 days to 15 days. In embodiments, the static culturing is for 3 days to 14 days. In embodiments, the static culturing is for 3 days to 13 days. In embodiments, the static culturing is for 3 days to 12 days. In embodiments, the static culturing is for 3 days to 11 days. In embodiments, the static culturing is for 3 days to 10 days. In embodiments, the static culturing is for 3 days to 9 days. In embodiments, the static culturing is for 3 days to 8 days. In embodiments, the static culturing is for 3 days to 7 days. In embodiments, the static culturing is for 3 days to 6 days. In embodiments, the static culturing is for 3 days to 5 days. In embodiments, the static culturing is for 3 days to 4 days. [0079] In embodiments, the maturation cocktail includes at least one exogenous cytokine, wherein the at least one exogenous cytokine includes GM-CSF, interleukin 4 (IL-4), interferon embodiments, the at least one exogenous cytokine includes GM-CSF. In embodiments, the at least one exogenous cytokine includes interleukin 4 (IL-4). In embodiments, the at least one one exogenous cytokine includes prostaglandin E2 (PGE-2). In embodiments, the at least one exogenous cytokine includes polyinosinic-polycytidylic acid (poly(I:C)). In embodiments, the at exogenous cytokine includes CD40L. In embodiments, one or more of the recited cytokines may be expressly excluded. [0080] In embodiments, the maturation cocktail includes GM-CSF and IL-4. In embodiments, PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0081] In embodiments, an mDC of the population of mDCs in step (e) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express one or more of CD14, TRA160, and SSEA-5. In embodiments, an mDC of the population of mDCs in step (e) expresses CD40. In embodiments, an mDC of the population of mDCs in step (e) expresses CD45. In embodiments, an mDC of the population of mDCs in step (e) expresses CD83. In embodiments, an mDC of the population of mDCs in step (e) expresses CD86. In embodiments, an mDC of the population of mDCs in step (e) expresses HLA-DR. In embodiments, an mDC of the population of mDCs in step (e) expresses HLA-A. In embodiments, an mDC of the population of mDCs in step (e) expresses CCR7. In embodiments, an mDC of the population of mDCs in step (e) expresses CD141. In embodiments, an mDC of the population of mDCs in step (e) expresses CD11c. In embodiments, an mDC of the population of mDCs in step (e) expresses CD45 and CD83. [0082] In embodiments, an mDC of the population of mDCs in step (e) does not express CD14. In embodiments, an mDC of the population of mDCs in step (e) does not express TRA160. In embodiments, an mDC of the population of mDCs in step (e) does not express SSEA-5. In embodiments, an mDC of the population of mDCs in step (e) does not express TRA160 and SSEA-5. [0083] In embodiments, an mDC of the population of mDCs in step (e) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express D14, CD45, TRA160, and SSEA-5. In embodiments, an mDC of the population of mDCs in step (e) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express D14, TRA160, and SSEA-5. [0084] In embodiments, at least 70% of the population of mDCs in step (e) express CD45. In embodiments, at least 75% of the population of mDCs in step (e) express CD45. In embodiments, at least 80% of the population of mDCs in step (e) express CD45. In embodiments, at least 85% of the population of mDCs in step (e) express CD45. In embodiments, at least 90% of the population of mDCs in step (e) express CD45. In embodiments, at least 95% of the population of mDCs in step (e) express CD45. In embodiments, at least 96% of the population of mDCs in step PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT (e) express CD45. In embodiments, at least 97% of the population of mDCs in step (e) express CD45. In embodiments, at least 98% of the population of mDCs in step (e) express CD45. In embodiments, at least 99% of the population of mDCs in step (e) express CD45. [0085] In embodiments, at least 70% of the population of mDCs in step (e) express CD83. In embodiments, at least 75% of the population of mDCs in step (e) express CD83. In embodiments, at least 80% of the population of mDCs in step (e) express CD83. In embodiments, at least 85% of the population of mDCs in step (e) express CD83. In embodiments, at least 90% of the population of mDCs in step (e) express CD83. In embodiments, at least 95% of the population of mDCs in step (e) express CD83. In embodiments, at least 96% of the population of mDCs in step (e) express CD83. In embodiments, at least 97% of the population of mDCs in step (e) express CD83. In embodiments, at least 98% of the population of mDCs in step (e) express CD83. In embodiments, at least 99% of the population of mDCs in step (e) express CD83. [0086] In embodiments, at least 70% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 75% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 80% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 85% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 90% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 95% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 96% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 97% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 98% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. In embodiments, at least 99% of the population of mDCs in step (e) express CD86, CD40, or HLA- A. [0087] In embodiments, at least 70% of the population of mDCs in step (e) express CD86. In embodiments, at least 75% of the population of mDCs in step (e) express CD86. In embodiments, at least 80% of the population of mDCs in step (e) express CD86. In embodiments, at least 85% of the population of mDCs in step (e) express CD86. In embodiments, at least 90% of the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT population of mDCs in step (e) express CD86. In embodiments, at least 95% of the population of mDCs in step (e) express CD86. In embodiments, at least 96% of the population of mDCs in step (e) express CD86. In embodiments, at least 97% of the population of mDCs in step (e) express CD86. In embodiments, at least 98% of the population of mDCs in step (e) express CD86. In embodiments, at least 99% of the population of mDCs in step (e) express CD86. [0088] In embodiments, at least 70% of the population of mDCs in step (e) express CD40. In embodiments, at least 75% of the population of mDCs in step (e) express CD40. In embodiments, at least 80% of the population of mDCs in step (e) express CD40. In embodiments, at least 85% of the population of mDCs in step (e) express CD40. In embodiments, at least 90% of the population of mDCs in step (e) express CD40. In embodiments, at least 95% of the population of mDCs in step (e) express CD40. In embodiments, at least 96% of the population of mDCs in step (e) express CD40. In embodiments, at least 97% of the population of mDCs in step (e) express CD40. In embodiments, at least 98% of the population of mDCs in step (e) express CD40. In embodiments, at least 99% of the population of mDCs in step (e) express CD40. [0089] In embodiments, at least 70% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 75% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 80% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 85% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 90% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 95% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 96% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 97% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 98% of the population of mDCs in step (e) express HLA-A. In embodiments, at least 99% of the population of mDCs in step (e) express HLA-A. [0090] In embodiments, at least 20% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 25% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 30% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 35% of the population of mDCs in step (e) express HLA-DR. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, at least 40% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 45% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 50% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 55% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 60% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 65% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 70% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 75% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 80% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 85% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 90% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 95% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 96% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 97% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 98% of the population of mDCs in step (e) express HLA-DR. In embodiments, at least 99% of the population of mDCs in step (e) express HLA-DR. [0091] In embodiments, less than 20% of the population of mDCs in step (e) express CD14. In embodiments, less than 19% of the population of mDCs in step (e) express CD14. In embodiments, less than 18% of the population of mDCs in step (e) express CD14. In embodiments, less than 17% of the population of mDCs in step (e) express CD14. In embodiments, less than 16% of the population of mDCs in step (e) express CD14. In embodiments, less than 15% of the population of mDCs in step (e) express CD14. In embodiments, less than 14% of the population of mDCs in step (e) express CD14. In embodiments, less than 13% of the population of mDCs in step (e) express CD14. In embodiments, less than 12% of the population of mDCs in step (e) express CD14. In embodiments, less than 11% of the population of mDCs in step (e) express CD14. In embodiments, less than 10% of the population of mDCs in step (e) express CD14. In embodiments, less than 9% of the population of mDCs in step (e) express CD14. In embodiments, less than 8% of the population of mDCs in step (e) express CD14. In embodiments, less than 7% PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT of the population of mDCs in step (e) express CD14. In embodiments, less than 6% of the population of mDCs in step (e) express CD14. In embodiments, less than 5% of the population of mDCs in step (e) express CD14. In embodiments, less than 4% of the population of mDCs in step (e) express CD14. In embodiments, less than 3% of the population of mDCs in step (e) express CD14. In embodiments, less than 2% of the population of mDCs in step (e) express CD14. In embodiments, less than 1% of the population of mDCs in step (e) express CD14. [0092] In embodiments, less than 20% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 19% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 18% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 17% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 16% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 15% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 14% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 13% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 12% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 11% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 10% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 9% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 8% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 7% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 6% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 5% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 4% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 3% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 2% of the population of mDCs in step (e) are CD45+ and CD83-. In embodiments, less than 1% of the population of mDCs in step (e) are CD45+ and CD83-. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0093] In embodiments, the third period of time is between about 1 day to about 10 days. In embodiments, the third period of time is between about 2 days to about 10 days. In embodiments, the third period of time is between about 3 days to about 10 days. In embodiments, the third period of time is between about 4 days to about 10 days. In embodiments, the third period of time is between about 5 days to about 10 days. In embodiments, the third period of time is between about 6 days to about 10 days. In embodiments, the third period of time is between about 7 days to about 10 days. In embodiments, the third period of time is between about 8 days to about 10 days. In embodiments, the third period of time is between about 9 days to about 10 days. [0094] In embodiments, the third period of time is between about 1 day to about 9 days. In embodiments, the third period of time is between about 1 day to about 8 days. In embodiments, the third period of time is between about 1 day to about 7 days. In embodiments, the third period of time is between about 1 day to about 6 days. In embodiments, the third period of time is between about 1 day to about 5 days. In embodiments, the third period of time is between about 1 day to about 4 days. In embodiments, the third period of time is between about 1 day to about 3 days. In embodiments, the third period of time is between about 1 day to about 2 days. [0095] In embodiments, the third period of time is between 1 day to 10 days. In embodiments, the third period of time is between 2 days to 10 days. In embodiments, the third period of time is between 3 days to 10 days. In embodiments, the third period of time is between 4 days to 10 days. In embodiments, the third period of time is between 5 days to 10 days. In embodiments, the third period of time is between about 6 days to about 10 days. In embodiments, the third period of time is between 7 days to 10 days. In embodiments, the third period of time is between 8 days to 10 days. In embodiments, the third period of time is between 9 days to 10 days. [0096] In embodiments, the third period of time is between 1 day to 9 days. In embodiments, the third period of time is between 1 day to 8 days. In embodiments, the third period of time is between 1 day to 7 days. In embodiments, the third period of time is between 1 day to 6 days. In embodiments, the third period of time is between 1 day to 5 days. In embodiments, the third period of time is between 1 day to 4 days. In embodiments, the third period of time is between 1 day to 3 days. In embodiments, the third period of time is between 1 day to 2 days. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0097] In embodiments, step (e) includes static culturing of the population of iDCs. In embodiments, the static culturing is for about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days. In embodiments, the static culturing is for about 4 days. In embodiments, the static culturing is for about 5 days. In embodiments, the static culturing is for about 6 days. In embodiments, the static culturing is for about 7 days. In embodiments, the static culturing is for about 8 days. In embodiments, the static culturing is for about 9 days. In embodiments, the static culturing is for about 10 days. In embodiments, the static culturing is for 4 days. In embodiments, the static culturing is for 5 days. In embodiments, the static culturing is for 6 days. In embodiments, the static culturing is for 7 days. In embodiments, the static culturing is for 8 days. In embodiments, the static culturing is for 9 days. In embodiments, the static culturing is for 10 days. [0098] In embodiments, any one of or both of steps (d) and (e) are performed under serum free conditions. In embodiments, step (d) is performed under serum free conditions. In embodiments, step (e) is performed under serum free conditions. In embodiments, steps (d) and (e) are performed under serum free conditions. In embodiments, any one of or both of steps (d) and (e) are performed in the absence of feeder cells. In embodiments, step (d) is performed in the absence of feeder cells. In embodiments, step (e) is performed in the absence of feeder cells. In embodiments, steps (d) and (e) are performed in the absence of feeder cells. In embodiments, any one of or both of steps (d) and (e) are performed in the absence of stromal cells. In embodiments, step (d) is performed in the absence of stromal cells. In embodiments, step (e) is performed in the absence of stromal cells. In embodiments, steps (d) and (e) are performed in the absence of stromal cells. [0099] In embodiments, steps (a), (b), and (c) are repeated after step (c), prior to step (d). In embodiments, steps (a), (b) and (c) are each repeated three times prior to step (d). In embodiments, steps (a), (b) and (c) combined are about three days to about five days. In embodiments, steps (a), (b) and (c) combined are about three days. In embodiments, steps (a), (b) and (c) combined are about four days. In embodiments, steps (a), (b) and (c) combined are about five days. In embodiments, steps (a), (b) and (c) combined are three days. In embodiments, steps PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT (a), (b) and (c) combined are four days. In embodiments, steps (a), (b) and (c) combined are five days. [0100] In embodiments, the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in steps (a) through (c). In embodiments, the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in step (a). In embodiments, the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in step (b). In embodiments, the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in step (c). In embodiments, at least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. In embodiments, at least about 80% of the undifferentiated and pluripotent cells express SSEA-5. In embodiments, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. In embodiments, at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog. In embodiments, at least about 70% of the undifferentiated and pluripotent cells express Oct-4. In embodiments, at least about 70% of the undifferentiated and pluripotent cells express Nanog. In embodiments, at least about 80% of the undifferentiated and pluripotent cells express SSEA-5, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60, at least about 70% of the undifferentiated and pluripotent cells express Oct-4, and at least about 70% of the undifferentiated and pluripotent cells express Nanog. [0101] In embodiments, step (b) further includes changing the growth medium to remove accumulated lactate. In embodiments, the growth medium includes Y-27632. In embodiments, the culturing in step (b) is dynamic. In embodiments, the cultured cells in step (b) include hematopoietic cell intermediates. In embodiments, the hematopoietic cell intermediates express CD34, CD90. In embodiments, the hematopoietic cell intermediates express CD34. In embodiments, the hematopoietic cell intermediates express CD90. In embodiments, the hematopoietic cell intermediates do not express CD38. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0102] In embodiments, the microcarrier includes one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof. In embodiments, the microcarrier includes polystyrene. In embodiments, the microcarrier includes cross-linked dextran. In embodiments, the microcarrier includes magnetic particles. In embodiments, the microcarrier includes microchips. In embodiments, the microcarrier includes cellulose. In embodiments, the microcarrier includes hydroxylated methacrylate. In embodiments, the microcarrier includes collagen. In embodiments, the microcarrier includes gelatin. In embodiments, the microcarrier includes polystyrene, plastic. In embodiments, the microcarrier includes glass. In embodiments, the microcarrier includes ceramic. In embodiments, the microcarrier includes silicon. In embodiments, one or more of the recited microcarriers may be expressly excluded. [0103] In embodiments, the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof. In embodiments, the microcarrier is spherical. In embodiments, the microcarrier is smooth. In embodiments, the microcarrier is macroporous. In embodiments, the microcarrier is rod-shaped. In embodiments, the microcarrier is not coated. In embodiments, the microcarrier is coupled with protamine or polylysine. In embodiments, the microcarrier is coupled with protamine. In embodiments, the microcarrier is coupled with polylysine. In embodiments, the microcarrier has a neutral charge. In embodiments, the microcarrier is negatively charged. In embodiments, the microcarrier is hydrophilic. In embodiments, one or more of the recited microcarriers may be expressly excluded. [0104] In embodiments, the ECM component comprises a matrigel, laminin, vitronectin, collagen, a derivative thereof, or any combination thereof. In embodiments, the ECM component includes a matrigel, a laminin, a vitronectin, a collagen, a derivative thereof, or any combination thereof. In embodiments, the ECM component includes a matrigel. In embodiments, the ECM component includes a laminin. In embodiments, the ECM component includes a vitronectin. In embodiments, the ECM component includes a collagen. In embodiments, the laminin is human PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT laminin. In embodiments, the human laminin is 511 E8 fragment. In embodiments, one or more of the recited ECM components may be expressly excluded. [0105] In embodiments, the mDCs are characterized by: (i) expression of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c, but not expressing CD14, CD45, TRA160, and SSEA-5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii). [0106] In embodiments, the mDCs are characterized by: (i) expression of at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c, but not expressing one or more of CD14, TRA160, and SSEA-5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii). [0107] In embodiments, the mDCs are characterized by expression of at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c. In embodiments, the mDCs are characterized by expression of CD40. In embodiments, the mDCs are characterized by expression of CD45. In embodiments, the mDCs are characterized by expression of CD83. In embodiments, the mDCs are characterized by expression of CD86. In embodiments, the mDCs are characterized by expression of HLA-DR. In embodiments, the mDCs are characterized by expression of HLA-A. In embodiments, the mDCs are characterized by expression of CD40. In embodiments, the mDCs are characterized by expression of CCR7. In embodiments, the mDCs are characterized by expression of CD141. In embodiments, the mDCs are characterized by expression of CD11c. In embodiments, the mDCs are characterized by expression of CD45 and CD83. [0108] In embodiments, the mDCs are characterized by not expressing one or more of CD14, TRA160, and SSEA-5. In embodiments, the mDCs are characterized by not expressing CD14. In embodiments, the mDCs are characterized by not expressing TRA160. In embodiments, the mDCs are characterized by not expressing SSEA-5. In embodiments, the mDCs are characterized by not expressing TRA160 and SSEA-5. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0109] In embodiments, the mDCs are characterized by exhibiting CCL19-induced migration. In embodiments, the mDCs are characterized by an ability to present an antigen at the cell surface. [0110] In embodiments, the mDCs are unable to replicate. In embodiments, the mDCs are irradiated to prevent replication. [0111] In embodiments, further including (f) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. In embodiments, further including (f) contacting the population of mDCs with an antigen. In embodiments, further including (f) contacting the population of mDCs with a nucleic acid encoding an antigen. In embodiments, further including (f) cryopreserving the population of mDCs. In embodiments, further including (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. In embodiments, further including (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen. In embodiments, further including (g) thawing the population of mDCs; and (h) contacting the population of mDCs with a nucleic acid encoding an antigen. In embodiments, the population of mDCs is contacted with two or more antigens. In embodiments, the antigen is a tumor antigen. In embodiments, the tumor antigen includes a tumor associated antigen (TAA). In embodiments, the antigen is a neoantigen. In embodiments, the antigen is a bacteria associated antigen. In embodiments, the antigen is a viral antigen. [0112] In embodiments, the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof. In embodiments, the tumor antigen includes a tumor-specific antigen (TSA). In embodiments, the nucleic acid includes an RNA. In embodiments, the RNA includes an mRNA encoding hTERT or fragment thereof. In embodiments, the fragment includes one or more immunogenic epitopes of hTERT. In embodiments, the RNA includes an mRNA encoding lysosomal-associated membrane protein 1 (LAMP1) or a fragment thereof. In embodiments, the RNA includes an mRNA encoding heat shock protein 96 (HSP96). In embodiments, the population of mDCs including the one or more antigens is cryopreserved. In embodiments, the cryopreserved population of mDCs including the one or more antigens is irradiated. In embodiments, the cryopreserved population of mDCs is irradiated to prevent replication. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT CELL COMPOSITIONS [0113] Antigen presenting cells of this invention are often referred to in this disclosure as “dendritic cells”. However, this is not meant to imply any morphological, phenotypic, or functional feature beyond what is explicitly required. The term is used to refer to cells that are phagocytic or can present antigen to T lymphocytes, falling within the general class of monocytes, macrophages, dendritic cells and the like, such as may be found circulating in the blood or lymph, or fixed in tissue sites. Phagocytic properties of a cell can be determined according to their ability to take up labeled antigen or small particulates. The ability of a cell to present antigen can be determined in a mixed lymphocyte reaction as described. Certain types of dendritic cells and antigen-presenting cells in the body are first identified in tissue sites such as the skin or the liver; but regardless of their origin, location, and developmental pathway, they are considered in the art to fall within the general category of hematopoietic cells. By analogy, the term dendritic cells used in this disclosure also fall in the broad category of hematopoietic cells, whether produced through the hematopoietic or direct paradigm framed earlier, or through a related or combined pathway. [0114] The putative role of hPS derived cells as antigen-presenting cells is provided in this disclosure as an explanation to facilitate the understanding of the reader. However, the theories expostulated here are not intended to limit the invention beyond what is explicitly required. The hPS derived cells of this invention may be used therapeutically regardless of their mode of action, as long as they achieve a desirable clinical benefit in a substantial proportion of patients treated. [0115] As such, antigen-presenting cells may include, but are not necessarily limited to, dendritic cells, macrophages, and B cells. In specific embodiments, the antigen-presenting cells are dendritic cells. [0116] In an aspect is provided a cell produced as provided herein including embodiments thereof, wherein the cell expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160 and SSEA-5. In embodiments, the cell expresses CD40. In embodiments, the cell expresses CD45. In embodiments, the cell expresses CD83. In embodiments, the cell expresses CD86. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the cell expresses HLA-DR. In embodiments, the cell expresses HLA-A. In embodiments, the cell expresses CD40. In embodiments, the cell expresses CCR7. In embodiments, the cell expresses CD45 and CD83. In embodiments, the cell does not express CD14. In embodiments, the cell does not express TRA160. In embodiments, the cell does not express SSEA-5. In embodiments, the cell does not express TRA160 and SSEA-5. [0117] In an aspect is provided a cell produced as provided herein including embodiments thereof, wherein the cell expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express CD14, CD83, TRA160 and SSEA-5. [0118] In another aspect is provided a cell produced as provided herein including embodiments thereof, wherein the cell (i) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA- A, CD40 and CCR7, but does not express one or more of CD14, TRA160, and SSEA-5; and (ii) includes the one or more antigens. [0119] In another aspect is provided a cell produced as provided herein including embodiments thereof, wherein the cell (i) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express CD14, CD83, TRA160, and SSEA-5; and (ii) includes the one or more antigens. CELL VACCINE COMPOSITIONS [0120] In an aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier. [0121] In an aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0122] In another aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. [0123] In embodiments, the one or more antigens include a tumor associated antigen (TAA), a tumor specific antigen (TSA), a neoantigen, or an antigen from an infectious organism. In embodiments, the one or more antigens include a tumor associated antigen (TAA). In embodiments, the one or more antigens include a tumor specific antigen (TSA). In embodiments, the one or more antigens include a neoantigen. In embodiments, the one or more antigens include an antigen from an infectious organism. In embodiments, the TAA includes a human telomerase reverse transcriptase (hTERT) or fragment thereof. In embodiments, the TAA includes a lysosomal-associated membrane protein 1 (LAMP1). In embodiments, the TAA includes a fusion protein. In embodiments, the TAA is an hTERT-LAMP1. In embodiments, the TAA includes HSP96. In embodiments, the TAA includes hTERT-LAMP1 and HSP96. In embodiments, the one or more antigens are not endogenous. In embodiments, the one or more antigens are encoded by a nucleic acid. [0124] In embodiments, the nucleic acid is introduced into the cell via electroporation, transfection, transduction. In embodiments, the nucleic acid is introduced into the cell via electroporation. In embodiments, the electroporation includes high throughput flow electroporation. In embodiments, the nucleic acid is introduced into the cell via transfection. In embodiments, the transfection includes chemical-based transfection. In embodiments, the chemical-based transfection includes a lipid-based transfection agent. In embodiments, the lipid- based transfection agent is a positively charged (e.g., cationic) lipid-based transfection agent. In embodiments, the chemical-based transfection includes calcium phosphate (e.g., Ca3(PO4)2). In embodiments, the nucleic acid is introduced into the cell via transduction. In embodiments, the transduction includes a viral vector. In embodiments, the viral vector is an Adenoviral vector, an Adeno-associated virus (AAV) vector, or a lentiviral vector. In embodiments, the viral vector is PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT an Adenoviral vector. In embodiments, the viral vector is an Adeno-associated virus (AAV) vector. In embodiments, the viral vector is a lentiviral vector. In embodiments, the nucleic acid is mRNA or DNA. In embodiments, the nucleic acid is mRNA. In embodiments, the nucleic acid is DNA. [0125] In another aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. [0126] In another aspect is provided a cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine including: (a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) including a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. [0127] In embodiments, the nucleic acid comprises a DNA. In embodiments, the DNA is under the control of a promoter. In embodiments, the promoter is an inducible promoter. In embodiments, further including (c) an inducing agent. In embodiments, the inducing agent is desferrioxamine. In embodiments, the inducing agent is tetracycline. In embodiments, the agent is picolinic acid. In embodiments, the inducing agent is desferrioxamine. In embodiments, the nucleic acid includes an RNA. In embodiments, the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent. [0128] In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.6 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.7 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.8 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the cell vaccine includes between about 0.9 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.0 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.1 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.2 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.3 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.4 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.5 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.6 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.7 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.8 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.9 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 2.0 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 2.1 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 2.2 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 2.3 x 107 dendritic cells to about 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 2.4 x 107 dendritic cells to about 2.5 x 107 dendritic cells. [0129] In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 2.4 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 2.3 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 2.2 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 2.1 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.9 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.8 x 107 dendritic cells. In embodiments, the cell vaccine includes PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT between about 0.5 x 107 dendritic cells to about 1.7 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.6 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.4 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.3 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.2 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.1 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 1.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 0.9 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 0.8 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 0.7 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 0.5 x 107 dendritic cells to about 0.6 x 107 dendritic cells. [0130] In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.6 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.7 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.8 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.9 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.0 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.1 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.2 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.3 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.4 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.5 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.6 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.7 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.8 x 107 dendritic cells PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.9 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 2.0 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 2.1 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 2.2 x 107 dendritic cells to 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 2.3 x 107 dendritic cells to abot 2.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 2.4 x 107 dendritic cells to 2.5 x 107 dendritic cells. [0131] In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 2.4 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to about 2.3 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 2.2 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 2.1 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.9 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.8 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.7 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.6 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.4 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.3 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.2 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.1 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 1.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 0.9 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 0.8 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 0.7 x 107 dendritic cells. In embodiments, the cell vaccine includes between 0.5 x 107 dendritic cells to 0.6 x 107 dendritic cells. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0132] In embodiments, the cell vaccine preferably includes between about 1,0 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.1 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.2 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.3 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.4 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.5 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.6 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.7 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.8 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1.9 x 107 dendritic cells to about 2.0 x 107 dendritic cells. [0133] In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.9 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.8 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.7 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.6 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.4 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.3 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.2 x 107 dendritic cells. In embodiments, the cell vaccine includes between about 1,0 x 107 dendritic cells to about 1.1 x 107 dendritic cells. [0134] In embodiments, the cell vaccine preferably includes between 1,0 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.1 x 107 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.2 x 107 dendritic cells to about 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.3 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.4 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.5 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.6 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.7 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.8 x 107 dendritic cells to 2.0 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1.9 x 107 dendritic cells to 2.0 x 107 dendritic cells. [0135] In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.9 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.8 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.7 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.6 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.5 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.4 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.3 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.2 x 107 dendritic cells. In embodiments, the cell vaccine includes between 1,0 x 107 dendritic cells to 1.1 x 107 dendritic cells. [0136] In embodiments, the cell vaccine more preferably includes about 1.0 x 107 dendritic cells. In embodiments, the cell vaccine includes about 1.0 x 107 dendritic cells. In embodiments, the cell vaccine more preferably includes 1.0 x 107 dendritic cells. In embodiments, the cell vaccine includes 1.0 x 107 dendritic cells. [0137] In embodiments, the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder. In embodiments, the disease or disorder is cancer. In embodiments, the disease or disorder is an immune disease or disorder. In embodiments, the disease or disorder is an infectious disease or disorder. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0138] In embodiments, the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer. In embodiments, the cancer is breast cancer. In embodiments, the cancer is ovarian cancer. In embodiments, the cancer is colon cancer. In embodiments, the cancer is prostate cancer. In embodiments, the cancer is bone cancer. In embodiments, the cancer is colorectal cancer. In embodiments, the cancer is gastric cancer. In embodiments, the cancer is lymphoma. In embodiments, the cancer is malignant melanoma. In embodiments, the cancer is liver cancer. In embodiments, the cancer is small cell lung cancer. In embodiments, the cancer is non-small cell lung cancer. In embodiments, the cancer is pancreatic cancer. In embodiments, the cancer is thyroid cancers. In embodiments, the cancer is kidney cancer. In embodiments, the cancer is cancer of the bile duct. In embodiments, the cancer is brain cancer. In embodiments, the cancer is cervical cancer. In embodiments, the cancer is maxillary sinus cancer. In embodiments, the cancer is bladder cancer. In embodiments, the cancer is esophageal cancer. In embodiments, the cancer is Hodgkin's disease. In embodiments, the cancer is adrenocortical cancer. [0139] In embodiments, further comprising one or more additional therapeutic agents. In embodiments, the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents. In embodiments, the one or more additional therapeutic agents is chemotherapy. In embodiments, the one or more additional therapeutic agents is adjuvants. In embodiments, the one or more additional therapeutic agents is radiation. In embodiments, the one or more additional therapeutic agents is gene therapy. In embodiments, the one or more additional therapeutic agents is surgery. In embodiments, the one or more additional therapeutic agents is one or more anti-inflammatory agents. In embodiments, the one or more additional therapeutic agents is one or more immunomodulatory agents. In embodiments, the one or more antigens is identified from the subject. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0140] In embodiments, the cell vaccine is formulated to be administered to the subject directly after thawing. In embodiments, directly administering includes that dead cells are not removed from the composition prior to administration to the subject. In embodiments, the cell vaccine is formulated to be administered without washing or reconstitution. In embodiments, the cell vaccine is formulated to be administered without washing. In embodiments, the cell vaccine is formulated to be administered without reconstitution. [0141] In embodiments, the cell vaccine is formulated for administration within about 4 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 3.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 3 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 2.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 2 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 1.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within about 1 hour of thawing. In embodiments, the cell vaccine is formulated for administration within about 30 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within about 20 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within about 10 minutes of thawing. [0142] In embodiments, the cell vaccine is formulated for administration within 4 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 3.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 3 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 2.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 2 hours of thawing. In embodiments, the cell vaccine is formulated for administration within 1.5 hours of thawing. In embodiments, the cell vaccine is formulated for administration within o1 hour of thawing. In embodiments, the cell vaccine is formulated for administration within 30 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within 20 minutes of thawing. In embodiments, the cell vaccine is formulated for administration within 10 minutes of thawing. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0143] In embodiments, the cell vaccine is formulated for administration more than 4 hours after thawing. [0144] In embodiments, the cell vaccine is formulated for administration within about 24 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 23 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 22 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 21 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 20 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 19 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 18 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 17 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 16 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 15 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 14 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 13 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 12 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 11 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 10 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 9 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 8 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 7 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 6 hours after thawing. In embodiments, the cell vaccine is formulated for administration within about 5 hours after thawing. [0145] In embodiments, the cell vaccine is formulated for administration within 24 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 23 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 22 hours after PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT thawing. In embodiments, the cell vaccine is formulated for administration within 21 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 20 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 19 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 18 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 17 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 16 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 15 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 14 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 13 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 12 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 11 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 10 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 9 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 8 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 7 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 6 hours after thawing. In embodiments, the cell vaccine is formulated for administration within 5 hours after thawing. METHODS OF MANUFACTURING [0146] In another aspect is provided a method of manufacturing a cell vaccine for treating cancer, the method including: (a) obtaining one or more mature dendritic cells (mDCs) according to the method provided herein including embodiments thereof; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer. [0147] In embodiments, further including (c) providing one or more additional therapeutic agents. In embodiments, optionally the one or more mDCs are cryopreserved and thawed before step (b). In embodiments, the one or more antigens is one or more tumor antigens. In embodiments, the one or more tumor antigens includes a TAA. In embodiments, the TAA PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT includes hTERT. In embodiments, the TAA is hTERT. In embodiments, the TAA includes LAMP1. In embodiments, the TAA is a fusion protein. In embodiments, the TAA is hTERT- LAMP1. In embodiments, the TAA includes HSP96. In embodiments, the TAA includes hTERT-LAMP1 and HSP96. In embodiments, the one or more tumor antigens includes a TSA. In embodiments, the one or more tumor antigens includes a neoantigen. In embodiments, the one or more nucleic acid molecules includes one or more RNA molecules. In embodiments, the one or more RNA molecules include one or more mRNA molecules. In embodiments, the one or more nucleic acid molecules includes on ore more DNA molecules. In embodiments, the one or more RNA molecules includes hTERT mRNA. In embodiments, the one or more RNA molecules includes hTERT-LAMP1 mRNA. In embodiments, the one or more RNA molecules includes HSP96 mRNA. [0148] In embodiments, the one or more nucleic acid molecules is introduced into the cell via electroporation, transfection, transduction. In embodiments, the one or more nucleic acid molecules is introduced into the cell via electroporation. In embodiments, the electroporation includes high throughput flow electroporation. In embodiments, the one or more nucleic acid molecules is introduced into the cell via transfection. In embodiments, the transfection includes chemical-based transfection. In embodiments, the chemical-based transfection includes a lipid- based transfection agent. In embodiments, the lipid-based transfection agent is a positively charged (e.g., cationic) lipid-based transfection agent. In embodiments, the chemical-based transfection includes calcium phosphate (e.g., Ca3(PO4)2). In embodiments, the one or more nucleic acid molecules is introduced into the cell via transduction. In embodiments, the transduction includes a viral vector. In embodiments, the viral vector is an Adenoviral vector, an Adeno-associated virus (AAV) vector, or a lentiviral vector. In embodiments, the viral vector is an Adenoviral vector. In embodiments, the viral vector is an Adeno-associated virus (AAV) vector. In embodiments, the viral vector is a lentiviral vector. [0149] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT is between about 0.6 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.7 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.8 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.9 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 1 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 2 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 3 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 4 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 5 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 6 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 7 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 8 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 9 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 10 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 11 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 12 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT molecules used for electroporation is between about 13 µg per million cells to about 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 14 µg per million cells to about 15 µg per million cells. [0150] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 14 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 13 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 12 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 11 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 10 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 9 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 8 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 7 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 6 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 5 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 4 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 3 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 2 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 1 µg per million cells. In embodiments, the concentration of the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 0.9 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 0.8 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 0.7 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between about 0.5 µg per million cells to about 0.6 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is preferably about 6 µg per million cells. [0151] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.6 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.7 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.8 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.9 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 1 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 2 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 3 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 4 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 5 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 6 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 7 µg per million cells PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 8 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 9 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 10 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 11 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 12 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 13 µg per million cells to 15 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 14 µg per million cells to 15 µg per million cells. [0152] In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 14 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 13 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 12 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 11 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 10 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 9 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 8 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 7 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 6 µg per million cells. In embodiments, the concentration of the one or more nucleic acid PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT molecules used for electroporation is between 0.5 µg per million cells to 5 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 4 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 3 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 2 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 1 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 0.9 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 0.8 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 0.7 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is between 0.5 µg per million cells to 0.6 µg per million cells. In embodiments, the concentration of the one or more nucleic acid molecules used for electroporation is preferably 6 µg per million cells. [0153] In embodiments, the manufacturing takes about 30 days to about 50 days. In embodiments, the manufacturing takes about 31 days to about 50 days. In embodiments, the manufacturing takes about 32 days to about 50 days. In embodiments, the manufacturing takes about 33 days to about 50 days. In embodiments, the manufacturing takes about 34 days to about 50 days. In embodiments, the manufacturing takes about 35 days to about 50 days. In embodiments, the manufacturing takes about 36 days to about 50 days. In embodiments, the manufacturing takes about 37 days to about 50 days. In embodiments, the manufacturing takes about 38 days to about 50 days. In embodiments, the manufacturing takes about 29 days to about 50 days. In embodiments, the manufacturing takes about 40 days to about 50 days. In embodiments, the manufacturing takes about 41 days to about 50 days. In embodiments, the manufacturing takes about 42 days to about 50 days. In embodiments, the manufacturing takes about 43 days to about 50 days. In embodiments, the manufacturing takes about 44 days to about 50 days. In embodiments, the manufacturing takes about 45 days to about 50 days. In PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT embodiments, the manufacturing takes about 46 days to about 50 days. In embodiments, the manufacturing takes about 47 days to about 50 days. In embodiments, the manufacturing takes about 48 days to about 50 days. In embodiments, the manufacturing takes about 49 days to about 50 days. [0154] In embodiments, the manufacturing takes about 30 days to about 49 days. In embodiments, the manufacturing takes about 30 days to about 48 days. In embodiments, the manufacturing takes about 30 days to about 47 days. In embodiments, the manufacturing takes about 30 days to about 46 days. In embodiments, the manufacturing takes about 30 days to about 45 days. In embodiments, the manufacturing takes about 30 days to about 44 days. In embodiments, the manufacturing takes about 30 days to about 43 days. In embodiments, the manufacturing takes about 30 days to about 42 days. In embodiments, the manufacturing takes about 30 days to about 41 days. In embodiments, the manufacturing takes about 30 days to about 40 days. In embodiments, the manufacturing takes about 30 days to about 39 days. In embodiments, the manufacturing takes about 30 days to about 38 days. In embodiments, the manufacturing takes about 30 days to about 37 days. In embodiments, the manufacturing takes about 30 days to about 36 days. In embodiments, the manufacturing takes about 30 days to about 35 days. In embodiments, the manufacturing takes about 30 days to about 34 days. In embodiments, the manufacturing takes about 30 days to about 33 days. In embodiments, the manufacturing takes about 30 days to about 33 days. In embodiments, the manufacturing takes about 30 days to about 31 days. [0155] In embodiments, the manufacturing takes 30 days to 50 days. In embodiments, the manufacturing takes 31 days to 50 days. In embodiments, the manufacturing takes 32 days to 50 days. In embodiments, the manufacturing takes 33 days to 50 days. In embodiments, the manufacturing takes 34 days to 50 days. In embodiments, the manufacturing takes 35 days to 50 days. In embodiments, the manufacturing takes 36 days to 50 days. In embodiments, the manufacturing takes 37 days to 50 days. In embodiments, the manufacturing takes about 38 days to 50 days. In embodiments, the manufacturing takes 29 days to 50 days. In embodiments, the manufacturing takes 40 days to 50 days. In embodiments, the manufacturing takes 41 days to 50 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT days. In embodiments, the manufacturing takes 42 days to 50 days. In embodiments, the manufacturing takes 43 days to 50 days. In embodiments, the manufacturing takes 44 days to 50 days. In embodiments, the manufacturing takes 45 days to 50 days. In embodiments, the manufacturing takes 46 days to 50 days. In embodiments, the manufacturing takes 47 days to 50 days. In embodiments, the manufacturing takes about 48 days to about 50 days. In embodiments, the manufacturing takes 49 days to 50 days. [0156] In embodiments, the manufacturing takes 30 days to 49 days. In embodiments, the manufacturing takes 30 days to 48 days. In embodiments, the manufacturing takes 30 days to 47 days. In embodiments, the manufacturing takes 30 days to 46 days. In embodiments, the manufacturing takes 30 days to 45 days. In embodiments, the manufacturing takes 30 days to 44 days. In embodiments, the manufacturing takes 30 days to 43 days. In embodiments, the manufacturing takes 30 days to 42 days. In embodiments, the manufacturing takes 30 days to 41 days. In embodiments, the manufacturing takes 30 days to 40 days. In embodiments, the manufacturing takes 30 days to 39 days. In embodiments, the manufacturing takes 30 days to 38 days. In embodiments, the manufacturing takes 30 days to 37 days. In embodiments, the manufacturing takes 30 days to 36 days. In embodiments, the manufacturing takes 30 days to 35 days. In embodiments, the manufacturing takes 30 days to 34 days. In embodiments, the manufacturing takes 30 days to 33 days. In embodiments, the manufacturing takes 30 days to 33 days. In embodiments, the manufacturing takes 30 days to 31 days. [0157] In embodiments, the one or more antigens is an antigen expressed by the cancer. In embodiments, the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. In embodiments, the cancer is non-small cell lung cancer (NSCLC) . In embodiments, the cancer is prostate cancer. In embodiments, the cancer is acute myelogenous leukemia. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT METHODS OF TREATMENT [0158] In another aspect is provided a method for treating or delaying progression of a disease or disorder in a subject, including administering to the subject a cell vaccine provided herein including embodiments thereof. In embodiments, the dendritic cells are allogeneic to the subject. [0159] In another aspect is provided a method for treating cancer, including administering to a subject having cancer a cell vaccine made by the method provided herein including embodiments thereof. [0160] In embodiments, the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. In embodiments, the cancer is non-small cell lung cancer (NSCLC). In embodiments, the cancer is prostate cancer. In embodiments, the cancer is acute myelogenous leukemia. EXAMPLES [0161] LIST OF ABBREVIATIONS Abbreviation Definition A AE Adverse Event
Figure imgf000059_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT or or
Figure imgf000060_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Example 1 [0162] LCT-VAC2 consists of mature dendritic cells (mDCs) derived from the H1 human embryonic stem cells (hESCs) line that have been transfected with mRNA coding for tumor- associated antigen hTERT/LAMP-1. [0163] The first in human clinical trial, conducted by Cancer Research United Kingdom (CRUK) in Great Britain, investigated VAC2 in HLA A*02:01 positive subjects with metastatic or locally advanced non-small cell lung cancer (NSCLC). The initial dose escalation clinical trial to be conducted in the United States will include similar subjects with refractory NSCLC. [0164] LCT-VAC2 Drug Product (DP) was cryopreserved as “Thaw-and Inject” formulation in CryoStor 5 (CS5) medium at a concentration of 6.25 x 106 cells per 500 µl per vial (Nunc Cryovial, Thermo-Fisher Scientific, 374086). The route of delivery was an intradermal (ID) injection, and the planned vaccination schedule utilized a cohort dose escalation scheme, starting with cohorts of subjects receiving either 1.25, 2.5, 5 or 10 million cells per administration weekly, for a total of six vaccinations. [0165] LCT-VAC2 is a novel allogeneic dendritic cell (DC) vaccine derived from H1 undifferentiated human embryonic stem cells (hESCs). LCT-VAC2 was produced by the stepwise differentiation of H1 hESCs into mature DCs (mDCs) which were then pulsed (electroporated) with messenger ribonucleic acid (mRNA) encoding the tumor associated antigen human telomerase reverse transcriptase (hTERT) fused to the cytoplasmic tail of lysosomal associated membrane protein 1 (LAMP-1), which facilitated human leukocyte antigen (HLA) II, as well as HLA I loading of hTERT. Both elements (hTERT and LAMP-1) were fused to the coding sequence for heat shock protein 96 (HSP96) to facilitate efficient shuttling of the translated protein to the endoplasmic reticulum. The electroporated mDCs were then cryopreserved and irradiated to eliminate the replication of any remaining cells with proliferative capability. The mDCs were engineered to present hTERT, a tumor-associated antigen found in most cancer cells, and LCT- VAC2 stimulated an immunogenic anti-tumor CD4+/CD8+ T cell response toward hTERT expressed on tumor cells. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0166] As described above, LCT-VAC2 was developed to target the telomerase enzyme since telomerase is expressed in >85% of human cancers and the safety and efficacy of this approach have been previously demonstrated. Telomerase is an established target to inhibit cancer growth since it is essential for maintaining the proliferative capacity and survival of tumor cells by preventing the shorting of the telomeres (Ouellette, Wright et al.2011). It is not usually expressed in normal tissues. Where it is observed (e.g., gastrointestinal tract and CD34+ hematopoietic cells), it is only transiently expressed at low levels. [0167] Brief History and Current Status of the Development Program [0168] The safety and immunogenicity of autologous DC immunotherapy has been demonstrated in many clinical trials in solid and liquid tumors (Su, Dannull et al.2005), including a previous clinical trial of VAC1 (Khoury, Collins et al.2010, Khoury, Collins et al.2017) which employed autologous DCs sourced from respective subjects. Safety data in the completed Phase 1 clinical trial of the allogeneic VAC2 in NSCLC confirmed that the vaccine appears well tolerated with an acceptable safety profile. While reports on tumor response to DC immunotherapy have been mixed to date and include a wide range of approaches, published meta-analyses indicate positive effects on overall survival, especially when applied in a minimal residual disease setting. [0169] Chemistry, Manufacturing and Controls [0170] The LCT-VAC2 manufacturing process, as outlined below, is an optimized differentiation process originated from the H1 hESCs line into mDCs, which are then electroporated with hTERT mRNA fused to the cytoplasmic tail (LAMP-1). Improvement of the process used to generate the original VAC2 utilized in the CRUK trial included: a scaled-up process for H1 expansion, enhanced in-process controls (IPC) during the differentiation process, and development of specific in-process and release tests. [0171] A specific IPC plan to monitor cell characteristics along the differentiation process was created based on hESCs-mDCs differentiation steps: hESCs pluripotent cells, hematopoietic progenitors, common myeloid precursors (CMP), monocytes, immature DC (iDCs), and finally, mature DCs (mDCs). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0172] The final LCT-VAC2 drug product was subjected to a panel of characterization assays developed to evaluate the cells’ viability, identity, and functionality. Altogether these activities led to a reproducible and tightly controlled manufacturing process and defined LCT-VAC2 drug product. [0173] Mature DCs (mDCs) were produced as part of LCT-VAC2 manufacturing from the hESCs by a differentiation process as an intermediate material. Characterization release tests of the intermediate material included evaluation of LCT-mDCs’ identity and functionality. Thus, LCT-mDCs were used as a source starting material in future applications by introduction of alternative mRNAs to induce different, specific immune responses. [0174] Preclinical Development [0175] To support use of VAC-2 in the CRUK study, a series of in vitro characterization assays was completed to demonstrate that VAC-2 had the characteristics of, and behaved biologically as, activated mDCs. Additionally, the mDCs were irradiated as an additional safety precaution, and the effectiveness of the irradiation demonstrated. Though the initial study in the UK started several years ago, there have been no reliable in vivo models developed since the initiation of that study that would allow for assessment of generated immune responses or potential anti-tumor effects. Humanized murine models exist (as detailed below), but due to minor-HLA mismatches, they are of questionable value in assessing dendritic cell vaccines. The safety profiles of the autologous VAC1 program, as well as other experimental DC vaccines, have also demonstrated that treatment is well tolerated, and adverse events (AEs) and serious adverse events (SAEs) have been as predicted based on the immune functional role of DCs. We did not observe any unexpected AEs or SAEs in the completed study of VAC2 in refractory NSCLC subjects in the UK. [0176] Clinical Development [0177] The initial starting dose was 4-fold less than previously used in the CRUK study of VAC2. The dose selection was aimed to bridge between the previous VAC2 clinical trial, which was well tolerated by all eight (8) subjects and the LCT-VAC2 product. Additional supply with an optimized dose concentration, which was not available for the completed study, allowed the PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT determination of an optimal dose to generate biological and immune activity following administration. The Phase 2a clinical trial with LCT-VAC2 was designed to evaluate the safety and tolerability of escalating target doses of LCT-VAC2 utilizing four (4) dose groups. LCT- VAC2 was given as six weekly intradermal (ID) injections in subjects with refractory Stage IIlB or IV NSCLC, who previously failed at least one approved immunotherapy regimen (e.g., PD-1 or PD-L1 checkpoint inhibitors or CTLA-4 receptor antibodies) and one platinum-based chemotherapy cycle, or a combination of both. [0178] SCIENTIFIC BACKGROUND AND CLINICAL RATIONALE [0179] Potential Role of LCT-VAC2 in Human Disease [0180] Novel immunotherapy agents, such as anti-CTLA4 and anti-PD-1 antibodies, as well as vaccines such as TG4010, have demonstrated that NSCLC is amenable to immunotherapy (Quoix, Ramlau et al.2011, Lynch, Bondarenko et al.2012, Reck, Bondarenko et al.2013, Quoix, Lena et al.2016, Chae, Arya et al.2018, De Giglio, Di Federico et al.2021, Mielgo-Rubio, Uribelarrea et al.2021). While these agents appear to improve outcome, they are only able to offer durable disease control to a subset of subjects (Chen, Fillmore et al.2014). The novel properties of LCT- VAC2 may address an unmet medical need via the use of defined HLA Class I and Class II potentiating activities. [0181] Dendritic cells are naturally able to process and present antigens, activating both CD4+ and CD8+ T cell responses, and thus functionally promote an inflammatory response against specific antigens, including tumor antigens. To date, there have been hundreds of clinical trials conducted focusing on the use of autologous or allogeneic DCs as a vaccination strategy for numerous oncology indications. The most frequently reported AEs associated with DCs vaccine strategies were flu-like symptoms, injection site reactions and injection site pain, which are anticipated results for a vaccination strategy designed to promote an inflammatory and memory immune response. Therefore, historical data suggest that DCs vaccination strategies are safe and generally well tolerated. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0182] hESCs-derived mature DCs migrate from the site of administration to the regional lymph nodes and are capable of stimulating T cell proliferation in response to viral and other antigens including the tumor antigen hTERT. These cells have phagocytic activity and release pro- inflammatory cytokines. Cell surface markers expressed on mature hESCs-derived DCs include CD83, CD86, HLA I and II. All these features are common properties of mature DCs and suggest that hESCs-derived DCs are functionally and phenotypically comparable with endogenous DCs. [0183] The antigen (hTERT) is a known tumor antigen and cancer target, which has been well- characterized as being overexpressed almost exclusively in a variety of tumors, including NSCLC. To date, dozens of studies have either targeted hTERT directly, or have used hTERT expression levels as an endpoint to determine efficacy of treatment and there have been no unexpected safety events of note. [0184] Sequences encoding LAMP-1 have been added to the hTERT to enable the presentation of endogenous protein by HLA II receptors; hTERT presented in this way elicits a CD4+ T cell response alongside the CD8+ response driven through HLA I. Such a dual response is desirable as the activated CD4+ cells can enhance the primary CD8+ response. [0185] Data from Completed VAC1 Autologous DC Phase 2 Clinical Trial [0186] As described above, a clinical trial was conducted that employed an autologous cellular immunotherapy approach (VAC1) administered to subjects with acute myeloid leukemia (AML) in a Phase 2 multicenter, open-label trial. Encouraging signs of increased overall survival and immune activity were observed and the AML data are shown in FIG.1. This study of autologous VAC1 vaccine demonstrated the safety, tolerability, immunogenicity, and clinical activity of the same TERT antigen product utilized in VAC2 for the NSCLC trial conducted by CRUK (i.e., telomerase-LAMP mRNA) (Khoury, Collins et al.2010, Khoury, Collins et al.2017) The described allogeneic LCT-VAC2 product under development currently builds upon the previous autologous VAC1 work by utilizing established pluripotent stem cell differentiation protocols to manufacture non-patient specific “off-the-shelf” mature DCs by means of what is designed to be a commercially scalable method of production with increased purity. A summary of the differences and similarities between the autologous VAC1 and VAC2 products is shown below in Table 1. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 1. Summary of the Product Characteristics for Autologous VAC1 and Allogeneic VAC2 Desi n characteristic Autolo ous Allo eneic Scientific rationale , d
Figure imgf000066_0001
[0187] Phase 1 Clinical Trial of Allogeneic VAC2 [0188] The Phase 1 trial of allogeneic VAC2 conducted in the UK, entitled, “A Cancer Research UK Phase 1 trial of AST-VAC2 (allogeneic dendritic cell vaccine) administered weekly via intradermal injection in subjects with advanced non-small cell lung cancer”, completed enrollment (N=8) in February 2022. The therapeutic vaccine was administered via two, equally split intradermal (ID) injections at a target dose of 1 x 107 cells on each vaccination day, administered weekly for 6 consecutive weeks. The target dose was 1 x 107 viable cells/day. [0189] In these 8 subjects, there were no unexpected safety events and no serious adverse events (SAEs) attributable to VAC2. Adverse events (AEs) reported at sites of VAC2 injection were mild, self-limiting and had no clinical sequelae. AEs reported by some of the subjects were suggestive of provocation or initiation of an adaptive immune response and included injection site PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT immunogenicity data in this study suggest the desired immune effects were generated in some VAC2 treated subjects. These data support further clinical investigation of VAC2. [0190] Clinical Rationale [0191] Lung cancer is the leading cause of cancer mortality among men and women and accounts for about one-fifth of all cancer-related deaths (Spira and Ettinger 2004). Lung cancer is broadly split into non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), with 80-85% classified as NSCLC. Even with remarkable recent advances in treatment, long-term survival rates for NSCLC and SCLC remain poor and new treatments are necessary. LCT-VAC2 may provide an additional treatment option for these subjects. [0192] The hTERT sequence (amino acids 168-1132) used to create VAC2, both in the completed CRUK study and the proposed trial, contains epitopes that are known to be presented by HLA A*02:01 (Lin, Guarnieri et al.1996). The HLA A*02:01 locus is expressed at high frequencies in the general population, including in approximately 46% of individuals of North European Caucasian ancestry (Morse, Coleman et al.1999, Eskelinen 2006). To ensure that the subjects enrolled in this trial are allogeneically compatible with the hTERT/DC system, and thus able to benefit from any therapeutic potential offered, they will be phenotyped prior to inclusion to ensure that they are also HLA A*02:01 positive. No attempt will be made to match subjects to the remaining alleles as it is hoped that this mismatch will induce a beneficial mixed leukocyte reaction. [0193] CHEMISTRY, MANUFACTURING AND CONTROLS [0194] H1 Cell Banking System [0195] H1 Master Cell Bank (MCB) Production [0196] The origin of the new H1 Master Cell Bank (MCB) was the H1 Bank Lot. No. MCBH101. MCBH101 was manufactured by Geron directly from the H1 Original Cell Bank (OCB) in 2009 (manufacturing of Geron’s MCBH101 Seed Bank is detailed in GR10-014 report). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0197] The production of H1 MCB (LCT-H1-001) was performed according to FIG.2 and involved the following steps - one vial of MCBH101 Passage 30 is thawed and seeded on rh- Laminin 521 (BioLamina, LN521) coated flasks in mTeSR plus (STEMCELL Technologies, 100- 0276). The cells were passaged using non-enzymatic dissociation reagent ‘ReLeSR’ (STEMCELL Technologies, 05872). The culture was expanded for 6 passages and evaluated for morphology, % confluency, and lactate concentration. The MCB was cryopreserved in CryoStor 10 cryopreservation solution (CS10, Biolife solutions, 210102) at 10 x 106 cells per 1 mL per 1 vial, using CryoMed controlled rate freezer. [0198] The characterization of Pilot LCT-H1-001 Master Cell Bank is summarized in Table 2. Table 2. Release Testing of PILOT LCT-H1-001 MCB Test Type Method Specification Result 5: : 5: g: t to
Figure imgf000068_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT ch
Figure imgf000069_0001
[0199] The extended biosafety testing of the Pilot LCT-H1-001 Master Cell Bank is presented in Table 3. Table 3. The Extended Adventitious Agents Tests Results of LCT-H1-001 MCB Test Type Specification Result [
Figure imgf000069_0002
g [0201] The production of H1 WCB was performed according to Table 3 and involved the following steps – one vial of LCT-H1-001 MCB was thawed and seeded on iMatrix-511 E8 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT (Matrixome, 892005) coated flasks in mTeSR plus (STEMCELL Technologies, 100-0276). The cells were passaged using non-enzymatic dissociation reagent ‘ReLeSR’ (STEMCELL Technologies, 05872). The culture was expanded for 4 passages and evaluated for morphology, % confluency, and lactate concentration. The WCB was cryopreserved in CryoStor 10 cryopreservation solution (CS10, Biolife solutions, 2010102) at 7 x 106 cells per 1 mL per 1 vial, using CryoMed controlled rate freezer. [0202] The planned characterization of H1 Working Cell Bank is summarized in Table 4. Table 4. Planned Release Testing of H1 WCB Quality Attribute Method Specification sm tly ead eat
Figure imgf000070_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0203] The planned extended biosafety testing of H1 WCB is presented in Table 5. Table 5. Planned Extended Adventitious Agents Tests of H1 WCB Test T e S ifi i
Figure imgf000071_0001
[0204] 5.1.3 Materials Used in Manufacturing of H1 MCB Table 6. Materials Used in Manufacturing of H1 Master Cell Bank Material Supplier #Cat Origin Step in the Manufacturing Pr 0
Figure imgf000071_0002
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0206] Five (5) years long-term stability (LTS) of H1 MCB test plan is on-going. The following quality attributes are tested during LTS assessment: Cell viability and strength (total viable cells/1 ml) Performance test for 2 passages: pluripotency biomarkers expression (%TRA-1- 60/SSEA5+ and %Oct4/Nanog+), morphology and population doubling Levels (PDL) Pluripotency of thawed cells (%TRA-1-60/SSEA5+ and %Oct4/Nanog+ expression) Sterility [0207] LCT VAC2 and LCT mDCs [0208] LCTVAC2 and LCT-FnDCr Process Overview [0209] LCT-VAC2 and LCT mDCs production process is described in FIG.4 and involved the following steps: hESCs thawing (WCB) and expansion prior to differentiation hESCs differentiation into monocytes and further into iDCs iDCs maturation into mDCs Harvesting mDCs The mDCs were then be processed by one of two options: o LCT-mDCs Cryopreservation of the mDCs o LCT-VAC2 Electroporation (EP) of mDCs with hTERT-LAMP1 mRNA Cryopreservation of the EP’d mDCs in a Thaw-and-Inject (TAI) formulation for long-term storage and used as needed X-ray irradiation of the cryopreserved EP’d mDCs [0210] H1 Expansion Prior to Differentiation [0211] One vial of H1 WCB (P30+6+4) was thawed and seeded on iMatrix-511 E8 (Matrixome, 892005) coated flasks in mTeSR plus (STEMCELL Technologies, 100-0276) media. The cells were passaged using non-enzymatic dissociation reagent ‘ReLeSR’ (STEMCELL Technologies, 05872). The culture was expanded for 2 passages (P30+6+4+3) and evaluated for morphology, % confluency, and lactate concentration. For the third passage, the hESCs were seeded on enhanced attachment microcarriers (Corning, 3779) in mTeSR plus media for 4 days in dynamic 0.5 L PBS wheels vessels (PBS Biotech, FA-0.5-D-001) (P30+6+4+4). During culturing, the cells were PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT morphologically evaluated, and lactate was measured. At the end of 4 days, the hESCs were harvested using TrypLE Select (Gibco, 12563029) and the MCs were removed using 70µm strainer (Pluriselect, 43-57070-51). [0212] hESCs Differentiation into Monocytes and iDCs Differentiation [0213] On day 0, to start the differentiation process, the harvested hESCs were seeded for aggregate formation in X-VIVO15 medium (Lonza, BEBP02-061Q) supplemented with 50 ng/ml rh-GM-CSF (Cellgenix, 001012-1000), 20 ng/ml rh-SCF (R&D systems, 255B-GMP-01M), 50 ng/ml rh-VEGF165 (R&D systems, 293-GMP-01M), 50 ng/ml rh-BMP4 (R&D systems, 314E- GMP-050), and 10µM of Y27632 (Tocris, 1254), from day 0 to day 3 of the differentiation. The aggregates were formed in 0.5L PBS wheel dynamic vessels. At the end of aggregate formation (day 3), lactate was measured, and the medium was replaced with fresh X-VIVO15 supplemented with 50 ng/ml rh-GM-CSF, 20 ng/ml rh-SCF, 50 ng/ml rh-VEGF165, and 50 ng/ml rh-BMP4. Then, the aggregates were transferred into static culture according to the lactate concentration and seeded in ULA CellSTACKs (Corning, 3303) (Lactate concentration was measured in the PBS wheel and the relevant volume according to Lactate was transferred per CellSTACK). The medium was replaced three times a week with X-VIVO15 supplemented with the following according to the relevant day of differentiation: Days 3-5 - 50 ng/ml rh-GM-CSF, 20 ng/ml rh-SCF, 50 ng/ml rh-VEGF165, and 50 ng/ml rh-BMP4 Days 6-10 - 50 ng/ml rh-GM-CSF, 20 ng/ml rh-SCF, and 50 ng/ml rh-VEGF165 Days 11-15 - 50 ng/ml rh-GM-CSF, and 20 ng/ml rh-SCF Days 15-27 - 50 ng/ml rh-GM-CSF [0214] Lactate concentration and culture morphology were evaluated during every media replacement. On day 0 and on day 3, the lactate concentration in culture served as IPC. On day 0 lactate concentration served a critical parameter for differentiation initiation, the cells were harvested only when the lactate was >13.00 mM. On day 3, the volume of aggregates seeded into each CellSTACK was completed according to the measured lactate concentration. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0215] During the differentiation process, up to day 28, the aggregates shed single cells, and the culture was composed of aggregates in suspension and single cells as a semi-adherent culture. [0216] On day 28 of differentiation, the aggregates were removed from the culture using customized 60 µm closed system filtration system (Meissner) and the single cells were cultured in CellGenix® GMP DC Medium (CG-DC, CellGenix, 20901-0500) supplemented with 50 ng/ml rh-GM-CSF and 50 ng/ml rh-IL-4 (R&D systems, 204-GMP-050) for 5 days in the CellSTACK. The medium was replaced on day 31 with fresh CG-DC media supplemented with 50 ng/ml rh- GM-CSF and 50 ng/ml rh-IL-4. In addition, lactate concentration measurement and morphology evaluation were performed. [0217] iDCs Maturation into LCT-mDCs [0218] On day 33 of differentiation, lactate and morphology of the cells were evaluated and 100% of the medium was replaced with maturation medium - CG-DC medium supplemented with PGE-2 (Sigma Aldrich, P6532). The medium was replaced every 24 hours for total of 48 hours. [0219] LCT-mDCs Harvest [0220] On day 35 of differentiation, lactate and morphology of the cells were evaluated and the mDCs were harvested using 1mM EDTA (BI, 01-862-1B) and the EDTA dissociation activity quenched using CG-DC. Harvested mDCs can be further processed to LCT-VAC2 by electroporation, cryopreservation, and X-Ray irradiation to yield the final LCT-VAC2. Alternatively, the harvested mDCs can be cryopreserved and processed to the final product as a separate stage. [0221] LCT-mDCs - Cryopreserved non-EP mDCs [0222] mDCs were cryopreserved at 25 x 106 cells per 1 ml per vial in CryoStor 5 cryopreservation solution (CS5, Biolife solutions, 205102) - LCT-mDCs intermediate product. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0223] These LCT-mDCs were thawed and electroporated with desired mRNA into the final product. The characterization of LCT-mDCs is detailed below. [0224] LCT-VAC2 - Cryopreserved mDCs EP’d with hTERT-LAMP1 mRNA and Irradiated [0225] Electroporation of LCT-mDCs with hTERT-LAMP1 mRNA [0226] For LCT-VAC2 mDCs, following harvest on day 35, cells in the quenching solution (1:3, 1mM EDTA quenched by CG-DC medium), as detailed above, were counted, diluted (1:10, v:v) with PBS minus (Sartorius, 02-023-1A) and centrifuged. After centrifugation the cells were resuspended and pooled at a cell concentration of 250 x 106 per mL Belzer UW Solution (Bridge to Life, BUWC). The cell suspension was adjusted to 100 x 106 cells per mL by adding Belzer UW solution, after which the cell suspension was divided into 0.3-1 x 109 aliquots each assigned for consecutive EP cycles, and kept at 2-8 °C. [0227] Per each aliquot of 0.3-1 x 109 cells, GMP grade hTERT-LAMP1 mRNA was added to the cell suspension in Belzer UW, to reach a final cell/mRNA mixture of 50 x 106 cells with 300 µg mRNA per mL. Cell/mRNA mixture was then transferred into the 4D Nucleofector inlet reservoir (Lonza, V4LR-1001W) via a Luer port. The inlet reservoir was placed on a stirrer at 300 RPM, keeping the mixture homogenous during EP. The EP cycles were performed using the 4D Nucleofector LV unit (Lonza) under vendor’s optimal electroporation program for introducing mRNA into human mature dendritic cells (CB150). Cell/mRNA mixture was pumped from the reservoir, through the LV cartridge (Lonza, V4LN-7020), electroporated, and finally transferred into the CG-DC medium - filled outlet bag, at fixed increments of 1 mL. Post EP, the cells were allowed to sit for 10 minutes at RT to recover from the EP process, at a cell concentration of 1 x 106 cells per mL. [0228] Cryopreservation of the EP’d LCT-mDCs in a Thaw-and-Inject (TAI) Formulation [0229] Post-EP cells were allowed to recover and are then filtered using a customized 60 µm closed-system filtration kit (Meissner), and then were counted using the NC-200 cell counter (DS counts). The cells were centrifuged and re-suspended in CryoStor 5 cryopreservation solution (CS5, Biolife solutions, 205102,). The cells were counted directly in CS5 while CS5 wasa PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT incrementally added followed by additional cell count until the required final cell concentration was achieved, prior to cryopreservation. After confirmation of target cell concentration, the cells were vialed at 6.25 x 106 cells per 500 µl per vial (Nunc Cryovial, Thermo-Fisher Scientific, 374086). The vials were then cryopreserved using the CryoMed controlled rate freezer. [0230] Irradiation of the Cryopreserved EP’d LCT-mDCs [0231] In the final step of LCT-VAC2 production, the cryopreserved electroporated mDCs were X-ray irradiated, to eliminate cell proliferation capabilities. Irradiation was carried out using Precision Xray’s XRAD 320 irradiation system. [0232] Determination of the irradiation dose parameters was performed with pure cryopreserved H1 hESCs, serving as a ‘worst case’ scenario to determine the irradiation dose parameters that are required to achieve mitotic arrest of these highly proliferating cells. Briefly, harvested single-cell H1 hESCs were cryopreserved using CS10 at 6.25 x 106 cells per 500 µl in NUNC cryovials (Thermo-Fisher Scientific, 374086), simulating the LCT-VAC2 final product vial type and cell concentration. Post irradiation evaluation of the proliferation capacity was done via a performance test, in which irradiated and non-irradiated H1 hESCs vials are thawed and cultured, for a 4-day culturing session, during which cell proliferation is evaluated by lactate measurements and morphological assessment from day 1 to 4. Lactate served as a direct indicator to the number of H1 hESCs in the culture, and the daily lactate accumulation as an indication of H1 hESCs proliferation capacity. Both lactate concentration and morphological appearance of irradiated hESCs are markedly distinguished from those of non-irradiated ESCs culture, as can be seen in Table 7, on day 4 post thawing non irradiated H1 cells reach above than 10mM lactate compared to 0 mM lactate of the irradiated H1 cells. The irradiation parameters that were selected and shown to be effective in arresting proliferation were: Dose: 1200 rad (12Gy), 130 kV, 12.5 mA, SSD (‘Source-to-Sample Distance’): 45/5 cm. Irradiation cycle time is in the range of approx.730-750 seconds. This dose was confirmed as having no effect on other attributes of LCT-VAC2 (viability, recovery, identity, and functional activity). [0233] Irradiation dose verification was done prior to each irradiation cycle by conducting a dry simulation (without vials) using a calibrated dose meter to verify the system’s operation. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Additionally, irradiation performance verification was done prior to each irradiation cycle with H1 cells, comparing irradiated and non-irradiated cells for proliferation potential as detailed above for the development of the irradiation parameters. [0234] An example of morphology test results for irradiated and non-irradiated H1 cells, are shown in FIG.5 and lactate measurements in Table 7. Table 7. H1 Post Irradiation Performance Test - Lactate Measurements IRR C cle Lactate (mM) 4 96 1
Figure imgf000077_0001
[0235] Based on these data, the process of irradiation using 1200 rad (12Gy), 130 kV, 12.5 mA, for approximately 730-750 seconds was sufficient to inhibit any proliferative abilities while retaining viability and functional activity. [0236] Process Performance Evaluation: In- Process Controls (IPCs) [0237] In-process control (IPC) markers monitoring during differentiation, LCT-mDCs characterization, and the LCT-VAC2 product release testing of key attributes were performed by flow cytometry. Specific IPC plan was created to monitor cell characteristics along the differentiation process. [0238] hESCs-mDCs differentiation was based on the following hematopoietic cell types: hematopoietic progenitors, common myeloid precursors (CMP), monocytes, immature DCs (iDCs) and finally mDCs. A detailed IPC plan according to the different differentiation steps is depicted in FIG.6. A list of markers tested at IPC sample points are listed in Table 8 below. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 8. List of FCM Based IPC Along LCT-mDCs Generation/ LCT-VAC2 Differentiation Process IPC Differentiation Tested tar et Rationale
Figure imgf000078_0001
i. IPC2-day 6: CD34 is a transmembrane phosphor-glycoprotein expressed on hematopoietic stem cells and progenitor cells which is tested on day 6 (Patente, Pinho et al.2018). Generally, sufficient fraction of CD34+ cells among the differentiating cells is needed to make the differentiation process robust. CD90 is a known progenitor marker expressed by pluripotent cells including hESCs. As the differentiation process progresses CD90 expression was expected to be downregulated. ii. IPC3-day 20: CD45 (leukocyte common antigen) is a unique and ubiquitous membrane glycoprotein that is expressed on all hematopoietic cells except for mature erythrocytes (Nakano, Harada et al.1990). CD45 expression was known to be elevated around day 20 of differentiation process while cells were still grown in aggregates, indicating cellular differentiation towards leukocyte lineage. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT iii. IPC4-Day 27: Enables evaluation of cells before filtration of aggregates on day 28 of the differentiation process. CD45 expression is expected to increase compared to expression on day 20 as most of the single cells are leucocytes. CD14 expression was tested to ensure cells undergo monocytic step as part of their differentiation (Landmann, Muller et al.2000). CD86 a DC marker, was tested as well as part of cellular differentiation towards DCs (Mir 2015). iv. 1PC5-Day 32: Time point for evaluation of cells post aggregate filtration and prior to maturation toward mDCs to ensure that CD86+ expression continues to rise compared to day 27 and that high CD45+ expression is maintained. CD83 is expressed by mDCs (Zhou, Schwarting et al.1992) and was tested as part of kinetics profile, and CD83 expression increased until the end of the process on day 35, indicating that cells acquired the desired mDCs fate. [0239] LCTVAC2 Final Drug Product Characterization [0240] LCT-VAC2 was subjected to a panel of characterization assays developed to evaluate cells’ identity and functionality. These final product characterizations included cells’ viability, identity, and functional activity, and are summarized in Table 9 below. LCT-VAC2 cryopreserved vials were thawed and evaluated according to established assay protocols. Table 9. LCT-VAC2 Final Product Characterization Quality attribute Method Viability (% Viable cells) NC-200
Figure imgf000079_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT
Figure imgf000080_0001
[0241] LCT-mDCs Characterization [0242] LCT-mDCs was subjected to a panel of characterization assays developed to evaluate cells’ identity and functionality. These characterizations are summarized in Table 10 below. LCT- mDCs cryopreserved vials were thawed and evaluated according to established assay protocols. Table 10. LCT-mDCs Intermediate Product Characterization Quality attribute Method % CD45+ Leukocyte marker
Figure imgf000080_0002
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT
Figure imgf000081_0001
[0243] Characterization Assays (mDCs and LCT-VAC2) [0244] Viability and Total Viable Cells (Strength) [0245] Viability and total viable cells were determined using “NC-200 Nuclecounter”, an automated quantitative method using special fluorescent dyes for counting dead/live cells. Evaluation of Total viable cells/ml was done directly on thawed cells still in CS5 cryopreservation medium. [0246] Purity/Identity [0247] Purity/identity measurement was performed by flow cytometry, a quantitative method capable of detecting and identifying cellular markers on a single-cell basis, which enables the determination of cellular purity/identity. This assay was based on the assessment of cells expressing a selected panel of mDCs characteristic markers as listed in Table 9 and Table 10 and specified below. i. CD45 antigen (leukocyte common antigen) is a unique and ubiquitous membrane glycoprotein that is expressed on almost all hematopoietic cells except for mature PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT erythrocytes (Nakano, Harada et al.1990). mDCs were expected to express it at high levels at the end of differentiation process. ii. CD83 is an identity marker for mDCs at the end of mDCs differentiation process. It is an immunoglobulin protein expressed on the surface of mDCs (Zhou, Schwarting et al.1992). iii. CD86 is a co-receptor expressed on antigen presenting cells in general and DCs, in particular (Mir 2015). iv. HLA-A is a common isotype of MHC-I molecule. It was used to evaluate MHC class I molecule that is expressed on all nucleated cells and it’s involved in antigen presenting processes (Bray 2001). v. HLA-DR is a common isotype of MHC-II molecule. It is expressed on antigen presenting cells (Neefjes, Jongsma et al.2011). It was used to evaluate MHC class II expression on mDCs at the end of the differentiation process. It is known to be essential for antigen presenting processes. vi. CD40 is a co-stimulatory protein found on antigen-presenting cells and is required for their activation. Binding of CD154 (CD40-L) leads to a cascade of downstream effects result in cell activation (Grewal and Flavell 1998). mDCs cells express it at high levels upon maturation. vii. CCR7 is a chemokine receptor which serves to direct DCs to lymph nodes (Dieu, Vanbervliet et al.1998). Its expression is known to be significantly elevated upon cell maturation to mature DCs. [0248] Non-Targeted Cell Population [0249] Evaluation of non-targeted cell population was also performed by flow cytometry, a quantitative method capable of detecting and identifying cellular markers on a single-cell basis. This assay was based on the assessment of cells expressing a selected panel of mDCs characteristic markers as listed in Table 10 and specified below: PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT i. CD14 is a monocyte/macrophage differentiation antigen presented on the surface of cells of myeloid lineage (Landmann, Muller et al.2000). CD14 was tested as part of final product characterization to examine the final product for non-targeted cell populations with monocyte characteristics. ii. CD45(+) and CD83(-) staining of cells was used to determine percentage of non- DC cells at the final product. [0250] Residual hESCs [0251] LCT-VAC2/LCT-mDCs was manufactured by differentiating hESCs to mDCs cells. The level of residual hESCs in the final product was measured using two-dimensional flow cytometry analysis for two markers of undifferentiated hESCs: TRA-1-60 and SSEA-5. [0252] Functional Activity Assays [0253] LCT-VAC2 cells’ mode of action in the patient was based on the premise that these matured DC cells EP with hTERT antigen can migrate to adjacent lymph node and successfully activate T cells by presenting the hTERT antigen. A set of functional activity assays was designed for characterization of LCT-VAC2 final product. These tests were based on mimicking the biological activity of LCT-VAC2 cells in the patient’s immune system and lymph nodes. i. using in-vitro Transwell system 6.5 mm with 8.0 µm Pore Polycarbonate Membrane Insert, Corning, #3422) based assay. Briefly, DC media supplemented cells were added to the top chambers. The number of cells that were migrated to the lower chamber was determined by counting in a NC-200 cell counter 4 hours post incubation. Percentage of migrated cells was calculated based on dividing the number of migrated cells by the number of cells initially cultured. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT ii. Mixed Leukocyte Reaction assay (MLR): The ability of LCT-VAC2 cells to promote allogeneic T cell proliferation was assessed using Mixed Leukocyte Reaction assay (MLR). Proliferation of fluorescently pre-labeled peripheral blood mononuclear cells (PBMC) following co-culture with LCT-VAC2 was measured using flow cytometry. Percent of proliferating CD8+ T cells was used to evaluate T cell activation. iii. hTERT Antigen presentation assay (for LCT-VAC2 only): The ability of LCT- VAC2 cells to present and activate hTERT specific T cells was evaluated by co- incubation of LCT-VAC 2 cells with hTERT specific T cells generated using co culture of HLA-A-02:01 PBMC derived mDCs differentiated from isolated monocytes with autologous T cells in the presence of hTERT peptide (Miltenyi following co culture of cells with allogeneic mDCs in the presence of hTERT peptide (Miltenyi Biotec #130-097-277). mDCs that were electroporated with GFP or cells not electroporated was used as negative control. The degree of T cell LCT-VAC 2 final product (post EP and Irradiation). iv. To assess the functional attributes of mDCs prior to electroporation, CMV Antigen presentation was used to assess the potential of mDCs to activate T cells via general antigen presentation via peptide uptake. Pp65 specific (Cytomegalovirus (CMV) known antigen) T cells isolated from commercially available Leukopak (Stem Cell Technologies # 200131) of a CMV positive donor. These antigen - specific T cells were co-cultured with LCT-mDCs at a ratio of 1:10 (Effector:Target), with 1 µg of pp65 peptide (PepTivatorCMV pp65, Miltenyi Biotec, #130-093-438) in 24-well plate (Corning, #3524) for 20-24 hours.100 IU/ml IL-2 (Miltenyi Biotec, #130-097-742) was added to the media. T cells co- cultured with mDCs but without peptide, or T cells alone were used as negative PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT controls for A INFy secretion calculation. INFy (Interferon gamma) secretion into culture media was evaluated using a commercially available ELISA assay (R&D Systems,# DIF50C). To evaluate the extent of antigen specific T cell activation, A secretion by negative control group without peptide. v. Expression of hTERT-LAMP-1 mRNA in the final LCT-VAC 2 product was confirmed by Western blot (protein) and RT-PCR (mRNA presence) methods. vi. Western blot (WB): LCT-VAC 2 cells were thawed and incubated at 37 °C for 5 hours to allow antigen expression, washed and snapfrozen in liquid nitrogen as cell pellet. Cell pellet was lysed before separation on an SDS-PAGE (4-12% Bis-Tris gel, Bio-Rad# 1610772) under reducing conditions, and bloted onto a nitrocellulose membrane. Detection of hTERT-LAMP-1 band was done by specialized monoclonal antibody. Assay was performed on thawed LCT-VAC 2 final product. Non-electroporated cells are used as a negative control. vii. qPCR: qPCR was performed to confirm the presence of hTERT-LAMP-1 mRNA in the final product after electroporation. Briefly, mRNA was extracted from cells using a Qiagen RNeasy Mini kit (Qiagen # 74106) and transcribed into cDNA. cDNA amplification was performed with hTERT-LAMP-1 construct specific primers (Tagman). Assay was performed on thawed LCT-VAC2 final product. Non-electroporated cells are used as a negative control. [0254] Materials Used in Manufacturing of LCT-VAC2
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 11. Materials Used for Manufacturing of LCT-VAC2 Drug Product i h n 27 m m m m m n - n 33 n 14
Figure imgf000086_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT n - n 10 n 5 n n n n for n
Figure imgf000087_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT g n n
Figure imgf000088_0001
[0255] Stability of LCT-VAC2 Drug Product [0256] The LCT-VAC2 drug product is planned to undergo a 5-year stability program. Each produced batch will be tested at 6- and 12-months post-manufacturing and annually for an additional 4 years thereafter. [0257] Shipment, Formulation and Administration [0258] LCT-VAC2 is cryopreserved as a TAI product in CS5 and maintained in the vapor phase in LN2. It will be shipped to clinical sites under the same conditions and will be thawed immediately prior to administration at the clinical site. [0259] NONCLINICAL PHARMACOLOGY AND TOXICOLOGY [0260] LCT-VAC2 has an immunologically complex mechanism of action. Consequently, there are no relevant animal models in which to evaluate the safety of LCT-VAC2. Immunologically PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT deficient animals would tolerate LCT-VAC2 without recognizing the cells as xenogeneic material but would lack the necessary effector systems to elicit LCT-VAC2-mediated biological effects. Given the species selectivity of the immune system, it is unlikely that LCT-VAC2 would function correctly in immunologically competent animals and the animal would probably rapidly clear any LCT-VAC2 since it would be recognized as a foreign material. Additionally, “humanized” mouse models rely on adoptive transfer of human peripheral blood mononuclear cells (PBMC) or CD34+ hematopoietic progenitor cells. Notably, PBMC transfer leads to graft versus host disease (GVHD) in the humanized mice in 3-6 weeks, and thus would complicate any safety or efficacy signal observed from a series of LCT-VAC2 injections. Additionally, human T cells that arise from CD34+ transfer are not educated by a human thymus. Therefore, it is unlikely that LCT- VAC2 antigen presentation will stimulate a T cell response in this context. Any data generated with LCT-VAC2 in a non-clinical in vivo model would potentially be misleading. [0261] PHARMACOLOGY STUDIES [0262] The ability of DCs to regulate immunity is dependent on the extent to which they have matured. DC maturation involves a multistep process including redistribution of HLA molecules from intracellular endocytic compartments to the cell surface, increases in the surface expression of co-stimulatory molecules, secretion of chemokines, cytokines and proteases, and surface expression of adhesion molecules and chemokine receptors (Wu, Guarnieri et al.1995). Following antigen exposure and activation, DCs migrate into T cell areas of lymphoid organs where they present the antigen as well as release co-stimulatory cell surface and soluble factors that stimulate T cell proliferation. hESCs-derived DCs exhibit a phenotype comparable with immature and of stimulating T cell proliferation in response to viral antigens and the tumor antigen hTERT. These cells have phagocytic activity and release pro-inflammatory cytokines. Cell surface markers on mature hESCs-derived DCs include CD83 and CD86 HLA I and II as well as CCR7. All these features are common attributes of mature DCs and suggest that hESCs-derived DCs are functionally and phenotypically comparable with endogenous DCs. hESCs-derived DCs transfected with hTERT/LAMP-1 mRNA were reactive towards hTERT-loaded PBMCs obtained PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT from HLA-A2+ individuals as assessed by ELISPOT analysis in vitro. LCT-VAC2 cells transfected with hTERT/LAMP-1 mRNA were capable of priming naïve T cells isolated from peripheral blood matched on 1, 2, or 4 HLA alleles. Importantly, from the perspective of potential antitumor activity, LCT-VAC2 has been shown to be capable of stimulating both memory and naïve semi-allogeneic T cells in an hTERT-dependent manner. [0263] Safety pharmacodynamic as well as pharmacodynamic interaction studies have not been undertaken with LCT-VAC2. This is partly due to the absence of a relevant non-clinical model but is also principally due to existing clinical data already demonstrating that hTERT/LAMP-1 mRNA transfected into autologous DCs (VAC1) does not demonstrate any evidence of secondary or adverse safety pharmacology (Khoury, Collins et al.2010, Khoury, Collins et al.2017). As LCT- VAC2 has been developed to be comparable with endogenous DCs, these published data suggest that the potential for any unintended pharmacology to manifest itself is low. In summary, hTERT/LAMP-1 mRNA transfected DCs derived from hESCs have functional and phenotypic features that are characteristic of PBM-derived DCs (Tseng, Nishimoto et al.2009). These cells are highly specific for HLA A2+ cells expressing hTERT and there is no evidence to suggest that secondary and/or safety pharmacology issues associated with LCT-VAC2 treatment are likely. [0264] Pharmacokinetics [0265] Conventional pharmacokinetics is not relevant for LCT-VAC2 and, therefore, no studies have been undertaken. However, there is a body of clinical data that describes the fate of autologous DCs following systemic administration. In summary, administered DCs show a depot effect for up to 48 hours post-injection. Over two thirds of the administered cells remain at the site of administration with the remainder undergoing translocation, principally to the regional draining lymph nodes. Activated DCs have a short biological half-life, approximately 2-3 days, before being phagocytosed (Nayak, Hokey et al.2006, Savina and Amigorena 2007). Therefore, given the LCT-VAC2 similar characteristics with endogenous and autologous DCs, it is considered that the fate of LCT-VAC2 in animals can be assumed. The potential for LCT-VAC2 to undergo classical pharmacokinetic interactions with other drugs is unlikely. Accordingly, no studies of the pharmacokinetic interactions of LCT-VAC2 have been performed. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0266] Toxicology Studies [0267] In the absence of suitable animal models, no animal toxicology studies have been undertaken. However, LCT-VAC2 has been shown to be phenotypically equivalent to autologous DCs which have previously demonstrated tolerability in numerous clinical trials involving many hundreds of subjects (Brunsvig, Kyte et al.2011, Dorrie, Schaft et al.2020). As described above, LCT-VAC2 differs from autologous DC therapy in one notable respect in that subjects treated with LCT-VAC2 are only partially haplotype matched to the potential subject. There have been no indications of unexpected SAEs or AEs of increased severity in the completed VAC2 CRUK study and no GVHD or cytokine release syndrome following administration of VAC2 or other allogeneic DC vaccines trials in both solid and hematological malignancies at similar doses as those proposed for the US study in NSCLC. [0268] LCT-VAC2 has been manufactured to produce terminally differentiated DCs, which have limited proliferative potential, minimizing a theoretical risk of ectopic growth or tumorigenicity. Furthermore, the product is irradiated to eliminate any residual proliferative potential of the cells (including the presence of any remaining undifferentiate stem cells). [0269] Pharmacodynamic studies have demonstrated that LCT-VAC2 presents hTERT epitopes on HLA I expressed upon their cell surfaces (and, by inference, hTERT expressed on HLA II as well since LAMP-1 epitopes are expressed on both HLA I and II). Accordingly, LCT-VAC2 would be expected to elicit an immune response towards cells that express hTERT and/or LAMP- 1. In adults, hTERT expression is restricted to the testes and gastrointestinal tract and LAMP-1 predominantly to exhausted T cells. Clinical trials have been conducted in which either hTERT peptide or hTERT/LAMP-1 mRNA transfected DCs have been given to subjects (Su, Dannull et al.2005, Khoury, Collins et al.2010, Khoury, Collins et al.2017). Overall, these studies suggest that vaccination with hTERT-(and LAMP-1-) expressing DCs is well tolerated. These studies failed to show any toxicity in tissues known to express hTERT or any evidence of any off-target effects due to conserved changes in antigens. [0270] In the dose escalation clinical trial, four target dose groups are planned (1.25, 2.5, 5, and 10 million cells per injection). Previous trials with VAC1, VAC2, and other DC vaccines PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT demonstrated that a dose of up to 2 x 107 cells per injection (the maximum dose in the CRUKD/17/003 study) is both tolerable and induces tumor antigen-associated T cell responses in cancer subjects with a range of solid and hematological malignancies including NSCLC. Each subject will initially receive a maximum of six immunizations (in Weeks 1, 2, 3, 4, 5 and 6) based on previous clinical experience with both the autologous VAC1 and the allogeneic VAC2, where six immunizations were found to induce the highest levels of anti-hTERT immune responses. [0271] In summary, the product has been shown to be a clinically well-tolerated experimental treatment for cancer. Acceptable safety events reported to date include mild, self-limiting, localized injections site reactions (ISR) as well as fatigue, mild-fever, chills etc. The partial HLA mismatch of LCT-VAC2 to subjects may contribute to any localized rejection reaction over and above what might be seen with an autologous product. However, both the number of cells administered, along with the transient nature of the cells following administration, will ensure that any rejection reaction manifested should be short-lived and may even be beneficial given the adjuvant immune-stimulatory environment it would create. The irradiation of LCT-VAC2 cells prevents the possibility of further cell division in vivo. Consequently, the potential for ectopic cell growth or malignant transformation is not considered to be an issue with LCT-VAC2. [0272] CLINICAL DEVELOPMENT [0273] Overview of Clinical Development Plan [0274] Phase 1 Study [0275] A Phase 1 trial (CRUKD/17/003) of VAC2 (AST VAC2) was conducted at two centers in the UK by Cancer Research UK administered the vaccines weekly via intradermal injection in subjects with advanced NSCLC. The study has completed enrollment in February 2022 and all subjects have either withdrawn or died due to the progressive and advanced stage of their disease.In this study, two intradermal injections with a total target dose of 1 x 107 cells of VAC2 were administered at six time points over a period of six consecutive weeks. [0276] Eight subjects received VAC2 and there were no unexpected SAEs, AEs or events of increased severity that would have mandated an interruption in scheduled dosing Non-Serious PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT AEs probably related to VAC2 observed in the trial were limited to injection site reactions (ISRs) and a single infusion related hypersensitivity reaction. Initial safety data from the eight subjects demonstrated that the vaccine was well tolerated, with the full (6x) regimen delivered to all trial subjects without treatment delays. Though limited, initial pentamer and ELISA assay data were highly compelling with respect to the magnitude and durability of antigen-specific T cell professionalization. Initial assessments of the immunogenicity data to date suggest the desired immune effects were generated in VAC2 treated subjects (Table 12). Of interest, a major radiological response was reported in one patient post-treatment. Table 12. Interim Results of Pentamer Assay in First Four Subjects Treated % of hTERT Reactive TCells in Patient Blood by Pentamer Assay hs on
Figure imgf000093_0001
[0277] Proposed Phase 2a Study [0278] The proposed Phase 2a study is designed to evaluate the safety and tolerability of escalating target doses of LCT-VAC2 utilizing 4 dose groups of n=3 subjects, (1.25, 2.5, 5 or 10 million cells per administration) given as six weekly intradermal (ID) injections in subjects with refractory Stage IIIB or IV NSCLC subjects who previously failed at least one approved immunotherapy regimen (e.g. PD-1 or PD-L1 checkpoint inhibitors or CTLA-4 receptor antibodies) and one platinum-based chemotherapy cycle, or a combination of both. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT REFERENCES [0279] 1. Bray, R. A. (2001). "Flow cytometry in human leukocyte antigen testing." Semin Hematol 38(2): 194-200. [0280] 2. Brunsvig, P. F., et al. (2011). "Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial." Clin Cancer Res 17(21): 6847-6857. [0281] 3. Chae, Y. K., et al. (2018). "Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC)." J Immunother Cancer 6(1): 39. [0282] 4. Chen, Z., et al. (2014). "Non-small-cell lung cancers: a heterogeneous set of diseases." Nat Rev Cancer 14(8): 535-546. [0283] 5. De Giglio, A., et al. (2021). "The Landscape of Immunotherapy in Advanced NSCLC: Driving Beyond PD-1/PD-L1 Inhibitors (CTLA-4, LAG3, IDO, OX40, TIGIT, Vaccines)." Curr Oncol Rep 23(11): 126. [0284] 6. Dieu, M. C., et al. (1998). "Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites." J Exp Med 188(2): 373-386. [0285] 7. Dorrie, J., et al. (2020). "Therapeutic Cancer Vaccination with Ex Vivo RNA- Transfected Dendritic Cells-An Update." Pharmaceutics 12(2). [0286] 8. Eskelinen, E. L. (2006). "Roles of LAMP-1 and LAMP-2 in lysosome biogenesis and autophagy." Mol Aspects Med 27(5-6): 495-502. Grewal, I. S. and R. A. Flavell (1998). "CD40 and CD154 in cell-mediated immunity." Annu Rev Immunol 16: 111-135. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0287] 10. Khoury, H. J., et al. (2010). "Prolonged Administration of the Telomerase Vaccine GRNVAC 1 Is Well Tolerated and Appears to Be Associated with Favorable Outcomes In High- Risk Acute Myeloid Leukemia (AML)." Blood 116(21): 2190. [0288] 11. Khoury, H. J., et al. (2017). "Immune responses and long-term disease recurrence status after telomerase-based dendritic cell immunotherapy in patients with acute myeloid leukemia." Cancer 123(16): 3061-3072. [0289] 12. Landmann, R., et al. (2000). "CD14, new aspects of ligand and signal diversity." Microbes Infect 2(3): 295-304. [0290] 13. Lin, K. Y., et al. (1996). "Treatment of established tumors with a novel vaccine that enhances major histocompatibility class II presentation of tumor antigen." Cancer Res 56(1): 21- 26. [0291] 14. Lynch, T. J., et al. (2012). "Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study." J Clin Oncol 30(17): 2046-2054. [0292] 15. Mielgo-Rubio, X., et al. (2021). "Immunotherapy in non-small cell lung cancer: Update and new insights." J Clin Transl Res 7(1): 1-21. [0293] 16. Mir, A. M. (2015). "Chapter 1 - Introduction to Costimulation and Costimulatory Molecules." Developing Costimulatory Molecules for Immunotherapy of Diseases: 1-43. [0294] 17. Morse, M. A., et al. (1999). "Migration of human dendritic cells after injection in patients with metastatic malignancies." Cancer Res 59(1): 56-58. [0295] 18. Nakano, A., et al. (1990). "Expression of leukocyte common antigen (CD45) on various human leukemia/lymphoma cell lines." Acta Pathol Jpn 40(2): 107-115. [0296] 19. Nayak, J. V., et al. (2006). "Phagocytosis induces lysosome remodeling and regulated presentation of particulate antigens by activated dendritic cells." J Immunol 177(12): 8493-8503. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0297] 20. Neefjes, J., et al. (2011). "Towards a systems understanding of MHC class I and MHC class II antigen presentation." Nat Rev Immunol 11(12): 823-836. [0298] 21. Ouellette, M. M., et al. (2011). "Targeting telomerase-expressing cancer cells." J Cell Mol Med 15(7): 1433-1442. [0299] 22. Patente, T. A., et al. (2018). "Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy." Front Immunol 9: 3176. [0300] 23. Quoix, E., et al. (2016). "TG4010 immunotherapy and first-line chemotherapy for advanced non-small-cell lung cancer (TIME): results from the phase 2b part of a randomised, double-blind, placebo-controlled, phase 2b/3 trial." Lancet Oncol 17(2): 212-223. [0301] 24. Quoix, E., et al. (2011). "Therapeutic vaccination with TG4010 and first-line chemotherapy in advanced non-small-cell lung cancer: a controlled phase 2B trial." Lancet Oncol 12(12): 1125-1133. [0302] 25. Reck, M., et al. (2013). "Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial." Ann Oncol 24(1): 75-83. [0303] 26. Savina, A. and S. Amigorena (2007). "Phagocytosis and antigen presentation in dendritic cells." Immunol Rev 219: 143-156. [0304] 27. Spira, A. and D. S. Ettinger (2004). "Multidisciplinary management of lung cancer." N Engl J Med 350(4): 379-392. [0305] 28. Su, Z., et al. (2005). "Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ T cell responses in patients with metastatic prostate cancer." J Immunol 174(6): 3798-3807. [0306] 29. Tseng, S. Y., et al. (2009). "Generation of immunogenic dendritic cells from human embryonic stem cells without serum and feeder cells." Regen Med 4(4): 513-526. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0307] 30. Wu, T. C., et al. (1995). "Engineering an intracellular pathway for major histocompatibility complex class II presentation of antigens." Proc Natl Acad Sci USA 92(25): 11671-11675. [0308] 31. Zhou, L. J., et al. (1992). "A novel cell-surface molecule expressed by human interdigitating reticulum cells, Langerhans cells, and activated lymphocytes is a new member of the Ig superfamily." J Immunol 149(2): 735-742. Table 13. Protocol Synopsis Name of S onsor e l c al g
Figure imgf000097_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e e s n d d e f h I g e e n y n d ll ll is n d s d
Figure imgf000098_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e - ) al r c e e o n y e t st
Figure imgf000099_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e r e m T L
Figure imgf000100_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e y e o e n s r g is
Figure imgf000101_0001
9 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e e r r e . n e m s r of t
Figure imgf000102_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e I/ , )
Figure imgf000103_0001
1 PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e e st e y e d d s h ll t nt ed
Figure imgf000104_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT e y s e e 5 s, be
Figure imgf000105_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 14. e, s f s
Figure imgf000106_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 15. n ) p d
Figure imgf000107_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 16. t d d s g al l s d n-
Figure imgf000108_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT a s
Figure imgf000109_0001
a release.
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 17. LCT-VAC2/mDCs – ER-RD#02 FCM based IPC results *Ba
Figure imgf000110_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT Table 18. LCT-VAC2/mDCs – ER-RD#02 Batch Release Results Product Name: VAC2 ER-RD#02 (hTERT) Cell Concentration (cells/ml): 6.25×106 cells/0.5ml Freezing Date: 19 May 2022
Figure imgf000111_0001
PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT
Figure imgf000112_0001
P EMBODIMENTS [0309] P Embodiment 1. A method of differentiating human embryonic stem cells (hESCs) into mature dendritic cells (mDCs), the method comprising: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce cultured cells;(c) removing the cultured cells from the suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs); and (e) contacting the population of iDCs with a maturation cocktail for a third period of time to differentiate the population of iDCs into a population of mature dendritic cells (mDCs). [0310] P Embodiment 2. The method of P Embodiment 1, wherein the culturing in step (b) is dynamic. [0311] P Embodiment 3. The method of P Embodiment 1, wherein the culturing in step (b) is static. [0312] P Embodiment 4. The method of any one of P Embodiments 1-3, wherein the differentiation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous cytokine comprises granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0313] P Embodiment 5. The method of any one of P Embodiments 1-4, wherein an iDC of the population of iDCs in step (d) expresses CD45 and CD86. [0314] P Embodiment 6. The method of any one of P Embodiments 1-5, wherein an iDC of the population of iDCs in step (d) does not express CD38. [0315] P Embodiment 7. The method of any one of P Embodiments 1-5, wherein step (d) comprises dynamic culturing of the cultured cells followed by static culturing of the cultured cells. [0316] P Embodiment 8. The method of P Embodiment 7, wherein the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days. [0317] P Embodiment 9. The method of P Embodiment 7 or 8, wherein the static culturing is for about 3 days to about 27 days. [0318] P Embodiment 10. The method of any one of P Embodiments 1-9, wherein the maturation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous [0319] P Embodiment 11. The method of any one of P Embodiments 1 to 10, wherein an mDC of the population of mDCs in step (e) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express CD14, CD45, TRA160, and SSEA-5. [0320] P Embodiment 12. The method of any one of P Embodiments 1 to 11, wherein step (e) comprises static culturing of the population of iDCs. [0321] P Embodiment 13. The method of P Embodiment 12, wherein the static culturing is for about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days. [0322] P Embodiment 14. The method of any one of P Embodiments 1-13, wherein any one of or both of steps (d) and (e) are performed under serum free conditions. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0323] P Embodiment 15. The method of any one of P Embodiments 1-13, wherein any one of or both of steps (d) and (e) are performed in the absence of feeder cells. [0324] P Embodiment 16. The method of any one of P Embodiments 1-13, wherein any one of or both of steps (d) and (e) are performed in the absence of stromal cells. [0325] P Embodiment 17. The method of any one of P Embodiments 1-16, wherein steps (a), (b), and (c) are repeated after step (c), prior to step (d). [0326] P Embodiment 18. The method of P Embodiment 17, wherein steps (a), (b) and (c) are each repeated three times prior to step (d). [0327] P Embodiment 19. The method of any one of P Embodiments 1-18, wherein steps (a), (b) and (c) combined are about three days to about five days. [0328] P Embodiment 20. The method of any one of P Embodiments 1-19, wherein the cultured cells of the suspendable expansion complex remain undifferentiated and pluripotent in steps (a) through (c). [0329] P Embodiment 21. The method of P Embodiment 20, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. [0330] P Embodiment 22. The method of P Embodiment 20, wherein at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog. [0331] P Embodiment 23. The method of P Embodiment 20, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60, at least about 70% of the undifferentiated and pluripotent cells express Oct-4, and at least about 70% of the undifferentiated and pluripotent cells express Nanog. [0332] P Embodiment 24. The method of any one of P Embodiments 1-23, wherein step (b) further comprises changing the growth medium to remove accumulated lactate. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0333] P Embodiment 25. The method of any one of P Embodiments 1-24, wherein the growth medium comprises Y-27632. [0334] P Embodiment 26. The method of any one of P Embodiments 17-25, wherein the culturing in step (b) is dynamic. [0335] P Embodiment 27. The method of any one of P Embodiments 1-26, wherein the cultured cells in step (b) comprise hematopoietic cell intermediates. [0336] P Embodiment 28. The method of P Embodiment 27, wherein the hematopoietic cell intermediates express CD34, CD90. [0337] P Embodiment 29. The method of P Embodiment 27 or 28, wherein the hematopoietic cell intermediates do not express CD38. [0338] P Embodiment 30. The method of P Embodiment 27, wherein the hematopoietic cell intermediates express CD45. [0339] P Embodiment 31. The method of P Embodiment 30, wherein the hematopoietic cell intermediates do not express CD38. [0340] P Embodiment 32. The method of any one of P Embodiments 1-30, wherein the microcarrier comprises one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof. [0341] P Embodiment 33. The method of any one of P Embodiments 1-32, the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof. [0342] P Embodiment 34. The method of any one of P Embodiments 1-33, wherein the microcarrier is not coated. [0343] P Embodiment 35. The method of any one of P Embodiments 1-34, wherein the microcarrier is coupled with protamine or polylysine. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0344] P Embodiment 36. The method of any one of P Embodiments 1-35, wherein the microcarrier has a neutral charge. [0345] P Embodiment 37. The method of any one of P Embodiments 1-36, wherein the microcarrier is negatively charged. [0346] P Embodiment 38. The method of any one of P Embodiments 1-37, wherein the microcarrier is hydrophilic. [0347] P Embodiment 39. The method of any one of P Embodiments 1-38, wherein the ECM component comprises a matrigel, laminin, vitronectin, collagen, a derivative thereof, or any combination thereof. [0348] P Embodiment 40. The method of P Embodiment 39, wherein the laminin is human laminin. [0349] P Embodiment 41. The method of P Embodiment 40, wherein the human laminin is 511 E8 fragment. [0350] P Embodiment 42. The method of any one of P Embodiments 1-41, wherein the mDCs are characterized by: (i) expression of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c, but not expressing CD14, CD45, TRA160, and SSEA-5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii). [0351] P Embodiment 43. The method of any one of P Embodiments 1-42, further comprising (f) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. [0352] P Embodiment 44. The method of P Embodiment 38, any one of P Embodiments 1-37, further comprising (f) cryopreserving the population of mDCs. [0353] P Embodiment 44a. The method of P Embodiment 38, any one of P Embodiments 1-43, further comprising (f) cryopreserving the population of mDCs. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0354] P Embodiment 45. The method of P Embodiment 44, further comprising (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. [0355] P Embodiment 46. The method of P Embodiment 43 or 45, wherein the population of mDCs is contacted with two or more antigens. [0356] P Embodiment 47. The method of any one of P Embodiments 43-45, wherein the antigen is a tumor antigen. [0357] P Embodiment 48. The method of P Embodiment 46, wherein the tumor antigen comprises a tumor associated antigen (TAA). [0358] P Embodiment 49. The method of P Embodiment 47, wherein the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof. [0359] P Embodiment 50. The method of P Embodiment 46, wherein the tumor antigen comprises a tumor-specific antigen (TSA). [0360] P Embodiment 51. The method of P Embodiment 46, wherein the tumor antigen comprises a neoantigen. [0361] P Embodiment 52. The method of P Embodiment 43 or 45, wherein the nucleic acid comprises an RNA. [0362] P Embodiment 53. The method of P Embodiment 52, wherein the RNA comprises an mRNA encoding hTERT or fragment thereof. [0363] P Embodiment 54. The method of P Embodiment 54, the wherein the fragment comprises one or more immunogenic epitopes of hTERT. [0364] P Embodiment 55. The method of P Embodiment 44 or 45, wherein the population of mDCs comprising the one or more antigens is cryopreserved. [0365] P Embodiment 56. The method of P Embodiment 54, wherein the cryopreserved population of mDCs comprising the one or more antigens is irradiated. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0366] P Embodiment 57. A cell produced by any one of P Embodiments 1-41, wherein the cell expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express CD14, CD83, TRA160 and SSEA-5. [0367] P Embodiment 58. A cell produced by any one of P Embodiments 43-55, wherein the cell (i) expresses CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express CD14, CD83, TRA160, and SSEA-5; and (ii) comprises the one or more antigens. [0368] P Embodiment 59. A cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine comprising:(a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier. [0369] P Embodiment 60. A cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine comprising: (a) one or more dendritic cells: (i) expressing CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing CD14, CD83, TRA160, and SSEA-5; and (ii) comprising a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. [0370] P Embodiment 61. The cell vaccine of P Embodiment 60, wherein the nucleic acid comprises a DNA. [0371] P Embodiment 62. The cell vaccine of P Embodiment 61, wherein the DNA is under the control of a promoter. [0372] P Embodiment 63. The cell vaccine of P Embodiment 62, wherein the promoter is an inducible promoter. [0373] P Embodiment 64. The cell vaccine of P Embodiment 63, further comprising (c) an inducing agent. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0374] P Embodiment 65. The cell vaccine of P Embodiment 64, wherein the inducing agent is desferrioxamine. [0375] P Embodiment 66. The cell vaccine of P Embodiment 60, wherein the nucleic acid comprises an RNA. [0376] P Embodiment 67. The cell vaccine of any one of P Embodiments 60-66, wherein the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent. [0377] P Embodiment 68. The cell vaccine of any one of P Embodiments 60-67, wherein the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder. [0378] P Embodiment 69. The cell vaccine of P Embodiment 68, wherein the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer. [0379] P Embodiment 70. The cell vaccine of any one of P Embodiments 60-69, further comprising one or more additional therapeutic agents. [0380] P Embodiment 71. The cell vaccine of P Embodiment 70, wherein the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents. [0381] P Embodiment 72. The cell vaccine of any one of P Embodiments 60-71, wherein the one or more antigens is identified from the subject. [0382] P Embodiment 73. A method of manufacturing a cell vaccine for treating cancer, the method comprising: (a) obtaining one or more mature dendritic cells (mDCs) according to the method of any one of P Embodiments 1-42; and (b) contacting the one or more mDCs with one or PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer. [0383] P Embodiment 74. The method of P Embodiment 73, further comprising (c) providing one or more additional therapeutic agents. [0384] P Embodiment 75. The method of P Embodiment 73, wherein optionally the one or more mDCs are cryopreserved and thawed before step (b). [0385] P Embodiment 76. The method of any one of P Embodiments 73-75, wherein the one or more antigens is one or more tumor antigens. [0386] P Embodiment 77. The method of P Embodiment 76, wherein the one or more tumor antigens comprises a TAA. [0387] P Embodiment 78. The method of P Embodiment 77, wherein the TAA is hTERT. [0388] P Embodiment 79. The method of P Embodiment 76, wherein the one or more tumor antigens comprises a TSA. [0389] P Embodiment 80. The method of P Embodiment 76, wherein the one or more tumor antigens comprises a neoantigen. [0390] P Embodiment 81. The method of any one of P Embodiments 73-80, wherein the one or more nucleic acid molecules comprises one or more RNA molecules. [0391] P Embodiment 82. The method of P Embodiment 81, wherein the one or more RNA molecules comprises hTERT mRNA. [0392] P Embodiment 83. The method of any one of P Embodiments 73-82, wherein the manufacturing takes about 30 days to about 50 days. [0393] P Embodiment 84. The method of any one of P Embodiments 73-83, wherein the one or more antigens is an antigen expressed by the cancer. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0394] P Embodiment 85. A method for treating or delaying progression of a disease or disorder in a subject, comprising administering to the subject a cell vaccine of any one of P Embodiments 59 to 72. [0395] P Embodiment 86. A method for treating cancer, comprising administering to a subject having cancer a cell vaccine made by the method of any one of P Embodiments 73-84. EMBODIMENTS [0396] Embodiment 1. A method of differentiating human embryonic stem cells (hESCs) into mature dendritic cells (mDCs), the method comprising: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce a cultured suspendable expansion complex comprising cultured cells; (c) removing the cultured cells from the cultured suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs); and (e) contacting the population of iDCs with a maturation cocktail for a third period of time to differentiate the population of iDCs into a population of mature dendritic cells (mDCs). [0397] Embodiment 2. The method of embodiment 1, wherein the culturing in step (b) is dynamic. [0398] Embodiment 3. The method of embodiment 1, wherein the culturing in step (b) is static. [0399] Embodiment 4. The method of any one of embodiments 1-3, wherein the removing of step (c) comprises an enzyme-free dissociation reagent. [0400] Embodiment 5. The method of any one of embodiments 1-4, wherein the removing of step (c) does not comprise manual dissection or manual scraping. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0401] Embodiment 6. The method of any one of embodiments 1-5, wherein the first period of time is between about 1 day to about 9 days. [0402] Embodiment 7. The method of any one of embodiments 1-6, wherein the differentiation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous cytokine comprises granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3). [0403] Embodiment 8. The method of any one of embodiments 1-7, wherein the differentiation cocktail comprises GM-CSF. [0404] Embodiment 9. The method of any one of embodiments 1-8, wherein the differentiation cocktail comprises GM-CSF and SCF. [0405] Embodiment 10. The method of any one of embodiments 1-9, wherein the differentiation cocktail comprises GM-CSF, SCF, and VEGF. [0406] Embodiment 11. The method of any one of embodiments 1-10, wherein the differentiation cocktail comprises GM-CSF, SCF, VEGF, and BMP-4. [0407] Embodiment 12. The method of any one of embodiments 1-11, wherein an iDC of the population of iDCs in step (d) expresses CD45 and CD86. [0408] Embodiment 13. The method of any one of embodiments 1-12, wherein an iDC of the population of iDCs in step (d) does not express CD38. [0409] Embodiment 14. The method of any one of embodiments 1-13, wherein the second period of time is between about 1 day to about 32 days. [0410] Embodiment 15. The method of any one of embodiments 1-14, wherein step (d) comprises dynamic culturing of the cultured cells followed by static culturing of the cultured cells. [0411] Embodiment 16. The method of embodiment 15, wherein the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0412] Embodiment 17. The method of embodiment 15 or 16, wherein the static culturing is for about 3 days to about 27 days. [0413] Embodiment 18. The method of any one of embodiments 1-17, wherein the maturation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous [0414] Embodiment 19. The method of any one of embodiments 1-18, wherein the maturation cocktail comprises GM-CSF and IL-4. [0415] Embodiment 20. The method of any one of embodiments 1-19, wherein the [0416] Embodiment 21. The method of any one of embodiments 1 to 18, wherein an mDC of the population of mDCs in step (e) expresses at least one of CD40, CD45, CD83, CD86, HLA- DR, HLA-A, CCR7, CD141, and CD11c, but does not express one or more of CD14, TRA160, and SSEA-5. [0417] Embodiment 22. The method of embodiment 1-21, wherein an mDC of the population of mDCs in step (e) co-expresses CD45 and CD83. [0418] Embodiment 23. The method of any one of embodiments 1-22, wherein at least 70% of the population of mDCs in step (e) express CD45. [0419] Embodiment 24. The method of any one of embodiments 1-23, wherein at least 70% of the population of mDCs in step (e) express CD83. [0420] Embodiment 25. The method of any one of embodiments 1-24, wherein at least 70% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. [0421] Embodiment 26. The method of any one of embodiments 1-25, wherein at least 20% of the population of mDCs in step (e) express HLA-DR. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0422] Embodiment 27. The method of any one of embodiments 1-24, wherein less than 20% of the population of mDCs in step (e) express CD14. [0423] Embodiment 28. The method of any one of embodiments 1-27, wherein less than 20% of the population of mDCs in step (e) are CD45+ and CD83-. [0424] Embodiment 29. The method of any one of embodiments 1-22, wherein the third period of time is between about 1 day to about 10 days. [0425] Embodiment 30. The method of any one of embodiments 1 to 29, wherein step (e) comprises static culturing of the population of iDCs. [0426] Embodiment 31. The method of embodiment 30, wherein the static culturing is for about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days. [0427] Embodiment 32. The method of any one of embodiments 1-31, wherein any one of or both of steps (d) and (e) are performed under serum free conditions. [0428] Embodiment performed in the absence of feeder cells. [0429] Embodiment 34. The method of any one of embodiments 1-31, wherein any one of or both of steps (d) and (e) are performed in the absence of stromal cells. [0430] Embodiment 35. The method of any one of embodiments 1-34, wherein steps (a), (b), and (c) are repeated after step (c), prior to step (d). [0431] Embodiment 36. The method of embodiment 35, wherein steps (a), (b) and (c) are each repeated three times prior to step (d). [0432] Embodiment 37. The method of any one of embodiments 1-36, wherein steps (a), (b) and (c) combined are about three days to about five days. [0433] Embodiment 38. The method of any one of embodiments 1-37, wherein the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in steps (a) through (c). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0434] Embodiment 39. The method of embodiment 38, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. [0435] Embodiment 40. The method of embodiment 38, wherein at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog. [0436] Embodiment 41. The method of embodiment 38, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60, at least about 70% of the undifferentiated and pluripotent cells express Oct-4, and at least about 70% of the undifferentiated and pluripotent cells express Nanog. [0437] Embodiment 42. The method of any one of embodiments 1-41, wherein step (b) further comprises changing the growth medium to remove accumulated lactate. [0438] Embodiment 43. The method of any one of embodiments 1-42, wherein the growth medium comprises Y-27632. [0439] Embodiment 44. The method of any one of embodiments 35-43, wherein the culturing in step (b) is dynamic. [0440] Embodiment 45. The method of any one of embodiments 1-44, wherein the cultured cells in step (b) comprise hematopoietic cell intermediates. [0441] Embodiment 46. The method of embodiment 45, wherein the hematopoietic cell intermediates express CD34, CD90. [0442] Embodiment 47. The method of embodiment 45 or 46, wherein the hematopoietic cell intermediates do not express CD38. [0443] Embodiment 48. The method of embodiment 45, wherein the hematopoietic cell intermediates express CD45. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0444] Embodiment 49. The method of embodiment 48, wherein the hematopoietic cell intermediates do not express CD38. [0445] Embodiment 50. The method of any one of embodiments 1-49, wherein the microcarrier comprises one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof. [0446] Embodiment 51. The method of any one of embodiments 1-50, the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof. [0447] Embodiment 52. The method of any one of embodiments 1-51, wherein the microcarrier is not coated. [0448] Embodiment 53. The method of any one of embodiments 1-52, wherein the microcarrier is coupled with protamine or polylysine. [0449] Embodiment 54. The method of any one of embodiments 1-53, wherein the microcarrier has a neutral charge. [0450] Embodiment 55. The method of any one of embodiments 1-54, wherein the microcarrier is negatively charged. [0451] Embodiment 56. The method of any one of embodiments 1-55, wherein the microcarrier is hydrophilic. [0452] Embodiment 57. The method of any one of embodiments 1-56, wherein the ECM component comprises a matrigel, laminin, vitronectin, collagen, a derivative thereof, or any combination thereof. [0453] Embodiment 58. The method of embodiment 57, wherein the laminin is human laminin. [0454] Embodiment 59. The method of embodiment 58, wherein the human laminin is 511 E8 fragment. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0455] Embodiment 60. The method of any one of embodiments 1-59, wherein the mDCs are characterized by: (i) expression of at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40, CCR7, CD141, and CD11c, but not expressing one or more of CD14, TRA160, and SSEA- 5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii). [0456] Embodiment 61. The method of any one of embodiments 1-60, wherein the mDCs are unable to replicate. [0457] Embodiment 62. The method of any one of embodiments 1-61, wherein the mDCs are irradiated to prevent replication. [0458] Embodiment 63. The method of any one of embodiments 1-62, further comprising (f) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. [0459] Embodiment 64. The method of any one of embodiments 1-62, further comprising (f) cryopreserving the population of mDCs. [0460] Embodiment 65. The method of embodiment 64, further comprising (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. [0461] Embodiment 66. The method of embodiment 63 or 65, wherein the population of mDCs is contacted with two or more antigens. [0462] Embodiment 67. The method of any one of embodiments 63-65, wherein the antigen is a tumor antigen. [0463] Embodiment 68. The method of embodiment 67, wherein the tumor antigen comprises a tumor associated antigen (TAA). [0464] Embodiment 69. The method of embodiment 68, wherein the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0465] Embodiment 70. The method of embodiment 67, wherein the tumor antigen comprises a tumor-specific antigen (TSA). [0466] Embodiment 71. The method of embodiment 67, wherein the tumor antigen comprises a neoantigen. [0467] Embodiment 72. The method of embodiment 63 or 65, wherein the nucleic acid comprises an RNA. [0468] Embodiment 73. The method of embodiment 72, wherein the RNA comprises an mRNA encoding hTERT or fragment thereof. [0469] Embodiment 74. The method of embodiment 7 , the wherein the fragment comprises one or more immunogenic epitopes of hTERT. [0470] Embodiment 75. The method of any one of embodiments 72-74, wherein the RNA comprises an mRNA encoding lysosomal-associated membrane protein 1 (LAMP1) or a fragment thereof. [0471] Embodiment 76. The method of any one of embodiments 72-75, wherein the RNA comprises an mRNA encoding heat shock protein 96 (HSP96). [0472] Embodiment 77. The method of any one of embodiments 63-76, wherein the population of mDCs comprising the one or more antigens is cryopreserved. [0473] Embodiment 78. The method of embodiment 77, wherein the cryopreserved population of mDCs comprising the one or more antigens is irradiated. [0474] Embodiment 79. The method of embodiment 78, wherein the cryopreserved population of mDCs is irradiated to prevent replication. [0475] Embodiment 80. A cell produced by any one of embodiments 1-59, wherein the cell expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160 and SSEA-5. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0476] Embodiment 81. The cell of embodiment 80, wherein the cell co-expresses CD45 and CD83. [0477] Embodiment 82. A cell produced by any one of embodiments 63-77, wherein the cell (i) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160, and SSEA-5; and (ii) comprises the one or more antigens. [0478] Embodiment 83. The cell of embodiment 82, wherein the cell co-expresses CD45 and CD83. [0479] Embodiment 84. A cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine comprising: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier. [0480] Embodiment 85. The cell vaccine of embodiment 84, wherein the one or more dendritic cells co-express CD45 and CD83. [0481] Embodiment 86. A cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine comprising: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) comprising a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. [0482] Embodiment 87. The cell vaccine of embodiment 86, wherein the one or more dendritic cells co-express CD45 and CD83. [0483] Embodiment 88. The cell vaccine of embodiment 86, wherein the nucleic acid comprises a DNA. [0484] Embodiment 89. The cell vaccine of embodiment 88, wherein the DNA is under the control of a promoter. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0485] Embodiment 90. The cell vaccine of embodiment 89, wherein the promoter is an inducible promoter. [0486] Embodiment 91. The cell vaccine of embodiment 90, further comprising (c) an inducing agent. [0487] Embodiment 92. The cell vaccine of embodiment 91, wherein the inducing agent is desferrioxamine. [0488] Embodiment 93. The cell vaccine of embodiment 86, wherein the nucleic acid comprises an RNA. [0489] Embodiment 94. The cell vaccine of any one of embodiments 86-93, wherein the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent. [0490] Embodiment 95. The cell vaccine of any one of embodiments 86-94, wherein the cell vaccine comprises between about 0.5 x 107 dendritic cells to about 2.5 x 107 dendritic cells. [0491] Embodiment 96. The cell vaccine of any one of embodiments 86-95, wherein the cell vaccine comprises between about 1.0 x 107 dendritic cells to about 2.0 x 107 dendritic cells. [0492] Embodiment 97. The cell vaccine of any one of embodiments 86-96, wherein the cell vaccine comprises about 1.0 x 107 dendritic cells. [0493] Embodiment 98. The cell vaccine of any one of embodiments 86-97, wherein the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder. [0494] Embodiment 99. The cell vaccine of embodiment 98, wherein the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0495] Embodiment 100. The cell vaccine of any one of embodiments 86-99, further comprising one or more additional therapeutic agents. [0496] Embodiment 101. The cell vaccine of embodiment 100, wherein the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents. [0497] Embodiment 102. The cell vaccine of any one of embodiments 86-101, wherein the one or more antigens is identified from the subject. [0498] Embodiment 103. The cell vaccine of any one of embodiments 86-102, wherein the cell vaccine is formulated to be administered to the subject directly after thawing. [0499] Embodiment 104. The cell vaccine of embodiment 103, wherein directly administering includes that dead cells are not removed from the composition prior to administration to the subject. [0500] Embodiment 105. The cell vaccine of embodiment 103 or 104, wherein the cell vaccine is formulated to be administered without washing or reconstitution. [0501] Embodiment 106. The cell vaccine of any one of embodiments 103-105, wherein the cell vaccine is formulated for administration within about 4 hours of thawing. [0502] Embodiment 107. The cell vaccine of any one of embodiments 103-105, wherein the cell vaccine is formulated for administration more than 4 hours after thawing. [0503] Embodiment 108. The cell vaccine of any one of embodiments 103-106, wherein the cell vaccine is formulated for administration within about 24 hours after thawing. [0504] Embodiment 109. A method of manufacturing a cell vaccine for treating cancer, the method comprising: (a) obtaining one or more mature dendritic cells (mDCs) according to the method of any one of embodiments 1-60; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0505] Embodiment 110. The method of embodiment 109, further comprising (c) providing one or more additional therapeutic agents. [0506] Embodiment 111. The method of embodiment 109, wherein optionally the one or more mDCs are cryopreserved and thawed before step (b). [0507] Embodiment 112. The method of any one of embodiments 109-111, wherein the one or more antigens is one or more tumor antigens. [0508] Embodiment 113. The method of embodiment 112, wherein the one or more tumor antigens comprises a TAA. [0509] Embodiment 114. The method of embodiment 113, wherein the TAA is hTERT. [0510] Embodiment 115. The method of embodiment 112, wherein the one or more tumor antigens comprises a TSA. [0511] Embodiment 116. The method of embodiment 112, wherein the one or more tumor antigens comprises a neoantigen. [0512] Embodiment 117. The method of any one of embodiments 109-116, wherein the one or more nucleic acid molecules comprises one or more RNA molecules. [0513] Embodiment 118. The method of embodiment 117, wherein the one or more RNA molecules comprises hTERT mRNA. [0514] Embodiment 119. The method of any one of embodiments 109-118, wherein the manufacturing takes about 30 days to about 50 days. [0515] Embodiment 120. The method of any one of embodiments 109-119, wherein the one or more antigens is an antigen expressed by the cancer. [0516] Embodiment 121. The method of any one of embodiments 109-120, wherein the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT [0517] Embodiment 122. A method for treating or delaying progression of a disease or disorder in a subject, comprising administering to the subject a cell vaccine of any one of embodiments 84 to 102. [0518] Embodiment 123. The method of embodiment 122, wherein the dendritic cells are allogeneic to the subject. [0519] Embodiment 124. A method for treating cancer, comprising administering to a subject having cancer a cell vaccine made by the method of any one of embodiments 109-120. [0520] Embodiment 125. The method of embodiment 124, wherein the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. [0521] Embodiment 126. The method of embodiment 124 or 125, wherein the mDCs are allogeneic to the subject.

Claims

PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT WHAT IS CLAIMED IS 1. A method of differentiating human embryonic stem cells (hESCs) into mature dendritic cells (mDCs), the method comprising: (a) expanding the hESCs in pluripotent state by combining: (i) the hESCs, (ii) an extracellular matrix (ECM) component, and (iii) a microcarrier in a growth medium to form a suspendable expansion complex; (b) culturing the suspendable expansion complex for a first period of time in the growth medium to produce a cultured suspendable expansion complex comprising cultured cells; (c) removing the cultured cells from the cultured suspendable expansion complex; and (d) contacting the cultured cells with a differentiation cocktail for a second period of time to differentiate the cultured cells into a population of immature dendritic cells (iDCs); and (e) contacting the population of iDCs with a maturation cocktail for a third period of time to differentiate the population of iDCs into a population of mature dendritic cells (mDCs). 2. The method of claim 1, wherein the culturing in step (b) is dynamic. 3. The method of claim 1, wherein the culturing in step (b) is static. 4. The method of any one of claims 1-3, wherein the removing of step (c) comprises an enzyme-free dissociation reagent. 5. The method of any one of claims 1-4, wherein the removing of step (c) does not comprise manual dissection or manual scraping. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 6. The method of any one of claims 1-5, wherein the first period of time is between about 1 day to about 9 days. 7. The method of any one of claims 1-6, wherein the differentiation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous cytokine comprises granulocyte-macrophage colony stimulating factor (GM-CSF), bond morphogenic protein 4 (BMP-4), vascular endothelial growth factor (VEGF), stem cell factor (SCF), fetal liver kinase ligand (FLT3L), thrombopoietin (TPO), or interleukin 3 (IL-3). 8. The method of any one of claims 1-7, wherein the differentiation cocktail comprises GM-CSF. 9. The method of any one of claims 1-8, wherein the differentiation cocktail comprises GM-CSF and SCF. 10. The method of any one of claims 1-9, wherein the differentiation cocktail comprises GM-CSF, SCF, and VEGF. 11. The method of any one of claims 1-10, wherein the differentiation cocktail comprises GM-CSF, SCF, VEGF, and BMP-4. 12. The method of any one of claims 1-11, wherein an iDC of the population of iDCs in step (d) expresses CD45 and CD86. 13. The method of any one of claims 1-12, wherein an iDC of the population of iDCs in step (d) does not express CD38. 14. The method of any one of claims 1-13, wherein the second period of time is between about 1 day to about 32 days. 15. The method of any one of claims 1-14, wherein step (d) comprises dynamic culturing of the cultured cells followed by static culturing of the cultured cells. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 16. The method of claim 15, wherein the dynamic culturing is for about 1 day, about 2 days, about 3 days, about 4 days, or about 5 days. 17. The method of claim 15 or 16, wherein the static culturing is for about 3 days to about 27 days. 18. The method of any one of claims 1-17, wherein the maturation cocktail comprises at least one exogenous cytokine, wherein the at least one exogenous cytokine comprises 19. The method of any one of claims 1-18, wherein the maturation cocktail comprises GM-CSF and IL-4. 20. The method of any one of claims 1-19, wherein the maturation cocktail 21. The method of any one of claims 1 to 18, wherein an mDC of the population of mDCs in step (e) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CCR7, CD141, and CD11c, but does not express one or more of CD14, TRA160, and SSEA-5. 22. The method of claim 1-21, wherein an mDC of the population of mDCs in step (e) co-expresses CD45 and CD83. 23. The method of any one of claims 1-22, wherein at least 70% of the population of mDCs in step (e) express CD45. 24. The method of any one of claims 1-23, wherein at least 70% of the population of mDCs in step (e) express CD83. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 25. The method of any one of claims 1-24, wherein at least 70% of the population of mDCs in step (e) express CD86, CD40, or HLA-A. 26. The method of any one of claims 1-25, wherein at least 20% of the population of mDCs in step (e) express HLA-DR. 27. The method of any one of claims 1-24, wherein less than 20% of the population of mDCs in step (e) express CD14. 28. The method of any one of claims 1-27, wherein less than 20% of the population of mDCs in step (e) are CD45+ and CD83-. 29. The method of any one of claims 1-22, wherein the third period of time is between about 1 day to about 10 days. 30. The method of any one of claims 1 to 29, wherein step (e) comprises static culturing of the population of iDCs. 31. The method of claim 30, wherein the static culturing is for about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, or about 10 days. 32. The method of any one of claims 1-31, wherein any one of or both of steps (d) and (e) are performed under serum free conditions. 33. The method of any one of claims 1-31, wherein any one of or both of steps (d) and (e) are performed in the absence of feeder cells. 34. The method of any one of claims 1-31, wherein any one of or both of steps (d) and (e) are performed in the absence of stromal cells. 35. The method of any one of claims 1-34, wherein steps (a), (b), and (c) are repeated after step (c), prior to step (d). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 36. The method of claim 35, wherein steps (a), (b) and (c) are each repeated three times prior to step (d). 37. The method of any one of claims 1-36, wherein steps (a), (b) and (c) combined are about three days to about five days. 38. The method of any one of claims 1-37, wherein the cultured cells of the cultured suspendable expansion complex remain undifferentiated and pluripotent in steps (a) through (c). 39. The method of claim 38, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5 and at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60. 40. The method of claim 38, wherein at least about 70% of the undifferentiated and pluripotent cells express Oct-4 and at least about 70% of the undifferentiated and pluripotent cells express Nanog. 41. The method of claim 38, wherein at least about 80% of the undifferentiated and pluripotent cells express SSEA-5, at least about 80% of the undifferentiated and pluripotent cells express TRA-1-60, at least about 70% of the undifferentiated and pluripotent cells express Oct-4, and at least about 70% of the undifferentiated and pluripotent cells express Nanog. 42. The method of any one of claims 1-41, wherein step (b) further comprises changing the growth medium to remove accumulated lactate. 43. The method of any one of claims 1-42, wherein the growth medium comprises Y-27632. 44. The method of any one of claims 35-43, wherein the culturing in step (b) is dynamic. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 45. The method of any one of claims 1-44, wherein the cultured cells in step (b) comprise hematopoietic cell intermediates. 46. The method of claim 45, wherein the hematopoietic cell intermediates express CD34, CD90. 47. The method of claim 45 or 46, wherein the hematopoietic cell intermediates do not express CD38. 48. The method of claim 45, wherein the hematopoietic cell intermediates express CD45. 49. The method of claim 48, wherein the hematopoietic cell intermediates do not express CD38. 50. The method of any one of claims 1-49, wherein the microcarrier comprises one or more of polystyrene, cross-linked dextran, magnetic particles, microchips, cellulose, hydroxylated methacrylate, collagen, gelatin, polystyrene, plastic, glass, ceramic, silicon, or a combination thereof. 51. The method of any one of claims 1-50, the microcarrier is spherical, smooth, macroporous, rod-shaped, or any combination thereof. 52. The method of any one of claims 1-51, wherein the microcarrier is not coated. 53. The method of any one of claims 1-52, wherein the microcarrier is coupled with protamine or polylysine. 54. The method of any one of claims 1-53, wherein the microcarrier has a neutral charge. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 55. The method of any one of claims 1-54, wherein the microcarrier is negatively charged. 56. The method of any one of claims 1-55, wherein the microcarrier is hydrophilic. 57. The method of any one of claims 1-56, wherein the ECM component comprises a matrigel, laminin, vitronectin, collagen, a derivative thereof, or any combination thereof. 58. The method of claim 57, wherein the laminin is human laminin. 59. The method of claim 58, wherein the human laminin is 511 E8 fragment. 60. The method of any one of claims 1-59, wherein the mDCs are characterized by: (i) expression of at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA- A, CD40, CCR7, CD141, and CD11c, but not expressing one or more of CD14, TRA160, and SSEA-5; (ii) exhibiting CCL19-induced migration; (iii) an ability to present an antigen at the cell surface; or (iv) any combination of (i), (ii), and (iii). 61. The method of any one of claims 1-60, wherein the mDCs are unable to replicate. 62. The method of any one of claims 1-61, wherein the mDCs are irradiated to prevent replication. 63. The method of any one of claims 1-62, further comprising (f) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 64. The method of any one of claims 1-62, further comprising (f) cryopreserving the population of mDCs. 65. The method of claim 64, further comprising (g) thawing the population of mDCs; and (h) contacting the population of mDCs with an antigen or a nucleic acid encoding an antigen. 66. The method of claim 63 or 65, wherein the population of mDCs is contacted with two or more antigens. 67. The method of any one of claims 63-65, wherein the antigen is a tumor antigen. 68. The method of claim 67, wherein the tumor antigen comprises a tumor associated antigen (TAA). 69. The method of claim 68, wherein the TAA is human telomerase reverse transcriptase (hTERT) or fragment thereof. 70. The method of claim 67, wherein the tumor antigen comprises a tumor- specific antigen (TSA). 71. The method of claim 67, wherein the tumor antigen comprises a neoantigen. 72. The method of claim 63 or 65, wherein the nucleic acid comprises an RNA. 73. The method of claim 72, wherein the RNA comprises an mRNA encoding hTERT or fragment thereof. 74. The method of claim 73, the wherein the fragment comprises one or more immunogenic epitopes of hTERT. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 75. The method of any one of claims 72-74, wherein the RNA comprises an mRNA encoding lysosomal-associated membrane protein 1 (LAMP1) or a fragment thereof. 76. The method of any one of claims 72-75, wherein the RNA comprises an mRNA encoding heat shock protein 96 (HSP96). 77. The method of any one of claims 63-76, wherein the population of mDCs comprising the one or more antigens is cryopreserved. 78. The method of claim 77, wherein the cryopreserved population of mDCs comprising the one or more antigens is irradiated. 79. The method of claim 78, wherein the cryopreserved population of mDCs is irradiated to prevent replication. 80. A cell produced by any one of claims 1-59, wherein the cell expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160 and SSEA-5. 81. The cell of claim 80, wherein the cell co-expresses CD45 and CD83. 82. A cell produced by any one of claims 63-77, wherein the cell (i) expresses at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but does not express one or more of CD14, TRA160, and SSEA-5; and (ii) comprises the one or more antigens. 83. The cell of claim 82, wherein the cell co-expresses CD45 and CD83. 84. A cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine comprising: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT (ii) presenting one or more antigens at the cell surface; and (b) a pharmaceutically acceptable carrier. 85. The cell vaccine of claim 84, wherein the one or more dendritic cells co- express CD45 and CD83. 86. A cell vaccine for treating or delaying progression of a disease or disorder in a subject, the cell vaccine comprising: (a) one or more dendritic cells: (i) expressing at least one of CD40, CD45, CD83, CD86, HLA-DR, HLA-A, CD40 and CCR7, but not expressing one or more of CD14, TRA160, and SSEA-5; and (ii) comprising a nucleic acid encoding one or more antigens; and (b) a pharmaceutically acceptable carrier. 87. The cell vaccine of claim 86, wherein the one or more dendritic cells co- express CD45 and CD83. 88. The cell vaccine of claim 86, wherein the nucleic acid comprises a DNA. 89. The cell vaccine of claim 88, wherein the DNA is under the control of a promoter. 90. The cell vaccine of claim 89, wherein the promoter is an inducible promoter. 91. The cell vaccine of claim 90, further comprising (c) an inducing agent. 92. The cell vaccine of claim 91, wherein the inducing agent is selected from 93. The cell vaccine of claim 86, wherein the nucleic acid comprises an RNA. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 94. The cell vaccine of any one of claims 86-93, wherein the one or more dendritic cells are induced to present the one or more antigens when contacted with the inducing agent. 95. The cell vaccine of any one of claims 86-94, wherein the cell vaccine comprises between about 0.5 x 107 dendritic cells to about 2.5 x 107 dendritic cells. 96. The cell vaccine of any one of claims 86-95, wherein the cell vaccine comprises between about 1.0 x 107 dendritic cells to about 2.0 x 107 dendritic cells. 97. The cell vaccine of any one of claims 86-96, wherein the cell vaccine comprises about 1.0 x 107 dendritic cells. 98. The cell vaccine of any one of claims 86-97, wherein the disease or disorder is cancer, an immune disease or disorder, an infectious disease or disorder. 99. The cell vaccine of claim 98, wherein the cancer is selected from breast cancer, ovarian cancer, colon cancer, prostate cancer, bone cancer, colorectal cancer, gastric cancer, lymphoma, malignant melanoma, liver cancer, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, thyroid cancers, kidney cancer, cancer of the bile duct, brain cancer, cervical cancer, maxillary sinus cancer, bladder cancer, esophageal cancer, Hodgkin's disease, and adrenocortical cancer. 100. The cell vaccine of any one of claims 86-99, further comprising one or more additional therapeutic agents. 101. The cell vaccine of claim 100, wherein the one or more additional therapeutic agents is selected from chemotherapy, adjuvants, radiation, gene therapy, surgery, one or more anti-inflammatory agents, and one or more immunomodulatory agents. 102. The cell vaccine of any one of claims 86-101, wherein the one or more antigens is identified from the subject. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 103. The cell vaccine of any one of claims 86-102, wherein the cell vaccine is formulated to be administered to the subject directly after thawing. 104. The cell vaccine of claim 103, wherein directly administering includes that dead cells are not removed from the composition prior to administration to the subject. 105. The cell vaccine of claim 103 or 104, wherein the cell vaccine is formulated to be administered without washing or reconstitution. 106. The cell vaccine of any one of claims 103-105, wherein the cell vaccine is formulated for administration within about 4 hours of thawing. 107. The cell vaccine of any one of claims 103-105, wherein the cell vaccine is formulated for administration more than 4 hours after thawing. 108. The cell vaccine of any one of claims 103-106, wherein the cell vaccine is formulated for administration within about 24 hours after thawing 109. A method of manufacturing a cell vaccine for treating cancer, the method comprising: (a) obtaining one or more mature dendritic cells (mDCs) according to the method of any one of claims 1-60; and (b) contacting the one or more mDCs with one or more nucleic acids encoding one or more antigens, thereby manufacturing the cell vaccine comprising antigen-presenting DCs for treating the cancer. 110. The method of claim 109, further comprising (c) providing one or more additional therapeutic agents. 111. The method of claim 109, wherein optionally the one or more mDCs are cryopreserved and thawed before step (b). PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 112. The method of any one of claims 109-111, wherein the one or more antigens is one or more tumor antigens. 113. The method of claim 112, wherein the one or more tumor antigens comprises a TAA. 114. The method of claim 113, wherein the TAA is hTERT. 115. The method of claim 112, wherein the one or more tumor antigens comprises a TSA. 116. The method of claim 112, wherein the one or more tumor antigens comprises a neoantigen. 117. The method of any one of claims 109-116, wherein the one or more nucleic acid molecules comprises one or more RNA molecules. 118. The method of claim 117, wherein the one or more RNA molecules comprises hTERT mRNA. 119. The method of any one of claims 109-118, wherein the manufacturing takes about 30 days to about 50 days. 120. The method of any one of claims 109-119, wherein the one or more antigens is an antigen expressed by the cancer. 121. The method of any one of claims 109-120, wherein the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. 122. A method for treating or delaying progression of a disease or disorder in a subject, comprising administering to the subject a cell vaccine of any one of claims 84 to 102. PATENT Attorney Docket No.: 058125-649001WO Client Ref. No.: LCTX-CC-600PCT 123. The method of claim 122, wherein the dendritic cells are allogeneic to the subject. 124. A method for treating cancer, comprising administering to a subject having cancer a cell vaccine made by the method of any one of claims 109-120. 125. The method of claim 124, wherein the cancer is non-small cell lung cancer (NSCLC), prostate cancer, or acute myelogenous leukemia. 126. The method of claim 124 or 125, wherein the mDCs are allogeneic to the subject.
PCT/US2023/079019 2022-11-08 2023-11-07 Compositions and method for expansion of embryonic stem cells WO2024102777A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263423501P 2022-11-08 2022-11-08
US63/423,501 2022-11-08

Publications (1)

Publication Number Publication Date
WO2024102777A2 true WO2024102777A2 (en) 2024-05-16

Family

ID=91033476

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/079019 WO2024102777A2 (en) 2022-11-08 2023-11-07 Compositions and method for expansion of embryonic stem cells

Country Status (1)

Country Link
WO (1) WO2024102777A2 (en)

Similar Documents

Publication Publication Date Title
EP2279253B1 (en) Engineering and delivery of therapeutic compositions of freshly isolated cells
EP2441468B1 (en) Immunotherapy by using cell capable of co-expressing target antigen and cd1d and pulsed with cd1d ligand
JP5156382B2 (en) Method for producing T cell population
EP2351599A1 (en) Method for producing dendritic cells
US20200208108A1 (en) Interferon primed plasmacytoid dendritic cells
JP2022065022A (en) Methods for generating engineered human primary blood dendritic cell lines
EP3362569B1 (en) Cxcr6-transduced t cells for targeted tumor therapy
JP2024506557A (en) Methods of producing modified tumor-infiltrating lymphocytes and their use in adoptive cell therapy
WO2010061930A1 (en) IMMUNOTHERAPEUTIC METHOD USING ALLO-CELLS WHICH CO-EXPRESS CD1d AND TARGET ANTIGEN
US20130189302A1 (en) Immunotherapeutic method using artificial adjuvant vector cells that co-express cd1d and target antigen
Takahashi et al. Transgenic expression of CD40L and interleukin-2 induces an autologous antitumor immune response in patients with non-Hodgkin's lymphoma
EP2647384B1 (en) IMMUNOTHERAPY USING ALLO-NKT CELLS, CELLS FOR IMMUNOTHERAPY IN WHICH alpha CHAIN OF T-CELL RECEPTOR (TCR) GENE HAS BEEN REARRANGED TO UNIFORM V alpha-J alpha , AND BANKING OF NKT CELLS DERIVED FROM SAID CELLS
WO2024102777A2 (en) Compositions and method for expansion of embryonic stem cells
AU2022259170A1 (en) Cell bank composed of ips cells for introducing t cell receptor gene
KR100975820B1 (en) Cells expressing CD40L for activation of B cells
JP5485139B2 (en) Method for producing transgenic cells
JP6782503B1 (en) A method for producing a cell composition for treating cancer, a cell composition for treating cancer produced thereby, and a method for treating cancer using the cell composition for treating cancer.
US20220233665A1 (en) Medicinal composition
EP4081634A1 (en) Immunotherapy for direct reprogramming of cancer cells into immune cells/antigen presenting cells/dendritic cells
KR101913353B1 (en) Immuno-suppressive dendritic cell-like cell and manufacturing method thereof
TW201900870A (en) Method for preparing and using embryonic mesenchymal precursor cells
EP4177344A1 (en) Novel transplantation cells having reduced immunogenicity
US20210017492A1 (en) Use of retinoic acid in t-cell manufacturing
US20130344095A1 (en) Method of using cd1d over-expression in human dendritic cells to enhance cd8+ t cell-based and invariant natural killer t cell-based antitumor immunity
SG193360A1 (en) METHOD OF USING CD1d OVER-EXPRESSION IN HUMAN DENDRITIC CELLS TO ENHANCE CD8+ T CELL-BASED AND INVARIANT NATURAL KILLER T CELL- BASED ANTITUMOR IMMUNITY