WO2024097051A1 - Compositions d'immunothérapie et procédés d'utilisation - Google Patents

Compositions d'immunothérapie et procédés d'utilisation Download PDF

Info

Publication number
WO2024097051A1
WO2024097051A1 PCT/US2023/035802 US2023035802W WO2024097051A1 WO 2024097051 A1 WO2024097051 A1 WO 2024097051A1 US 2023035802 W US2023035802 W US 2023035802W WO 2024097051 A1 WO2024097051 A1 WO 2024097051A1
Authority
WO
WIPO (PCT)
Prior art keywords
cov
sars
cells
tumor
peptide
Prior art date
Application number
PCT/US2023/035802
Other languages
English (en)
Inventor
Archis BAGATI
Original Assignee
Pyrojas 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 Pyrojas Inc. filed Critical Pyrojas Inc.
Publication of WO2024097051A1 publication Critical patent/WO2024097051A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/13Tumour cells, irrespective of tissue of origin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus

Definitions

  • FIELD OF THE DISCLOSURE [0003] The present disclosure relates in general to the field of immunotherapy and more specifically to compositions and methods of developing cellular and viral immunotherapy which confers protective immunity against human cancers. BACKGROUND OF THE DISCLOSURE [0004] While cancer vaccines are effective at preventing disease, they have limited therapeutic potential due to four major reasons. First, central and peripheral tolerance of the immune system. The immune system detects “self” antigens expressed by cancer cells and becomes tolerant towards them instead of killing them. Second, heterogeneity of tumor cells.
  • TIMEs Tumor cells that lack the antigen or have defective antigen presentation evade detection and killing by antigen specific immune cells.
  • the immune-privileged TIMEs tumor immune microenvironment. Immunosuppressive molecules and cells in the TIME limit the anti-tumor immune response. And four, most patients with advanced cancers are immunocompromised which makes treating established malignancies challenging.
  • the disclosure describes the composition of the genetic and the chemical modifications used to create autologous or allogenic cellular immunotherapies, or viral immunotherapies which stimulate the patient’s own immune system to detect a broad repertoire of cancer specific antigens and kill heterogeneous cancer cells.
  • Cancer vaccine platforms include cellular vaccines, viral or bacterial vaccines, and molecular vaccines utilizing DNA, RNA, or peptides.
  • Autologous APCs, antigen presenting cells, specifically dendritic cells are either loaded with peptide antigen or transfected with antigen genes to create DC vaccines.
  • the first FDA approved cancer vaccine sipuleucel-T (Provenge) is used for the treatment of metastatic castration-resistant prostate cancer (mCRPC).
  • This vaccine generated by enriching DCs after leukapheresis and activating them ex-vivo with a chimeric protein GM-CSF fused to the antigen PAP, are safe but have limited efficacy.
  • mCRPC metastatic castration-resistant prostate cancer
  • Several other DC vaccine-based phase I and II trials with autologous DCs pulsed with cancer antigen peptide or transduced with an adenovirus encoding the antigen are ongoing.
  • Whole cell vaccines Autologous and allogeneic whole cell vaccines have been investigated in phase II and phase III trials but have yet to receive FDA approval.
  • Autologous tumor vaccines were clinically evaluated in the 1970s by administration of patient-derived tumor cells with an adjuvant or virus to stimulate a polyclonal immune response to TAAs, tumor-specific antigens. More recently, GVAX whole tumor cell vaccine genetically modified to secrete the immune stimulatory cytokine, GM-CSF, granulocyte-macrophage colony- stimulating factor has been evaluated in autologous and allogeneic settings. GVAX enhances the recruitment and maturation of hematopoietic precursors into professional antigen presenting (APC) dendritic cells (DC).
  • APC professional antigen presenting
  • DC dendritic cells
  • GVAX induces immune responses and tumor regression in murine tumor models
  • its clinical efficacy is limited in prostate, melanoma, lung, and pancreatic cancer.
  • GM-CSF has been widely used in cancer vaccine trials with limited results.
  • Microorganisms to stimulate an immune response or deliver tumor antigens Heat- inactivated bacteria was first used by Coley to stimulate significant anti-tumor immune responses in patients with cancer.
  • a live attenuated strain of Mycobacterium bovis, Bacillus Calmette-Guérin has been used for over 35 years to treat bladder carcinoma. While several reports have rationalized how BCG induces an anti-tumor immune response, the precise cellular and molecular mechanisms remain unclear.
  • Listeria, Salmonella, Lactococcus, and Shigella are other species of bacteria have been used as effective vaccine vectors as well.
  • treatment with attenuated strains of Listeria monocytogenes results in the internalization of the bacteria by APCs, and delivery of DNA- or RNA-encoded tumor antigens 2 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO to induce potent anti-tumor immunity.
  • attenuated Listeria-based cancer vaccines have a favorable safety profile but limited efficacy.
  • the tumors express self-proteins which tolerize the immune system.
  • Tumor antigen specific vaccines generally show poor antigenicity due to immune tolerance and fail to activate a robust, clinically meaningful antitumor immune response.
  • vaccines are effective when used in a prophylactic setting but have limited efficacy in when the disease is well-established (therapeutic setting).
  • a cancer vaccine needs to overcome tolerance by either stimulating low affinity or rare TAA- reactive T cells that remain or stimulate novel TAA-reactive T cells.
  • Adjuvants, activators of antigen presentation, and serial vaccinations that promote the expansion of tumor-antigen- reactive T cells, particularly low-affinity T cells are needed.
  • presentation of the complete repertoire of tumor antigens to the immune system is needed to hedge against tolerogenic epitopes.
  • TAA-specific vaccines such as peptide or whole protein vaccines, or antigen-specific DNA or mRNA vaccines require knowledge of the pattern of expression of targetable tumor antigens, and their immunogenicity in cancer patients. The efficacy of these TAA-specific approaches relies heavily on the expression pattern and representation of target antigen in the patient’s tumor.
  • the expression pattern of the antigen is often heterogeneous and the immunogenicity of the antigen, variable. Further most cancer vaccines aim to stimulate an immune response against one or a limited set of tumor antigens. It is not sufficient to stimulate an immune response against a limited set of predicted neo-antigens for various reasons.
  • the expression of the entire tumor antigen repertoire is essential for activating an immune response against a broad repertoire of cancer antigens to target heterogeneous cancers.
  • Immunosuppressive cells and molecules in the TIME suppress anti-tumor immunity. Principally, the cytokine TGF-#, Transforming Growth Factor beta ligands TGF-#1, -#2, -#3 are the cause of the significant immunosuppression in the TIME.
  • TGF-# is ubiquitously expressed in human cancers and correlates with disease progression and poor patient prognosis. 3 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO It causes anergy of immune cells in the TIME, suppresses the cytotoxic potential of CD8 + and CD4 + T cells, and converts na ⁇ ve T cells into immunosuppressive regulatory T cells. TGF-# also potently blocks maturation of DCs by GM-CSF, the expression co-stimulatory molecules, and MHC class II, important for presentation of antigens to CD4 + T cells.
  • TGF-# directly inhibits antigen presentation capacity of macrophages and dendritic cells and limits the cytotoxic potential of cytotoxic CD8 + T cells and CD4 + T cells. Therefore, overcoming the immunosuppression induced by TGF-# in the TIME is critical to enable an effective anti-tumor immune response to persist.
  • Neutralizing anti-TGF-# antibodies, RNAi, expression of a mutated TGF- #1 precursors or dominant negatives have been utilized to neutralize TGF-# activity.
  • Tertiary Lymphoid Structures are highly differentiated compartments responsible for coordinating and guiding the differentiation and proliferation of lymphocytes.
  • Bone marrow and thymus are primary lymphoid organs where na ⁇ ve B and T lymphocytes mature from immature hematopoietic precursors.
  • Secondary lymphoid organs SLOs including the spleen, lymph nodes, and mucosal&associated lymphoid tissue (MALT), coordinate trafficking of lymphocytes and maintain immune tolerance.
  • Tertiary lymphoid organs TLOs or tertiary lymphoid structures (TLS) are highly organized aggregates of lymphocytes that accompany chronic inflammation, persistent infections, autoimmune transplant rejection, and cancer.
  • TLS are characterized by (i) a distinct T&lymphocytes rich zone enclosing a central B&cell rich area, similar to a germinal center; (ii) follicular dendritic cells (FDCs) and activated stromal mesenchymal cells (fibroblasts) (iii) plasma blasts and (iv) high endothelial venules (HEVs); blood vessels that promote the migration of na ⁇ ve lymphocytes into SLOs.
  • TLS express lymphoid associated chemokines and lymphotoxins (LTs), contain germinal centers (GCs) that are sites for in situ B cell differentiation, somatic hypermutation, oligoclonal expansion, and antibody production.
  • LTs lymphoid associated chemokines and lymphotoxins
  • GCs germinal centers
  • Chemokines are small (7–12 kDa) chemotactic polypeptides that direct lymphocyte recruitment and organize the architecture of lymphoid organs.
  • Chemokines CXCL12, CXCL13, CCL19, and CCL21 are constitutively expressed in lymphoid organs where they regulate lymphocyte migration, and segregation of B and T cells into T cell zones and germinal centers.
  • Lymphoid chemokines and LTs work in concert with numerous cytokines to shape the cellular microenvironment during tertiary lymphoid neogenesis.
  • the present disclosure relates to the field of cancer immunotherapy, offering a range of compositions and methods for the treatment of cancer and benign tumors, including leiomyomas.
  • the present disclosure provides a composition including an autologous cancer cell from a target subject, wherein the autologous cancer cell includes: (a) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces an immune response; (b) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces an inflammatory cell death response; (c) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor; or (d) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the formation of tumor-associated tertiary lymphoid structures.
  • the recombinant protein or peptide that induces an immune response is selected from one of the following proteins: (a) SARS-Cov-2 Spike (S); (b) SARS- Cov-2 Envelope (E); (c) SARS-Cov-2 Membrane (M); (d) SARS-Cov-2 Nucleocapsid (N); (e) SARS-CoV-2 Spike S-2P and RBD antigen; (f) SARS-Cov-2 ORF3a; (g) SARS-Cov-2 ORF7a; (h) SARS-Cov-2 ORF8; (i) SARS-Cov-2 Replicase 1AB; (j) Enhanced Green Fluorescent Protein (EGFP); or (k) Influenza Hemagglutinin (HA).
  • S SARS-Cov-2 Spike
  • E SARS- Cov-2 Envelope
  • M SARS-Cov-2 Membrane
  • N SARS-Cov-2 Nucleocapsid
  • the recombinant protein or peptide that induces an inflammatory cell death response is selected from one of the following proteins: (a) Gasdermin D (GSDMD); (b) Gasdermin E (GSDME); (c) Absent In Melanoma-2 (AIM2); (d) Interleukin 33 (IL33); (e) Thioredoxin-Interacting Protein (TXNIP); (f) Interleukin 1 Receptor Associated Kinase 1 (IRAK1); or (g) NLR Family Pyrin Domain Containing 3 (NLRP3).
  • GSDMD Gasdermin D
  • GSDME Gasdermin E
  • AIM2 Absent In Melanoma-2
  • IL33 Interleukin 33
  • TXNIP Thioredoxin-Interacting Protein
  • IRAK1 Interleukin 1 Receptor Associated Kinase 1
  • NLRP3 NLR Family Pyrin Domain Containing 3
  • the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor and a recombinant protein or peptide that induces that induce the formation of tumor- associated tertiary lymphoid structures is selected from one of the following proteins: (a) Chemokine Ligand 9 (CXCL9); (b) Chemokine Ligand 10 (CXCL10); (c) Chemokine Ligand 12 (CXCL12); (d) Chemokine Ligand 13 (CXCL13); (e) C-C Motif Chemokine Ligand 19 (CCL19); (f) C-C Motif Chemokine Ligand 21 (CCL21); (g) Interleukin 1 Beta (IL1#); (h) Interleukin 4 (IL4); (i) Interleukin 4 Receptor (IL4R); (j) Interleukin 21 (IL21); (k) Interleukin 9 (CX
  • the autologous cancer cell is contacted with a mixture of cytokines and chemokines.
  • at least one of the cytokines is Tumor Necrosis Factor Alpha (TNF") or Interferon Gamma (IFN$).
  • TNF Tumor Necrosis Factor Alpha
  • IFN$ Interferon Gamma
  • the present disclosure provides a pharmaceutical composition formulated for delivery with a lipid nanoparticles, electroporation, or other delivery mechanisms, including the autologous cancer cell.
  • the present disclosure provides a method for treating cancer and benign tumors (including leiomyomas) in a subject, including the composition described above. 6 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • present disclosure provides a composition including an allogenic cell including: (a) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces an immune response; (b) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces an inflammatory cell death response; (c) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor; or (d) d) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the formation of tumor-associated tertiary lymphoid structures.
  • the recombinant protein or peptide that induces an immune response is selected from one of the following proteins: (a) SARS-Cov-2 Spike (S); (b) SARS-Cov-2 Envelope (E); (c) SARS-Cov-2 Membrane (M); (d) SARS-Cov-2 Nucleocapsid (N); (e) SARS- CoV-2 Spike S-2P and RBD antigen; (f) SARS-Cov-2 ORF3a; (g) SARS-Cov-2 ORF7a; (h) SARS-Cov-2 ORF8; (i) SARS-Cov-2 Replicase 1AB; (j) Enhanced Green Fluorescent Protein (EGFP); or (k) Influenza Hemagglutinin (HA).
  • S SARS-Cov-2 Spike
  • E SARS-Cov-2 Envelope
  • M SARS-Cov-2 Membrane
  • N SARS-Cov-2 Nucleocapsid
  • recombinant protein or peptide that induces an inflammatory cell death response is selected from one of the following proteins: (a) Gasdermin D (GSDMD); (b) Gasdermin E (GSDME); (c) Absent In Melanoma-2 (AIM2); (d) Interleukin 33 (IL33); (e) Thioredoxin-Interacting Protein (TXNIP); (f) Interleukin 1 Receptor Associated Kinase 1 (IRAK1); or (g) NLR Family Pyrin Domain Containing 3 (NLRP3).
  • GDMD Gasdermin D
  • GSDME Gasdermin E
  • AIM2 Absent In Melanoma-2
  • IL33 Interleukin 33
  • TXNIP Thioredoxin-Interacting Protein
  • IRAK1 Interleukin 1 Receptor Associated Kinase 1
  • NLRP3 NLR Family Pyrin Domain Containing 3
  • the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor and a recombinant protein or peptide that induces that induce the formation of tumor- associated tertiary lymphoid structures is selected from one of the following proteins: (a) Chemokine Ligand 9 (CXCL9); (b) Chemokine Ligand 10 (CXCL10); (c) Chemokine Ligand 12 (CXCL12); (d) Chemokine Ligand 13 (CXCL13); (e) C-C Motif Chemokine Ligand 19 (CCL19); (f) C-C Motif Chemokine Ligand 21 (CCL21); (g) Interleukin 1 Beta (IL1#); (h) Interleukin 4 (IL4); (i) Interleukin 4 Receptor (IL4R); (j) Interleukin 21 (IL21); (k) Interleukin 9 (CX
  • TGF#RIII PYRO1100-3WO III
  • TGF#RIII - ectodomain, soluble
  • a fusion protein consisting of the ectodomains of one or more of: Transforming Growth Factor Beta Receptor I (TGF#RI), Transforming Growth Factor Beta Receptor II (TGF#RII), or Transforming Growth Factor Beta Receptor III (TGF#RIII) in any permutation
  • TGF#RI Transforming Growth Factor Beta Receptor I
  • TGF#RII Transforming Growth Factor Beta Receptor II
  • TGF#RIII Transforming Growth Factor Beta Receptor III
  • the allogenic cell is contacted with an autologous cancer cell from a subject, wherein the autologous cancer cell is first contacted with a mixture of cytokines and chemokines.
  • the present disclosure provides a pharmaceutical composition formulated for delivery using lipid nanoparticles, electroporation, or other suitable delivery mechanisms, including the allogenic cells described above.
  • the present disclosure provides a method for treating cancer and benign tumors (including leiomyomas) in a subject, including administering to the subject the pharmaceutical composition described above.
  • the present disclosure provides a composition including a recombinant virus including: (a) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces an immune response; (b) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces an inflammatory cell death response; (c) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor; or (d) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the formation of tumor-associated tertiary lymphoid structures.
  • the recombinant nucleic acid encoding the recombinant protein or peptide that induces an immune response is selected from a gene encoding one of the following proteins: (a) SARS-Cov-2 Spike (S); (b) SARS-Cov-2 Envelope (E); (c) SARS-Cov-2 Membrane (M); (d) SARS-Cov-2 Nucleocapsid (N); (e) SARS-CoV-2 Spike S-2P and RBD antigen (f) SARS-Cov-2 ORF3a; (g) SARS-Cov-2 ORF7a; (h) SARS-Cov-2 ORF8; (i) SARS- Cov-2 Replicase 1AB; (j) Enhanced Green Fluorescent Protein (EGFP); or (k) Influenza Hemagglutinin (HA).
  • S SARS-Cov-2 Spike
  • E SARS-Cov-2 Envelope
  • M SARS-Cov-2 Membrane
  • N SARS-Co
  • the recombinant nucleic acid encoding the recombinant protein or peptide that induces an inflammatory cell death response is selected from a gene encoding one of the following proteins: (a) Gasdermin D (GSDMD); (b) Gasdermin E (GSDME); (c) Absent 8 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • AIM2 Melanoma-2
  • IL33 Interleukin 33
  • TXNIP Thioredoxin-Interacting Protein
  • IRAK1 Interleukin 1 Receptor Associated Kinase 1
  • NLRP3 NLR Family Pyrin Domain Containing 3
  • the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor and induce the formation of tumor-associated tertiary lymphoid structures is selected from one of the following proteins: (a) Chemokine Ligand 9 (CXCL9); (b) Chemokine Ligand 10 (CXCL10); (c) Chemokine Ligand 12 (CXCL12); (d) Chemokine Ligand 13 (CXCL13); (e) C-C Motif Chemokine Ligand 19 (CCL19); (f) C-C Motif Chemokine Ligand 21 (CCL21); (g) Interleukin 1 Beta (IL1#); (h) Interleukin 4 (IL4); (i) Interleukin 4 Receptor (IL4R); (j) Interleukin 21 (IL21); (k) Interleukin 22 (IL22); (l) Interleukin
  • the present disclosure provides a pharmaceutical composition for delivery to a subject using lipid nanoparticles, electroporation, or other suitable delivery mechanisms, including the recombinant virus or delivery system described above.
  • the present disclosure provides a method for treating cancer and benign tumors (including leiomyomas) in a subject, including administering to the subject the pharmaceutical composition described above.
  • the present disclosure provides a composition including an allogenic T cell, wherein the allogenic T cell includes: (a) a chimeric antigen receptor (CAR) or an antigen specific T cell receptor (TCR) that makes the T cells antigen specific; and (b) a recombinant protein or peptide or mRNA encoding the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor. (c) a recombinant protein or peptide or mRNA encoding the 9 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • CAR chimeric antigen receptor
  • TCR antigen specific T cell receptor
  • the chimeric antigen receptor (CAR) or an antigen specific T cell receptor (TCR) is specific for one of the following proteins: (a) SARS-Cov-2 Spike (S); (b) SARS-Cov-2 Envelope (E); (c) SARS-Cov-2 Membrane (M); (d) SARS-Cov-2 Nucleocapsid (N); (e) SARS-CoV-2 Spike S-2P and RBD antigen; (f) SARS-Cov-2 ORF3a; (g) SARS-Cov-2 ORF7a; (h) SARS-Cov-2 ORF8; (i) SARS-Cov-2 Replicase 1AB; (j) Enhanced Green Fluorescent Protein (EGFP); or (k) Influenza Hemagglutinin (HA).
  • SARS-Cov-2 Spike S
  • SARS-Cov-2 Envelope E
  • M SARS-Cov-2 Membrane
  • N SARS-Cov-2 Nucleocapsid
  • the recombinant protein or peptide that induces the activation and persistence of antigen-presenting cells and cytotoxic T cells within the microenvironment of a tumor and induce the formation of tumor-associated tertiary lymphoid structures is selected from one of the following proteins: (a) Chemokine Ligand 9 (CXCL9); (b) Chemokine Ligand 10 (CXCL10); (c) Chemokine Ligand 12 (CXCL12); (d) Chemokine Ligand 13 (CXCL13); (e) C-C Motif Chemokine Ligand 19 (CCL19); (f) C-C Motif Chemokine Ligand 21 (CCL21); (g) Interleukin 1 Beta (IL1#); (h) Interleukin 4 (IL4); (i) Interleukin 4 Receptor (IL4R); (j) Interleukin 21 (IL21); (k) Interleukin 22 (IL22); (l) Interleukin 22
  • the present disclosure provides a pharmaceutical composition suitable for delivery to a subject using lipid nanoparticles, electroporation, or other suitable delivery mechanisms, including the allogenic cell described above.
  • the present disclosure provides a method for treating cancer and benign tumors (including leiomyomas) in a subject, including administering to the subject the pharmaceutical composition. 10 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • the present disclosure provides a composition including a recombinant DNA, an mRNA, lipid nanoparticles, or an electroporation system, wherein the recombinant DNA or mRNA encodes a recombinant protein or peptide that induces an immune response.
  • the recombinant nucleic acid encoding the recombinant protein or peptide that induces an immune response is selected from the gene encoding one of the following proteins: (a) SARS-Cov-2 Spike (S); (b) SARS-Cov-2 Envelope (E); (c) SARS-Cov- 2 Membrane (M); (d) SARS-Cov-2 Nucleocapsid (N); (e) SARS-CoV-2 Spike S-2P and RBD antigen (f) SARS-Cov-2 ORF3a; (g) SARS-Cov-2 ORF7a; (h) SARS-Cov-2 ORF8; (i) SARS- Cov-2 Replicase 1AB; (j) Enhanced Green Fluorescent Protein (EGFP); or (k) Influenza Hemagglutinin (HA).
  • the present disclosure provides a pharmaceutical composition for delivery to the target subject using lipid nanoparticles, electroporation, or other delivery mechanisms, including the recombinant DNA, mRNA, or delivery system described above.
  • the present disclosure provides a method for treating cancer and benign tumors (including leiomyomas) in a subject, including administering to the subject the pharmaceutical composition described above. + 639.BRIEF DESCRIPTION OF THE DRAWINGS [0046]
  • FIGURE 1A is a schematic diagram illustrating the process and design to generate PyroCells TM cellular immunotherapies.
  • FIGURE 1B is a schematic diagram illustrating the process and design to generate Allo-PyroCellTM immunotherapies.
  • FIGURE 1C is a schematic diagram illustrating the process and design to generate PyroVirTM viral immunotherapies.
  • FIGURE 2 is a schematic diagram illustrating the concept that challenges associated with therapeutic cancer vaccines and how PyroVax TM vector encoding PyroHIM TM , PyroACT TM , and PyroTIME TM can be used to overcome several of these challenges (right).
  • FIGURE 3 is a schematic diagram illustrating PyroVax TM components PyroHIM TM , PyroACT TM , and PyroTIME TM in a non-viral expression plasmid, including PyroVax TM in a lentiviral vector.
  • FIGURE 4 is a schematic diagram illustrating PyroVax TM components PyroHIM TM , PyroACT TM , and PyroTIME TM in a non-viral expression plasmid, including PyroVax TM in an AAV vector.
  • FIGURE 5 is a schematic diagram illustrating PyroVax TM components PyroHIM TM , PyroACT TM , and PyroTIME TM in a non-viral expression plasmid, including PyroVax TM in an AAV vector.
  • FIGURE 6 is a schematic diagram illustrating PyroVax TM components PyroHIM TM , PyroACT TM , and PyroTIME TM in a non-viral expression plasmid, including a backbone.
  • FIGURE 7A is a graph illustrating contour plots showing expression of indicated proteins in human TNBC cells transduced with lentiviral vectors encoding the indicated genes.
  • FIGURE 7B is a graph illustrating quantification of data from FIGURE 7A.
  • FIGURE 7C is a set of graphs illustrating contour plots showing expression levels of indicated constructs using a non-viral approach.
  • FIGURE 7D is a set of graphs illustrating flow plots showing the tetramer positive CD8+ T cells present in the blood of the donor described on the left.
  • FIGURE 7E is a chart describing a certificate of analysis highlighting critical information about the donor from who the T cells shown in FIGURE 7D and FIGURE 7E were isolated.
  • FIGURE 7F is a set of graphs illustrating A IFN$ assay (top) and a cytotoxicity assay (bottom) testing the cellular response to peptides encoding the T cells specific-reactive peptide.
  • FIGURE 8A is a set of bioluminescent images illustrating the immunological rejection of PyroCellsTM over time. By three weeks there is no detectable presence of PyroCells TM (left). On Day 22, vaccinated mice were challenged with parental tumor (red) or not (blue) and compared to unvaccinated controls which also received the same parental tumor. While vaccinated mice were protected from tumor challenge, all unvaccinated mice succumb to disease and died. Day 28 is 7 days after tumor challenge.
  • FIGURE 8B is a line graph quantifying the bioluminescence signal in FIGURE 8A. Data is represented as mean and standard deviation.
  • FIGURES 9A and FIGURE 9B are schematic representations of the PyroTIMER construct, highlighting the fusion of ectodomains of TGF#RI and TGF#RII, integrated into Jurkat T cells.
  • FIGURE 9C is a set of graphs illustrating a co-culture killing assay using PyroTIMER CD19 CAR-T cells and CD19 antigen-expressing Raji lymphoma cells. Displayed 12 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • FIGURE 9D is a set of graphs illustrating stability assays showing the persistence of PyroTIMER and CD19 CAR expression in Jurkat T cells over time, post-transduction.
  • FIGURE 9E is a graphical representation of the effector-to-target ratios used in the co-culture killing assay, highlighting the superior killing efficacy of PyroTIMER CD19 CAR- T cells across all ratios.
  • FIGURE 10A is a set of graphs illustrating generation and functional characterization of PyroTIMER CD19 CAR-T cells using primary human CD3 + T cells, including contour flow plots showing purity of enrichment of CD3+ T cells from human donor PBMCs.
  • FIGURE 10B is a graph illustrating quantification of FIGURE 10A.
  • FIGURE 10C is a set of graphs illustrating contour plots showing the frequency of CD4+ (top) and CD8+ (bottom) PyroTIMER CD19 CAR-T cells following enrichment by cell sorting.
  • FIGURE 10D is a set of graphs illustrating contour flow plots showing the frequency of CD4+ and CD8+ among the CD3+ T cells.
  • FIGURE 10E is a graph illustrating quantification of FIGURE 10D.
  • FIGURE 10F is a set of graphs illustrating contour flow plots showing the generation of CD19 CAR-T cells at high efficiency after chemical selection (puromycin) and cell sorting (RFP).
  • FIGURE 10G is a graph illustrating the number of live CD19 expressing Raji lymphoma cells, 24hrs after being in co-culture with either CD19 CAR-T cells (CAR) or PyroTIMER CD19 CAR-T cells (PyroTIMER CAR) at indicated effector to target (E:T) ratios in the presence of TGF #1.
  • FIGURE 10H is a set of graphs illustrating the percentage of CD69 + activated T cells in FIGURE 10G. Un-transduced T cells were used as controls. Experiments was replicated twice in technical triplicates. Data is a percentage of total and represented as an average +/- S.E.M. An unpaired two-tailed student t test was used to compared the killing (in G) and the CD69+ cells (in H) in either co-culture (CAR vs PyroTIMER CAR) at specific E:T ratios. A p ⁇ 0.05 was considered statistically significant.
  • FIGURES 11A and FIGURE 11B are a set of images and a graph illustrating results of an in vivo study using NOD.Cg-PrkdcscidIL2Rgtm1Wjl/Sz mice illustrating the efficacy of PyroTIMER CAR-T cells against CD19+ Raji lymphoma disease burden.
  • the comparative 13 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO tumor growth curves and survival rates between PyroTIMER CAR-T cell-treated mice and traditional CD19 CAR-T cell-treated mice are showcased.
  • the present disclosure relates to the field of cancer immunotherapy, offering a range of compositions and methods for the treatment of cancer and benign tumors, including leiomyomas.
  • the disclosure leverages the therapeutic potential of autologous and allogenic cells, particularly cancer cells and T cells, which have been genetically modified or contacted with specific recombinant proteins, or nucleic acids encoding the recombinant proteins or peptides. These modified cells are primed to elicit various immune and inflammatory responses in the tumor microenvironment, thereby bolstering the body's natural ability to recognize and destroy tumor cells.
  • Such autologous cells include cells where the immune response is triggered by proteins produced by viruses like SARS-CoV-2 or other entities like EGFP and Influenza Hemagglutinin.
  • Autologous cells of the present disclosure also include primed to induce inflammatory cell death by targeting or inhibiting proteins such as Gasdermins, AIM2, and IL33.
  • Further autologous cells include cells enhanced to bolster the activation of immune cells and the formation of lymphoid structures in the tumor environment, leveraging proteins or inhibitors like CXCL9, CCL21, IL1#, and PD-L1, among others.
  • Allogenic cells including T cells, are modified similarly to the autologous cells, allowing for a broader applicability in treatments.
  • Recombinant viruses are engineered to deliver the same therapeutic entities directly into the tumor.
  • Recombinant DNAs, mRNAs, lipid nanoparticles, or electroporation systems encoding specific proteins or peptides, especially those inducing an immune response, are described in the present disclosure.
  • the disclosure provides pharmaceutical compositions suitable for delivery to the subject using mechanisms like lipid nanoparticles and electroporation, ensuring efficient uptake and functionality of the therapeutic entities.
  • the disclosure offers methods for treating malignancies and benign tumors in subjects using the described compositions.
  • Immune cells continuously survey the body for pathogens like bacteria and viruses, or normal cells that might be becoming cancerous and kills them. However, in subjects who are immunocompromised, the heterogeneity of tumor cells allows for sub-populations of cells to evade detection and killing by immune cells and establish disease. These immune evasive tumor cells either do not present antigens that are vulnerable to detection by immune cells or completely halt antigen presentation. Since these cancer cells express most of the proteins and antigens that normal cells express, the immune system recognizes the cell as “self” and becomes “tolerant” of the cancer cells.
  • This disclosure describes a novel autologous and allogeneic cellular immunotherapy, and a viral immunotherapy which break central and peripheral immune tolerance against tumor antigens, enhances the immunogenicity of cancer cells, and remodels the immunosuppressive TIME.
  • the approach emphasizes the presentation of the entire available repertoire of subject tumor specific antigens to activate an anti-tumor immune response against a broad repertoire of cancer specific antigens. Notably, this approach redirects an existing repository of anti-viral immune cells to treat cancer. This approach is important because subjects with cancer are often immunocompromised and do not contain a robust repository of na ⁇ ve immune cells to develop a significant anti-tumor immune response.
  • isolated indicates that the cell or cells, or the peptide(s) or nucleic acid molecule(s) has/have been removed from its/their normal physiological environment, e.g. a natural source, or that a peptide or nucleic acid is synthesized.
  • isolated indicates that a naturally occurring sequence has been removed from its normal cellular (i.e., chromosomal) environment.
  • the sequence may be in a cell-free solution or placed in a different cellular environment.
  • An isolated cell or isolated cells may for instance be included in a different medium such as an aqueous solution than provided originally, or placed in a different physiological environment.
  • isolated cells, peptides or nucleic acid molecule(s) constitute a higher fraction of the total cells, peptides or nucleic acid molecule(s) present in their environment, e.g., solution/ suspension as applicable, than in the environment from which they were taken.
  • treatment refers to a prophylactic or preventative measure having a therapeutic effect and slowing down, or at least partially alleviating or abrogating an undesirable condition in the organism of a subject.
  • Those in need of treatment include those already with the condition as well as those prone to having the condition or those in whom the condition is to be prevented (prophylaxis).
  • a treatment reduces, stabilizes, or inhibits progression of a symptom that is associated with the presence and/or progression of an undesirable condition.
  • therapeutic effect refers to the inhibition or activation of factors causing or contributing to the undesirable condition. A therapeutic effect relieves to some extent one or more of the symptoms of an undesirable condition or disease.
  • administer refers to any way a treatment is given to a patient or subject.
  • administer refers to any mode of transferring, delivering, introducing, or 16 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO transporting matter such as a compound, e.g. a pharmaceutical compound, or other agent such as an antisense oligonucleotide, to a subject.
  • Administering can be accomplished via topical, intravenous, intramuscular, systemic, oral, or parenteral methods.
  • the present disclosure provides the composition of and methods to develop an autologous cellular immunotherapy, namely PyroCells TM .
  • PyroCells TM These are composed of patient derived (autologous) tumor cells which are modified ex-vivo to express the PyroVax TM vector and chemically modified with PyroStim TM .
  • PyroCells TM activate an immune response against a broad repertoire of tumor specific antigens conferring long term recurrence free survival in patients with advanced solid and hematological cancers.
  • Another embodiment of the present disclosure provides the composition of and methods to develop auto-allogeneic cellular immunotherapies namely, Allo-PyroCell TM , which are created by combining ex-vivo PyroStim TM treated autologous patient tumor derived cells with allogeneic cells (normal or transformed) which have been modified ex-vivo to express the PyroVax TM vector.
  • Allo-PyroCell TM activate a polyclonal immune response against a broad repertoire of tumor specific antigens conferring long-term recurrence free survival.
  • this allogeneic approach overcomes several manufacturing, technical, and cost challenges associated with an autologous approach.
  • Another embodiment of the present disclosure provides the composition of and methods to develop viral immunotherapies, namely, PyroVir TM .
  • the virus is delivered intratumorally and incorporate the components of PyroVax TM namely, PyroHIM TM , PyroActTM , PyroTIME TM , as well as PyroStim TM into the tumor to activate a polyclonal immune response against a broad repertoire of tumor specific antigens conferring long-term recurrence free survival.
  • the following section summarizes the three major components of the PyroVax TM vector namely, PyroHIM TM , PyroAct TM , and PyroTIME TM , and the composition of the chemicals with relevant doses which make up PyroStim TM . It also summarizes the methods to use these components to create PyroCells TM , Allo-PyroCells TM , and PyroVir TM . [0091] The present disclosure provides the composition of nucleotide sequences used to generate PyroVax TM vector.
  • the PyroVax TM vector is comprised of nucleic acids encoding three essential ORFs, open reading frames, among various other regulatory components.
  • PyroHIM TM a highly immunogenic molecule expressed at supraphysiological levels which are non-self and have high antigenic potential
  • PyroAct TM an activator of 17 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO inflammatory immunogenic cell death
  • PyroTIME TM a remodeler of the immunosuppressive TIME, tumor immune microenvironment.
  • Table 1 showing PyroVax TM components [0093] The present disclosure describes a method of making an autologous cellular immunotherapy (PyroCell TM ) including: a) Harvesting the surgically resected tumor tissue in the sterile container and ship to PyroLabs TM b) Dissociation of the tumor mechanically and chemically into a single cell suspension. 18 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO c) Enrichment of tumor cells using antibody mediated negative selection. d) Electroporation or viral transduction of autologous tumor cells to incorporate P yroVaxTM vector.
  • the present disclosure describes a method of making a combination of an autologous and allogeneic cellular immunotherapy (Allo-PyroCell TM ) including: a) Harvesting the surgically resected autologous tumor tissue in the sterile container. b) Dissociation of the tumor mechanically and chemically into a single cell suspension. c) Enrichment of tumor cells using antibody mediated negative selection.
  • the present disclosure describes a method of making a cancer-specific viral immunotherapy (PyroVir TM ) including: a) Generation of AAV and/or onco-specific viruses (HSV) containing PyroVax TM vectors for intra-tumoral delivery.
  • the present disclosure describes a method of redirecting an immune response generated in a prophylactic setting towards cancer cells in a therapeutic setting. Specifically, the disclosure describes the composition of the genetic and chemical modifications and the methods used to generate autologous or allogeneic cellular immunotherapies which re-direct the immune cells that detect SARS-Cov-2 antigens and control viral infection to detect cancer antigens and kill cancer cells.
  • Autologous or allogeneic cells are modified ex-vivo such that when introduced into the subject, these cellular immunotherapies stimulate an immune response or redirect an existing immune repository to detect cancer specific antigens and kill cancer cells.
  • cells genetically modified to manufacture the cellular immunotherapy may be autologous, expansion of cells derived from a xenograft of the autologous tumor cells, allogeneic tumor cells, allogeneic tumor cells expanded in xenografts, or a combination of one or more.
  • allogenic cells are established from normal or transformed cell lines, or normal fibroblasts, or endothelial cells, or immortalized cell lines.
  • HIMs include full length proteins or immunogenic regions selected from the proteins including but not limited to enhanced green fluorescent protein (eGFP), the full-length protein or peptide derivatives of the SARS-CoV-2 (accession number NC_045512.2) structural proteins, including the spike glycoprotein (S), envelope protein (E), membrane protein (M), and nucleocapsid phosphoprotein (N), among others (see table 1).
  • eGFP enhanced green fluorescent protein
  • S spike glycoprotein
  • E envelope protein
  • M membrane protein
  • N nucleocapsid phosphoprotein
  • PyroHIM TM the full-length protein or immunogenic peptides produced by the SARS-Cov-2 virus are encoded as PyroHIMs TM in the PyroVax TM construct operably linked with an activator of pyroptotic cell death (PyroAct TM ), and a remodeler of the TIME, tumor immune microenvironment (PyroTIME TM ). 20 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • PyroHIMs TM encode for the full-length protein(s) or peptide(s) of the SARS-CoV-2 (accession number NC_045512.2) structural proteins, including the spike glycoprotein (S), envelope protein (E), membrane protein (M), and nucleocapsid phosphoprotein (N).
  • the immunogenic peptides are used alone, or in combination with other peptide fragments connected with linkers or not to enhance immunogenicity.
  • the epitopes used to develop the FDA approved vaccinations by Moderna and Pfizer among other vaccine manufacturers are used.
  • the activation of pyroptosis is achieved using SARS- Cov-2 antigens ORF3a and envelope protein (E) which are direct inducers of pyroptosis.
  • SARS-Cov-2 antigens ORF3a and envelope protein (E) which are direct inducers of pyroptosis.
  • PyroCells TM containing PyroVax TM vectors encoding SARS-Cov-2 specific epitopes as PyroHIMs TM redirects the immune response generated against SARS-Cov-2 to treat cancer.
  • PyroCells TM cellular immunotherapies are effective in both prophylactic and therapeutic settings, able to control disease, and significantly extend overall survival.
  • PyroCells TM containing PyroVax TM vectors encoding SARS-Cov-2 specific epitopes as PyroHIMs TM activate a pre-existing pool of anti-viral cytotoxic CD8 + and CD4 + T cells and redirect it against cancer.
  • PyroHIM TM epitopes are linked to a reporter molecule to enable supraphysiological expression, detection, and enrichment of autologous or allogeneic PyroCells TM that express the PyroVax TM vector.
  • PyroCells TM containing PyroVax TM vectors encoding SARS-Cov-2 specific epitopes as PyroHIMs TM are operably linked together or not, derived from reactive T and B cell epitopes most prevalent in humans exposed to or vaccinated against SARS-Cov-2.
  • EGFP Enhanced green fluorescent protein
  • AAB02572 239 aa, Accession: AAB02572, Version: AAB02572.1 (SEQ ID NO:1) 1 mvskgeelft gvvpilveld gdvnghkfsv sgegegdaty gkltlkfict tgklpvpwpt 61 lvttltygvq cfsrypdhmk qhdffksamp egyvqertif fkddgnyktr aevkfegdtl 121 vnrielkgid fkedgnilgh kleynynshn vyimadkqkn gikvnfkirh niedgsvqla 181 dhyqqntpig dgpvllpdnh ylstqsalsk dpne
  • GFP is a model non-self-antigen to test the utility of PyroHIMs TM in PyroVax TM because well- established in vitro and in vivo tools and model systems utilizing GFP as an antigen exist and are well validated (4, 5).
  • Immunosuppressive molecules and cells in the TIME, tumor immune microenvironment [0119] PyroTIME TM is the component of the PyroVax TM vector that encodes for the molecules which remodel the immunosuppressive TIMEs by promoting tumor antigen presentation by APCs and killing by cytotoxic immune cells.
  • PyroTIME TM works by suppressing immunosuppressive molecules and cells in the TIME and promotes an environment that generates TS-TLS, tumor specific-tertiary lymphoid structures. Specifically, PyroTIME TM inactivates the expression of specific genes and/or increase the expression of specific cytokines, chemokines, and soluble receptor antagonists which promote the recruitment of T cells (organized into T cell zones), B-cells (organized as a germinal center), and follicular dendritic cells giving rise to tumor specific tertiary lymphoid structures. [0120] The present disclosure describes the composition of and methods to remodel the immunosuppressive TIME by suppressing the levels of the immunosuppressive cytokine, TGF- f in the TIME.
  • PyroTIME TM encodes for competitive antagonists of TGFf including soluble TGF-f$receptors (TGFfRII or TGFfRIII), or dominant negative receptors.
  • PyroTIME TM encodes guide RNAs which inactivate specific TGFf signaling genes including SMAD2/3, TGFfRII, TGFfRIII, among others (see table 1, PyroTIME TM ).
  • the gRNA (crRNA + trRNA tracer RNA) and Cas9 are complexed as RNPs and electroporated or nucleofected into host cells.
  • the gRNA and Cas9 are encoded in an expression vector.
  • PyroTIME TM is composed of nucleotide sequences which encode for genes that express soluble or dominant negative TGFfRII and/or TGFfRIII to inhibit the levels of TGFf in the TIME. 22 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO [0123] Another embodiment of the present disclosure describes the composition of and methods to remodel the immunosuppressive TIME to promote increased intra-tumoral infiltration and persistence of immune cells using PyroTIME TM .
  • the disclosure describes the composition of PyroTIME TM , DNA sequences encoding for molecules CXCL13 and CCL19 which promote recruitment of B cells, dendritic cells, cytotoxic T cells, formation of germinal centers, T cell zones, and finally tertiary lymphoid structures containing follicular dendritic cells.
  • the present disclosure describes PyroTIME TM , a component of PyroVax TM which remodels the immunosuppressive TIME and promotes the formation of tumor specific TLSs, Tertiary Lymphoid Structures for durable anti-tumor immune surveillance.
  • PyroTIME TM encodes molecules important for the recruitment, and maturation of dendritic cells, especially follicular dendritic cells which have significant potential to cross- present tumor antigen. These include but are not limited to either of the following, used alone, or in combination with one or more additional molecule: CXCL13, CXCL12, CCL19, CCL21, soluble TGFfRI/II/III, IL-13, IL-4, IL-4R, IL-21, IL-22, IL-22R, IL-23.
  • Molecules that are inactivated to promote antigen presentation and tumor cell uptake by APC include CD274, CD47, TGFfR, SMAD2/3, among others (See table 1).
  • the PyroTIME TM construct is expressed in normal (fibroblast or stromal cells), and/or transformed cells to generate PyroTIMECells TM . These stromal fibroblast-like cells are mixed with tumor cells or tumor lysates to generate tumor specific TLS, TS-TLS in vivo.
  • PyroTIMECells TM create a persistent anti-tumor immune response against a broad repertoire of tumor specific antigens conferring significant anti-tumor immunity
  • PyroTIMECells TM are developed from normal cells derived from an autologous or allogeneic setting and in another aspect from transformed cells, derived from an autologous or allogeneic setting.
  • the present disclosure describes the composition of chemical and methods to stimulate antigen presentation and type I and II interferon signaling in tumor cells using PyroStim TM .
  • the present disclosure describes the composition of and methods to stimulating antigen presentation in tumor cells by expressing B2M, beta-2-macroglobulin gene using PyroStim TM .
  • Limited diversity of anti-tumor immune cells 23 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO
  • the present disclosure describes the composition of and methods to activate inflammatory tumor cell death to cause the endogenous T-cell response against antigens to be activated and diversified towards non–cross reactive epitopes selected from the same antigen (intramolecular spreading) or other antigens (intermolecular spreading) using PyroAct TM .
  • the activation signal for PyroAct TM is functionally linked to PyroHIM TM such that the antigen encoded in PyroHIM TM stimulates an immune response which drives selective inflammatory cell death.
  • This antigen-restricted approach to activation of pyroptosis allows for targeted activation of epitope spreading in tumors.
  • the present disclosure describes the composition of and methods to develop autologous or allogeneic PyroCell TM which redirect the existing immune repertoire previously generated against SARS-CoV-2 Spike S-2P and RBD antigen or influenza antigens in patients prophylactically vaccinated or those that recovered from the disease to treat cancer.
  • the present disclosure describes the composition of and methods to develop a live cancer vaccine containing the construct, PyroKill TM , which allows for the killing of any injected tumor cells that were not rejected by the immune system. [0136] Attenuation of live vaccines negatively impacts cytokine production [0137] The present disclosure describes the composition of and methods to develop a live cancer vaccine containing the construct, PyroKill TM , which allows for the killing of any injected tumor cells that were not rejected by the immune system.
  • Allo- PyroCells TM which is composed of patient derived autologous tumor cells mixed with an allogeneic cell line (Allo) expressing PyroHIM TM , PyroAct TM , and PyroTIME TM . Auto cells are treated with PyroStim TM prior to mixing with Allo-PyroCells TM .
  • the source of the Allo-PyroCells TM would be tumor cells derived from allogeneic tumor cells, allogeneic tumor cells expanded in xenografts, normal cells including fibroblasts, macrophages, or dendritic cells, or a combination of one or 24 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO more.
  • B2M deficient Allo-PyroCells TM would be created and used to avoid cross-reactivity with allogenic epitopes.
  • the present disclosure describes the composition of and methods to develop Auto- Allo-PyroCell TM which is composed of patient derived autologous tumor cells (Auto) mixed with an allogeneic cell line expressing PyroHIM TM , PyroAct TM , and PyroTIME TM .
  • Auto patient derived autologous tumor cells
  • PyroHIM TM patient derived autologous tumor cells
  • PyroAct TM PyroAct TM
  • PyroTIME TM PyroTIME TM
  • Elaborate timelines for manufacturing of personalized vaccines are long extending out to approximately 4 months and rely on computationally derived prediction of TAA. Advanced and metastatic human cancers grow aggressively and don’t have time and the power of prediction is poor.
  • the present disclosure describes methods to generate PyroCell TM in 2 weeks.
  • the present disclosure describes methods to generate Allo-PyroCell TM in 1 week.
  • Ex-vivo culture associated modifications [0149] The present disclosure describes the composition of and methods to develop PyroVir TM which is a viral construct expressing PyroHIM TM , PyroAct TM , PyroTIME TM , and PyroStim TM delivered intratumorally avoid the need for ex-vivo modifications.
  • PyroVir TM is a viral construct expressing PyroHIM TM , PyroAct TM , PyroTIME TM , and PyroStim TM delivered intratumorally avoid the need for ex-vivo modifications.
  • the two oncolytic HSV2 vectors were developed from the HG52 (6) strain.
  • Modifications include deletion of the ICP47 and ICP34.5 genes and insertion of a GFP expression cassette expressing PyroHIM TM , PyroAct TM , PyroTIME TM , and PyroStim TM .
  • Patients with advanced cancers are often immunodeficient
  • Patients with advanced solid human cancers do not contain a robust repository of cytotoxic immune cells for various reasons including central and peripheral immune tolerance, age, immunosuppressive conditioning, chemotherapy, among others (1).
  • the present disclosure describes the composition of and methods to develop autologous or allogeneic PyroCell TM which redirects the existing immune repository against SARS-CoV-2 or influenza antigens to treat cancer.
  • the genetically modified cellular therapies can be administered as a stand-alone therapy, or in combination with other therapies.
  • the PyroVax TM vaccine may be combined with another therapeutic agent including, but not limited to IFN$, checkpoint inhibitors (anti-PD1, anti-CTLA-4), and adoptive T cell therapies.
  • the present disclosure PyroCell TM can also be used in combination with monoclonal antibodies against targets including, but not limited to, PD-1, PD-L1, CTLA- 25 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • the disclosure describes the composition of the genetic and chemical modifications used to create autologous or allogenic cellular immunotherapies, or viral immunotherapies. These immunotherapies stimulate the patient’s own immune system to detect a broad repertoire of cancer specific antigens and kill heterogeneous cancer cells, conferring patients long term immunity from their cancer.
  • the disclosure describes PyroCells TM cellular immunotherapies which are created by genetically and chemically modifying the patient’s own tumor cells.
  • PyroCells TM When re-introduced into the same patient PyroCells TM stimulate an anti-tumor immune response against a broad repertoire of cancer specific antigens.
  • PyroCells TM immunotherapies are effective in a prophylactic and therapeutic setting helping control disseminated and advanced heterogeneous human cancers.
  • PyroVax TM DNA or mRNA encoding vectors are used to genetically modify autologous or allogeneic tumor and normal cells and turn them into cellular immunotherapies which activate a significant anti-tumor immune response against a broad repertoire of cancer specific antigens.
  • PyroVax TM has three components: PyroHIM TM , PyroAct TM , and PyroTIME TM . Together, these components help break immune tolerance against tumor specific antigens and activate a polyclonal immune response against a broad repertoire of tumor specific antigens.
  • PyroHIM TM encodes for a highly immunogenic molecule which is non-self and expressed at supra-physiological levels.
  • PyroActTM encodes for an activator of inflammatory cell death which triggers immunological epitope spreading and diversification of the anti-tumor T cell repertoire.
  • PyroTIME TM encodes for molecules which remodel the immunosuppressive TIME, tumor immune microenvironment allowing cytotoxic immune cells to persist and kill tumor cells.
  • PyroVax TM is a DNA plasmid or mRNA vector(s) encoding: i) PyroHIMs TM , highly immunogenic molecules, ii) PyroActs TM , molecules that stimulate inflammatory cell death, and PyroTIME TM , molecules that remodels the immunosuppressive TIME, and support activation, 26 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • FIG. 1A shows schematic of the PyroVax TM vector(s), table below shows examples of each of the three components).
  • the SARS-Cov-2 epitopes shown below are representative examples of peptide sequences of partial length or full length proteins that would be used as model antigens in humans and murine models (see table 1 and claims for others).
  • Human Epitopes 27 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO
  • the full-length protein or peptide derivatives of the Influenza A structural proteins including the HA antigen (HA) and epitopes utilized for 30 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO immunization, are used as antigens.
  • Immunogenic epitopes are used alone, or in combination connected with linkers or not to enhance immunogenicity.
  • CTL Cytotoxic T lymphocyte
  • LBL 16-mer linear B-lymphocyte epitopes were identified using a threshold of 0.5 (ABCpreds server).
  • the ElliPro tool of IEDB was utilized to predict linear and conformational B-cell epitopes.
  • Polypeptide structure validation The ProtParam server is used to interrogate the physical and chemical properties of the construct, such as amino acid composition, molecular weight, theoretical isoelectric point (pI), grand average of hydropathicity (GRAVY), aliphatic and instability index, and half-life.
  • the secondary structural properties of the polypeptide were analyzed using SOPMA server, modeled and refined the 3D structure model using the GalaxyWEB server.
  • RAMPAGE server and ProSA-web tools were used to validate the refined 3D model.
  • PyroHIMTM (Highly Immunogenic Molecules)
  • PyroHIMTM encodes highly immunogenic molecules. These molecules are designed to provoke a robust immune response when introduced into the patient's body. They act as antigens, triggering the immune system to recognize them as foreign and initiate an immune response. PyroHIMTM is able to make cancer cells more visible to the immune system. Cancer cells often evade the immune system by appearing similar to healthy cells. By introducing highly immunogenic molecules, PyroHIMTM helps the immune system recognize and attack cancer cells more effectively.
  • PyroHIMTM can include epitopes (small protein fragments) from various sources, such as SARS-CoV-2 proteins, Influenza A antigens, or other tumor-specific antigens. These 31 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO epitopes are chosen because they are known to stimulate a strong immune response. By encoding these epitopes in the PyroVaxTM vector, the immune system is primed to target cancer cells displaying these antigens.
  • PyroActTM Activator of Inflammatory Cell Death
  • PyroActTM encodes molecules that trigger inflammatory cell death, a process known as pyroptosis.
  • Pyroptosis is a form of programmed cell death that results in the release of pro- inflammatory signals, which further stimulate the immune system.
  • PyroActTM amplifies the immune response. When cancer cells undergo pyroptosis, they release signals that attract immune cells to the site of cell death. This not only eliminates the cancer cell but also recruits more immune cells to the tumor microenvironment. PyroActTM encodes molecules like Gasdermin E (GSDME) or other proteins associated with pyroptosis. When these molecules are expressed in cancer cells, they lead to pyroptosis upon exposure to specific triggers. This results in the release of danger signals, such as cytokines, which alert the immune system to the presence of cancer cells.
  • GSDME Gasdermin E
  • PyroTIMETM (Tumor Immune Microenvironment Remodeler): [0177] PyroTIMETM encodes molecules that remodel the tumor immune microenvironment (TIME). It works to reduce the immunosuppressive factors within the TIME and promote a pro-immune response. PyroTIMETM creates a more favorable environment for immune cells to function effectively. Many tumors create an immunosuppressive environment that hinders immune cell activity. PyroTIMETM aims to counteract this by suppressing immunosuppressive molecules and promoting the recruitment and maturation of immune cells within the tumor. PyroTIMETM encodes various molecules, including cytokines, chemokines, soluble receptor antagonists, and gene-editing tools like guide RNAs (gRNA).
  • gRNA guide RNAs
  • the PyroVaxTM vector is a sophisticated tool for cancer immunotherapy. PyroHIMTM makes cancer cells more visible to the immune system, PyroActTM amplifies the immune response and recruits immune cells, and PyroTIMETM remodels the tumor microenvironment to create a more immune-supportive environment. Together, these components aim to overcome the challenges associated with cancer immunotherapy by harnessing the power of the patient's immune system to target and eliminate cancer cells effectively. 32 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • PyroHIMTM Molecular Selection
  • PyroHIMTM is designed to enhance the immunogenicity of the PyroCells vaccine. It serves as an adjuvant, stimulating a potent immune response against tumor cells.
  • SARS-CoV-2-derived molecules for PyroHIMTM is grounded in several scientific principles, including high immunogenicity, broad pre-existing immunity, epitope spreading potential, molecular selection criteria, the spike protein, safety profile, and strategic synergy.
  • SARS-CoV-2 has demonstrated an exceptional capacity to provoke robust immune responses in infected individuals.
  • PyroHIMTM acts as a beacon, drawing upon these pre- existing immune cells and redirecting them to target tumor cells, effectively repurposing the body's existing defenses for cancer immunotherapy.
  • SARS-CoV-2-derived molecules including the spike protein, contain multiple immunogenic epitopes. When presented to the immune system, these epitopes can trigger epitope spreading, a process where the immune response broadens to recognize and attack a variety of antigens. The present disclosure discusses methods to exploit this phenomenon to diversify the anti-tumor immune response. As PyroHIMTM directs immune cells towards tumor cells while incorporating SARS-CoV-2 epitopes, it encourages the immune system to identify and target a broader range of tumor-specific antigens.
  • SARS-CoV-2 The specific molecules chosen from SARS-CoV-2 for PyroHIMTM are selected based on their immunogenicity, safety, and strategic fit for cancer immunotherapy: [0185]
  • the spike protein of SARS-CoV-2 is a key antigenic target of the immune response during natural infection and vaccination. Its prominent role in eliciting strong immune reactions makes it an ideal choice for PyroHIMTM. Furthermore, the spike protein's widespread recognition by the immune system enhances the likelihood of recruiting memory T cells and antibodies to target tumor cells. 33 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • SARS-CoV-2 molecules have a well-characterized safety profile from extensive research during the COVID-19 pandemic. This safety record is crucial, ensuring that PyroHIMTM does not introduce unexpected adverse effects or immune reactions.
  • the choice of SARS-CoV-2 molecules aligns strategically with the disclosure's broader goals. By repurposing the immune response developed against SARS-CoV-2 for cancer treatment, the present disclosure describes methods and compounds to increase the utility of existing immunity, making PyroCells an innovative and efficient therapy.
  • PyroHIMTM 's scientific rationale for selecting specific molecules, particularly those related to SARS-CoV-2, is rooted in their proven immunogenicity, safety, and potential to induce epitope spreading. This unique approach leverages the global experience with SARS- CoV-2 to advance the field of cancer immunotherapy, offering a promising solution to address the unmet needs of cancer patients.
  • PyroHIM TM was designed using several strategies and with multiple pathways in mind, including selective immune cell recruitment, antigen presentation enhancement, T cell activation and expansion, epitope spreading, and inflammatory cell death (pyroptosis).PyroHIMTM is designed to attract and recruit specific immune cell populations to the tumor site.
  • SARS-CoV-2-derived epitopes which act as molecular "baits” to draw immune cells.
  • These epitopes have a twofold purpose: they serve as targets for pre-existing memory T cells generated against SARS- CoV-2, and they stimulate the maturation and activation of antigen-presenting cells (APCs) at the tumor site.
  • APCs antigen-presenting cells
  • APCs The presence of SARS-CoV-2 epitopes within PyroHIMTM triggers APCs to engulf tumor cells and present a diverse array of tumor antigens, including patient- specific neoantigens. This process, called cross-presentation, is critical for the activation of cytotoxic T cells that can target and eliminate tumor cells.
  • T cells Upon encountering the tumor-derived antigens presented by APCs, T cells are activated.
  • PyroHIMTM enhances this activation by providing a rich source of co-stimulatory signals and cytokines, such as GM-CSF (Granulocyte-Macrophage Colony-Stimulating Factor).
  • GM-CSF Granulocyte-Macrophage Colony-Stimulating Factor
  • GM-CSF further stimulates the maturation of dendritic cells and promotes the expansion and activation of effector T cells, specifically cytotoxic CD8+ T cells.
  • PyroActTM's incorporation of SARS-CoV-2 epitopes known to be highly immunogenic, induces epitope spreading. This phenomenon broadens the immune response 34 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO beyond the initially targeted tumor-specific antigens. As a result, the immune system becomes attuned to a wider spectrum of tumor antigens, reducing the risk of tumor immune escape due to antigenic variation.
  • PyroTIMETM Remodeling the Tumor Immune Microenvironment
  • PyroTIMETM is strategically designed to disrupt the immunosuppressive microenvironment that often surrounds tumors. It accomplishes this through the targeted release of cytokines and chemokines, such as IFN-$ (Interferon-gamma) and CXCL9/10. These signaling molecules attract immune cells, particularly cytotoxic T cells and natural killer (NK) cells, into the tumor microenvironment.
  • cytokines and chemokines such as IFN-$ (Interferon-gamma) and CXCL9/10. These signaling molecules attract immune cells, particularly cytotoxic T cells and natural killer (NK) cells, into the tumor microenvironment.
  • NK natural killer
  • PyroTIMETM doesn't merely attract immune cells; it also reprograms them to adopt a more aggressive anti-tumor phenotype. IFN-$, released by PyroTIMETM, enhances the cytotoxic activity of T cells and NK cells, making them more effective at recognizing and eliminating tumor cells.
  • PyroTIMETM complements the antigen presentation process initiated by PyroHIMTM. By creating an inflamed tumor microenvironment, it encourages APCs to be more efficient at engulfing tumor material, processing it, and presenting it to T cells. This synergy between PyroHIMTM and PyroTIMETM leads to a robust and sustained anti-tumor immune response. [0199] PyroTIMETM also exerts anti-angiogenic effects by inhibiting the recruitment of new blood vessels to the tumor site.
  • PyroHIMTM and PyroTIMETM work synergistically to enhance tumor immunogenicity and remodel the Tumor Immune Microenvironment (TIME).
  • PATENT Attorney Docket No. PYRO1100-3WO recruits and activates immune cells, induces epitope spreading, and amplifies the immune response, while PyroTIMETM disrupts the immunosuppressive TIME, reprograms immune cells for a more aggressive anti-tumor function, and enhances antigen presentation.
  • PyroHIMTM Interactions Between PyroHIMTM and PyroTIMETM
  • the present disclosure discusses use of various techniques and cells in order to enhance anti-tumor immune responses, including immune cell recruitment and activation, remodeling the tumor microenvironment, reducing immunosuppressive cells, increasing inflammatory cytokines, enhancing antigen presentation, and the synergistic effect of modification of multiple techniques and materials.
  • PyroHIMTM is designed to recruit and activate immune cells, including dendritic cells, T cells, and natural killer (NK) cells, within the Tumor Immune Microenvironment (TIME).
  • TIME Tumor Immune Microenvironment
  • PyroTIMETM complements PyroHIMTM by altering the Tumor Microenvironment (TME) to be more immunogenic. It can potentially reduce immunosuppressive factors and cells within the TME.
  • TME Tumor Microenvironment
  • This remodeling includes: [0205] PyroTIMETM may reduce the presence of regulatory T cells (Tregs) and myeloid- derived suppressor cells (MDSCs), which are known to suppress anti-tumor immune responses. This reduction helps create a more permissive immune environment.
  • PyroTIMETM can increase the production of pro-inflammatory cytokines (e.g., IL-2, IFN-$) within the TME.
  • pro-inflammatory cytokines e.g., IL-2, IFN-$
  • PyroTIMETM By promoting the maturation of dendritic cells and increasing the expression of major histocompatibility complex (MHC) molecules, PyroTIMETM improves antigen presentation. This allows for better recognition of tumor antigens by T cells.
  • MHC major histocompatibility complex
  • PyroHIMTM and PyroTIMETM act synergistically to create an immune-favorable TME. PyroHIMTM recruits immune cells, while PyroTIMETM helps create an environment that activates these recruited cells. This combined effect amplifies the anti-tumor immune response, potentially overcoming the immunosuppression typically seen in the TME.
  • PyroStimTM further boosts this activation through the inclusion of immune checkpoint inhibitors and immunostimulatory cytokines.
  • PyroHIMTM enhances antigen presentation by promoting the maturation of dendritic cells. PyroStimTM complements this by including tumor-specific antigens, ensuring a diverse array of tumor antigens are presented to immune cells. This combination enhances the immune system's recognition of tumor cells.
  • PyroStimTM contains immune checkpoint inhibitors, such as anti-PD-L1 antibodies, which block the PD-L1/PD-1 interaction. This prevents T cell exhaustion and enhances their cytotoxic activity.
  • the immune checkpoint blockade When used in conjunction with PyroHIMTM and PyroTIMETM, the immune checkpoint blockade is applied in an environment where immune cells are actively engaged, maximizing its effectiveness.
  • PyroHIMTM and PyroTIMETM potentially promote epitope spreading by creating conditions where dying tumor cells release a variety of tumor antigens. PyroStimTM enhances this process by boosting the immune response against these released antigens. This synergy can lead to a broader and more potent anti-tumor immune response.
  • All three components can be personalized based on the subject's tumor profile. This ensures that the treatment is tailored to the unique characteristics of the subject's cancer, maximizing its efficacy.
  • PyroHIMTM, PyroTIMETM, and PyroStimTM work in a coordinated manner to create a highly immunogenic and pro-inflammatory tumor microenvironment. They recruit immune cells, remodel the immune-suppressive elements within the tumor, activate the immune response, and enhance antigen presentation. This comprehensive approach addresses various aspects of the anti-tumor immune response, potentially leading to a more effective and durable therapeutic effect.
  • Technology of PyroCellsTM 37 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO
  • the present disclosure provides mechanisms, techniques and materials to redirect immune response, modify immunological memory, increase function of epitope cross- reactivity, activate immune cells, and modify supraphysiological expression.
  • Redirecting the immune response is one aspect of the present disclosure, initially generated against SARS-CoV-2, towards the targeted elimination of cancer cells. This redirection is achieved through the PyroCellTM immunotherapy system and the PyroVaxTM vector, which have been meticulously designed to exploit the immunological memory and response elicited by SARS-CoV-2 exposure or vaccination. This section elaborates on the mechanism, rationale, and preliminary evidence supporting this redirection. [0220] The immune system's memory is a key component of this redirection.
  • the PyroVaxTM vector incorporates highly immunogenic epitopes from SARS-CoV- 2 antigens, such as the spike glycoprotein (S), envelope protein (E), membrane protein (M), nucleocapsid phosphoprotein (N), as well as other immunodominant antigens. These epitopes have been strategically selected based on their propensity for cross-reactivity with cancer- specific antigens.
  • PyroVaxTM includes PyroHIMsTM, which encode highly immunogenic molecules. These molecules serve as potent activation signals for immune cells, triggering their effector functions, including cytokine release and cytotoxic activity. By linking PyroHIMsTM to SARS- CoV-2-derived epitopes, PyroVaxTM ensures the activation of memory immune cells with specificity for SARS-CoV-2 antigens. [0223] In some embodiments, PyroHIMsTM are linked to reporter molecules, enabling supraphysiological expression, detection, and enrichment of PyroCellsTM expressing the PyroVaxTM vector.
  • PyroCells are designed to stimulate a robust immune response against cancer cells. T cells, a critical component of the immune system, play a central role in this process. Upon activation, cytotoxic T cells, in particular, release molecules such as granzyme B.
  • Granzyme B is a protease enzyme released by cytotoxic T cells. It is a key mediator of the immune response against infected or cancerous cells. Granzyme B can enter target cells, including cancer cells. [0228] Inside the cancer cells targeted by PyroCells, granzyme B initiates a cascade of events that ultimately lead to the activation of gasdermin E. Gasdermin E is a critical protein in pyroptosis; it forms pores in the cell membrane. [0229] Gasdermin E pores created in the cell membrane disrupt its integrity. This results in the release of cellular contents, including pro-inflammatory cytokines and danger-associated molecular patterns (DAMPs).
  • DAMPs danger-associated molecular patterns
  • the release of pro-inflammatory cytokines and DAMPs signals neighboring immune cells, particularly macrophages and dendritic cells, to mount an immune response. This response includes the recruitment of more immune cells to the site and the activation of an adaptive immune response against the cancer cells.
  • the disruption of the cell membrane integrity by gasdermin E pores leads to cell lysis, or cell bursting. These releases additional pro-inflammatory molecules and cellular debris.
  • the pyroptotic mechanism is a powerful tool in cancer immunotherapy, there are potential challenges and side effects to consider: The hallmark of pyroptosis is the rapid and robust inflammatory response.
  • PYRO1100-3WO PYRO1100-3WO
  • PyroCells are personalized for each patient, targeting their specific cancer cells. This approach minimizes off-target effects, as the vaccine is tailored to the individual's unique cancer profile. Careful dose optimization is performed to balance a strong immune response with safety. This involves determining the appropriate amount of PyroCells to minimize excessive inflammation while ensuring efficacy. Rigorous monitoring of patients during clinical trials allows for the early detection of adverse effects, enabling timely intervention and adjustment of treatment plans.
  • the present disclosure explores combination therapies, including immunosuppressive agents or anti-inflammatory drugs, to modulate the immune response and mitigate potential side effects.
  • the pyroptotic mechanism involving T cell-secreted granzyme B and gasdermin E is one aspect of PyroCells' therapeutic action against cancer. While there are challenges and potential side effects associated with this mechanism, personalized treatment, dose optimization, vigilant monitoring, and strategic combination therapies help to mitigate these risks. These efforts aim to increase the benefits of pyroptosis while ensuring patient safety and treatment effectiveness.
  • Allogeneic Mechanism of Action [0236] The mechanism of action of the proposed therapy, which involves the use of allogenic cells, includes a complex but highly orchestrated process that harnesses the power of the immune system to target and eliminate cancer cells.
  • Enhanced Antigen Presentation PyroCellsTM are designed to enhance antigen presentation within the tumor microenvironment.
  • MHC Class I Molecules PyroCellsTM increase the expression of Major Histocompatibility Complex (MHC) Class I molecules on the surface of tumor cells. MHC Class I molecules play a critical role in presenting tumor antigens to CD8+ T cells.
  • Activation of Dendritic Cells PyroCellsTM stimulate the maturation and activation of dendritic cells within the tumor microenvironment. Dendritic cells are professional antigen- presenting cells that capture, process, and present tumor antigens to CD8+ T cells, initiating an immune response. 40 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • CD8+ T Cell Activation The CD8+ T cells recruited to the tumor site encounter the tumor-specific antigens presented by MHC Class I molecules on PyroCellsTM and dendritic cells. This interaction activates the CD8+ T cells, turning them into cytotoxic effector T cells.
  • Tumor Cell Killing Activated CD8+ T cells recognize and specifically target tumor cells expressing the presented antigens. They release cytotoxic molecules, such as perforin and granzymes, to induce apoptosis (cell death) in the tumor cells.
  • Epitope Spreading As tumor cells are killed, they release additional tumor antigens into the tumor microenvironment.
  • PyroStimTM Composition, Mechanism of Action, Advantages
  • PyroStimTM is a carefully composed therapeutic composition with the potential to significantly enhance the anti-tumor immune response. Its multi-faceted mechanism of action, including immune checkpoint blockade, cytokine-mediated enhancement, and antigen presentation, makes it a promising addition to Pyrojas' arsenal in the fight against cancer.
  • composition PyroStimTM is a proprietary composition designed to enhance the effectiveness of Pyrojas' cancer immunotherapy. It is composed of a carefully selected combination of molecules and factors that collectively boost the anti-tumor immune response.
  • the composition includes: [0246] Immune Checkpoint Inhibitors: PyroStimTM incorporates immune checkpoint inhibitors (ICIs) such as anti-PD-L1 (Programmed Death-Ligand 1) antibodies. These ICIs block the PD-L1/PD-1 interaction, preventing the tumor from evading the immune system by suppressing T cell activity.
  • ICIs immune checkpoint inhibitors
  • Cytokines PyroStimTM contains specific cytokines, including interleukins (e.g., IL- 2 and IL-12), interferons (e.g., IFN-$), and TNF alpha family memebers known for their roles in enhancing immune responses. These cytokines stimulate the activation and proliferation of cytotoxic T cells, natural killer (NK) cells, and other immune effector cells.
  • Mechanism of Action PyroStimTM operates through a multi-faceted mechanism to potentiate the immune response against the tumor:
  • Immune Checkpoint Blockade The inclusion of immune checkpoint inhibitors in PyroStimTM, such as anti-PD-L1 antibodies, plays a central role.
  • PyroStimTM By blocking the PD-L1/PD-1 41 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO interaction, PyroStimTM prevents the inhibition of T cell activity, allowing activated T cells to target and eliminate tumor cells more effectively.
  • Cytokine-Mediated Enhancement Cytokines within PyroStimTM, such as IL-2, IL- 12, and IFN-$, act as potent immunostimulants. They activate immune effector cells, particularly cytotoxic T cells and NK cells, which are critical for tumor cell recognition and destruction.
  • Antigen Presentation Inclusion of tumor-specific antigens in PyroStimTM enhances antigen presentation.
  • PyroStimTM synergizes with PyroHIMTM and PyroTIMETM, which recruit and activate immune cells and remodel the Tumor Immune Microenvironment (TIME). The combined effect amplifies the anti-tumor response, potentially overcoming immunosuppression within the tumor and promoting durable tumor control.
  • TIME Tumor Immune Microenvironment
  • PyroStimTM is designed to work synergistically with Pyrojas' other therapeutic components, including PyroHIMTM and PyroTIMETM. This combination approach increases the chances of a robust and sustained anti-tumor response.
  • PYRO1100-3WO Challenges and Optimizations Ensuring the purity of the target cell line is crucial. Regular STR profiling and mycoplasma testing are conducted to maintain cell line integrity. Strict aseptic procedures are followed to prevent contamination during cell culture. [0261] Transfection with PyroVaxTM Vector (DNA or mRNA) [0262] Techniques and Equipment: Transfection Reagents: ipofection or electroporation based on cell type. Commercial transfection reagents or custom-made formulations are employed. PyroVaxTM Vector Preparation: The PyroVaxTM vector is purified and quantified using established molecular biology techniques.
  • Transfection Efficiency Optimization of transfection conditions (e.g., voltage, reagent concentration) is crucial to increase vector uptake by target cells.
  • Vector Quality Control Rigorous quality control of the PyroVaxTM vector ensures consistent results.
  • Selection and Expansion of Transfected Cells [0264] Techniques and Equipment: Selection Markers: Cells are often transfected with a selectable marker gene, e.g., antibiotic resistance or fluorescent proteins. Cell Expansion: Transfected cells are expanded in culture to obtain enough for downstream applications.
  • Selection Pressure Optimizing the concentration of selection agents is necessary to balance cell survival and marker expression.
  • Cell Growth Optimization Media composition, seeding density, and passage intervals are modified to promote cell growth while maintaining transgene expression.
  • PyroCellTM Production Techniques and Equipment: Cell Harvest: Transfected cells are harvested at the desired stage of growth. Quality Control: Cell viability, transgene expression, and other relevant parameters are assessed. Potential Challenges and Optimizations: Cell Viability: Careful timing of cell harvest is critical to increase cell viability. Consistency: Batch-to-batch consistency is maintained through rigorous quality control measures.
  • Cryopreservation Techniques and Equipment Cryoprotectants: Cells are cryopreserved using validated cryoprotectant solutions.
  • Cryopreservation Containers Vials or cryobags are used for storage.
  • Tumor cells are enriched by removing stromal cells, including immune cells, endothelial cells, and fibroblasts, using antibody-mediated negative selection.
  • the PyroVaxTM composed of either DNA vector or mRNA construct encoding the components of PyroHIMTM, PyroActTM, and PyroTIMETM, is introduced into tumor cells using viral transduction or electroporation.
  • PyroVaxTM expressing cells termed PyroCellsTM, are chemically enriched and treated with PyroStimTM, a cocktail of cytokines and chemokines which increase tumor antigen presentation, and the gene expression levels of ISGs, interferon-stimulated genes.
  • GMP compliant immunotherapeutic tumor cells are expanded to achieve therapeutic doses (0.1, 0.5, 1 x 10 ⁇ 6), rendered replication incompetent, frozen into individual doses, and shipped to the physician for the treatment of the patient.
  • the entire manufacturing process is GMP compliant and is completed within two weeks.
  • Allo-PyroCellTM Briefly, patient-derived autologous tumor cells are chemically modified using PyroStimTM and mixed with allo-PyroCellTM, an allogeneic cell line modified with either a DNA vector or mRNA construct encoding the components of PyroVaxTM, which includes sequences of PyroHIMTM, PyroActTM, and PyroTIMETM (at either of the ratios: 1:1, 1:2, 1:5, 1:10, 1:100). This mixture is then shipped to the physician for the treatment of the patient. The entire manufacturing process is GMP compliant and is completed within two weeks.
  • allogeneic cellular therapies have abundantly available TAAs, are standardizable, scalable, less variable, and cost-effective.
  • Cell lines can be developed from solid or liquid cancers, metastatic or circulating cancer cells, and can be derived from one or multiple individuals.
  • PyroVir TM To manufacture PyroVir TM to deliver PyroVax TM intratumorally.
  • the present disclosure describes the composition of and methods to generate PyroVir TM , an AAV, HSV1/2, or other cancer specific virus, expressing the PyroVax TM vector. When administered intratumorally, the virus infects tumor cells and stimulate an immune response against PyroHIMs TM and a broad repertoire of cancer specific antigens.
  • PyroVir TM overcomes the various challenges associated with ex vivo modifications of autologous or allogeneic cells.
  • PyroVax TM components the PyroHIMs TM , PyroActs TM , and PyroTIME TM are quantified using RT-PCR, and immunoblotting or flow cytometry. Efficient delivery of PyroVax TM into normal or transformed cells is confirmed using immunoblotting, flow cytometry, and ELISA based studies. [0275] PyroCells TM cellular and PyroVir TM viral immunotherapies induce pyroptotic tumor cell death. [0276] Next, the impact of the expression of PyroVax TM in normal and transformed human and murine cells is functionally validated.
  • the PyroAct TM molecule serves as a trigger for the inflammatory cell death of PyroCells TM when killed by PyroHIM TM antigen-specific T cells.
  • PyroCells TM and PyroAct TM deficient PyroCells TM are killed by PyroHIM TM antigen specific-CTLs, although with noteworthy differences.
  • PyroAct TM deficient PyroCells TM die predominantly via Caspase-1 mediated apoptosis
  • PyroAct TM expressing PyroCells TM die preferentially via pyroptosis.
  • LDH enzyme release is used to monitor pyroptosis (7), among other markers including cytokine released because of inflammasome activation: IL-1# and IL-1RA. Activation of the inflammasome induces the formation of Gasdermin-D pores on the cell membrane, causing IL-1# and IL-18 secretion, and the influx of water molecules leading to cell swelling and subsequent rupture (pyroptosis).
  • SARS-Cov-2 antigens themselves are shown to have a significant intrinsic potential to activate the inflammasome and pyroptosis.
  • PyroHIMs TM encoding the SARS-CoV-2 proteins E, M, and specific ORFs induced efflux of K + ions creating an imbalance which results in oxidative stress, damage to the mitochondria, and activation of the NLRP3 inflammasome (8, 9).
  • ORF3a and ORF8b activate the inflammasome directly (8, 9).
  • Immunization and validation of mice against anti-SARS-Cov-2 [0283] Construction, expression, and purification of recombinant spike protein.
  • the code optimized recombinant DNA encoding the Spike protein expressed by the SARS-Cov-2 virus was introduced into a mammalian gene expression lentiviral vector (pLV[Exp]-Puro-CMV) vector (Vector Builder, USA). The DNA sequence of the assembled vector was verified by DNA sequencing. The sequence of the inserts and vectors are provided.
  • pLV[Exp]-Puro-CMV mammalian gene expression lentiviral vector
  • Immunized mice are intranasally challenged with mouse-adapted strain at passage 6, MASCp6 (1.6 ⁇ 10 4 PFU), and lung tissues are collected for virological and histopathological analysis at 5 days after challenge, as described previously (10). All the PBS-treated mice sustain high amounts of viral RNA loads in the lung at 5 days after challenge. By contrast, immunized mice demonstrate a significant reduction in viral RNA loads. Immunofluorescence staining for 46 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO SARS-CoV-2 S protein is used to detect viral proteins in the lungs of immunized mice versus PBS-immunized mice.
  • ELISA ELISA is performed to detect SARS-CoV-2-specific IgG antibodies. Briefly, ELISA plates are precoated with SARS-CoV-2 Spike protein (1 %g/ml) overnight at 4' and blocked with 2% milk in PBST for 2 h at 37'. Serially diluted sera are added to the plates and incubated for 2 h at 37'.
  • HRP horseradish peroxidase
  • TMB Tetramethylbenzidine
  • SARS-CoV-2 neutralization assay A micro-neutralization assay is carried out to detect neutralizing antibodies against SARS-CoV-2 as previously described (11, 12).
  • mice sera at 2-fold serial dilutions are incubated with SARS-CoV-2 (100 TCID50) for 1 h at 37' and added to Vero cells.
  • the cells are observed daily for the presence or absence of virus- induced cytopathic 9 effects (CPE) and recorded at 3 dpi.
  • CPE virus- induced cytopathic 9 effects
  • Neutralizing antibody titers are expressed as the reciprocal of the highest dilution of serum that completely prevent CPE in Vero cells.
  • PyroCells TM and PyroVir TM viral immunotherapies are safe and tolerable.
  • mice immunized against SARS-Cov-2 are treated with the PyroCells TM cellular or PyroVirTM viral immunotherapy and monitored for acute safety concerns and long-term adverse outcomes. Mice maintain their weight and show no signs of morbidity throughout the course of the treatment and on long-term follow up. Gross and histological assessment of all major organs systems by a pathologist is performed to evaluate any signs of toxicity or unintended adverse outcomes including auto-immune reactions. The impact of combining the therapeutic vaccines with the current standard of care (chemotherapy) or with immune-checkpoint inhibitors is also be tested. [0289] PyroCells TM cellular and PyroVir TM viral immunotherapies control disease burden and improve survival outcomes.
  • the anti-tumor efficacy of the immunotherapy is determined using murine models of solid (TNBC, melanoma, ovarian cancer) and hematological (leukemias, 47 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO lymphomas, and myelomas) human cancers.
  • TNBC solid-derived mammal-derived melanoma, ovarian cancer
  • hematological leukemias, 47 ACTIVE ⁇ 1604904070.2
  • PATENT Attorney Docket No. PYRO1100-3WO lymphomas, and myelomas human cancers.
  • the MTD determined previously is utilized to test the anti-tumor efficacy of PyroCells TM immunotherapies in vivo.
  • Treatment of mice bearing established disease with PyroCells TM immunotherapy alone, or in combination with immune checkpoint inhibitors control the disease burden and increase the overall survival of mice.
  • ELISPOT and tetramer-based flow cytometry are used to 48 ACTIVE ⁇ 1604904070.2
  • PATENT Attorney Docket No. PYRO1100-3WO confirm the presence of PyroHIM TM antigen specific CD4 + and CD8 + T cells, SARS-Cov-2 specific T cells, and notably several novel TAA-specific CD8 + and CD4 + T cells in mice.
  • Paired scRNA-seq and TCRa/b sequencing are used to test for clonal expansions of polyvalent T cells against a broad repertoire of cancer specific antigens besides the immunodominant PyroHIM TM epitope.
  • mice develop cross-primed T cells to known cancer antigens and importantly to newly arising TAAs, tumor-associated antigens.
  • Transcriptomics analysis of tumors is used to demonstrate a significant decrease in the expression of target antigens and/or TAAs in tumors derived from mice that received the PyroCells TM immunotherapy as compared to control counterparts. No adverse auto-immune reactions occur in humans or mice.
  • CTLs retrieved from the spleens of mice that received the vaccination or not are compared to previously unreactive cancer specific antigens using T cell and tumor cell co-culture killing assays and ELISPOT cytokine release assay.
  • CTLs derived from mice treated with PyroCells TM kill the parental PyroVax TM deficient tumor cells but CTLs derived from PyroAct TM deficient PyroCells TM have no reactivity towards parental cells.
  • mice treated with PyroCells TM develop antigen specific T cells which recognize not only the immunodominant PyroHIM TM antigen, but notably various other tumor-associated antigens.
  • Treatment of mice bearing established disease with PyroCells TM immunotherapy alone, or in combination with immune checkpoint inhibitors control the disease burden and increase the overall survival of mice.
  • re-challenge of complete responders with parental tumor cells not containing the epitope results in complete rejection of the parental tumor highlighting the diversification or spreading of the epitope and the presence of an immunological memory response.
  • the allo- PyroCells TM immunotherapies inhibit the emergence and establishment of disease burden by parent tumor cells not expressing PyroHIMs TM .
  • Immune evasion by tumor cells is driven by a loss of antigen presentation molecules like MHC-I or B2M. Therefore, the impact of vaccines on tumors which are predominantly B2M or MHC-I deficient is evaluated in vivo.
  • PyroCells TM render MHC-I or B2M deficient cancer cells sensitive to killing by CD8 + T cells whereas PyroAct TM extend the sensitivity to diverse additional tumor epitopes in vivo.
  • PyroTherapies TM redirect the anti-SARS-Cov-2 immune response against uterine leiomyomas [0301]
  • the safety and efficacy of using PyroTherapiesTM for the treatment of benign tumors, including uterine leiomyomas or fibroids, termed FibroTherapy TM is tested.
  • the immunocompetent Eker rat model of spontaneous uterine fibroids and an immunodeficient murine model of human uterine fibroid xenografts are used.
  • immunocompetent Eker rat with spontaneous uterine fibroids or immunocompromised Balb/c mice with transplanted human patient derived uterine fibroid xenografts are prophylactically vaccinated against an immunogen.
  • the M-spike protein expressed by the SARS-Cov-2 virus is used as a model immunogen derived from PyroHIMs TM , others could be used as well. All rats develop a robust humoral and adaptive immune response against the immunizing epitope several weeks after immunization.
  • a luciferase tagged episomal expression vector (EEV) or viral vector containing the DNA encoding the immunogen used for prophylactic immunization is directly injected into the fibroids.
  • EAV episomal expression vector
  • intra-fibroid injection is performed laparoscopically under MRI-guidance, whereas for the human PDX model direct injection is used because the transplant is accessible.
  • Bioluminescent imaging is used to confirm fibroid specific expression of the DNA vector in both models.
  • antigen specific cell therapy either TCR-T cells, or CAR-T/NK cells targeting the immunogenic antigen is administered.
  • FIGURE 7B A comprehensive quantification of this data (FIGURE 7B) emphasized the efficiency of the lentiviral transduction process, s featuring significant protein expression in the transduced cells as compared to control cells. Support: Demonstrating efficient transduction and protein expression in human TNBC cells reinforces the feasibility of the proposed approach. Successful expression in a challenging cell line like TNBC vouches for the efficacy and adaptability of the proposed constructs and transduction methods. [0307] Non-viral Approach for Protein Expression: Exploring alternative methodologies, a non-viral approach was also employed to achieve protein expression in breast cancer cells. Contour plots representing this data (FIGURE 7C) displayed comparable expression levels to the lentiviral method, suggesting the potential versatility of constructs for diverse delivery mechanisms.
  • mice bearing specific tumor burdens were imaged at indicated time intervals (FIGURE 8A). After a span of 21 days, a subset of these mice underwent a rechallenge with the parental tumor. Interestingly, the data (FIGURE 8B) showcased a substantial protective effect in mice previously immunized with the vaccine, indicating its potential not only in 51 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • the results provide a comprehensive account of the in vitro and in vivo efficacy of the therapeutic constructs. From effective protein expression in human TNBC cells to significant reduction in tumor burden in murine models, the findings pave the way for potential clinical applications of these constructs in cancer therapy. Support: The extended time-course study further endorses the long-term therapeutic potential of the constructs. Showing control over cancer growth and recurrence prevention in an extended setting makes a compelling case for this application, as it promises long-term patient benefits.
  • the CD3+ T lymphocytes were enriched, activated using CD3/CD28 Dynabeads, and expanded in IL-2 media. Following transduction with the PyroTIMER construct and a second-generation anti-human CD19 CAR, single- and double-positive cells were selected and enriched using chemical selection and flow-based cell sorting to achieve stable expression of both the PyroTIMER and the CAR over time (FIGURE 9C, FIGURE 10C and FIGURE 10F). Proper controls, including non-transfected cells and cells transfected with a non-functional version of PyroTIMER, are utilized to rule out non-specific effects.
  • FIGURE 10G demonstrates the superior cytotoxic prowess of PyroTIMER CD19 CAR-T cells
  • FIGURE 10H accentuates the heightened activation, evident from the surge in CD69+ activated T cells.
  • Predominance in TGF-" Rich Environments The data underscores the PyroTIMER CAR-T cells' unparalleled performance, particularly in TGF-# rich milieus.
  • the co-culture killing assays with CD19-expressing Raji lymphoma cells, presented in FIGURE 9D, FIGURE 9E, FIGURE 10E, and FIGURE 10F, were executed in the presence of 100 53 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • PYRO1100-3WO pM TGF-#1 replicating the immunosuppressive ambience of tumors overflowing with TGF- #.
  • the superior killing efficacy of PyroTIMER CD19 CAR-T cells in comparison to traditional CD19 CAR-T cells in this environment offers incontrovertible evidence of their enhanced functionality in settings abundant in TGF-#.
  • PyroTIMER technology improves the cytotoxicity of CD19 CAR-T cells against CD19-expressing cells in TGF-f rich microenvironments.
  • inhibiting the TGF-f pathway in hematological malignancies using CAR-T cells is particularly crucial since TGF-# plays a critical role in creating an immunosuppressive TIME in CD19 malignancies.
  • Uterine Leiomyomas (Fibroids) as a Model: [0325] 1. Prevalence and Clinical Significance: Uterine leiomyomas, commonly referred to as fibroids, are noncancerous growths of the uterus that often occur in women of reproductive age. They are highly prevalent, with estimates suggesting that up to 70-80% of women may develop fibroids during their lifetime. Fibroids can vary in size, number, and location within the uterus, leading to a range of clinical symptoms, including heavy menstrual bleeding, pelvic pain, and reproductive issues. Due to their prevalence and clinical significance, fibroids represent a substantial burden on women's health and healthcare systems.
  • FibroTherapyTM was initially developed for uterine leiomyomas (fibroids), its underlying principles and mechanisms of action hold promise for potential extensions to other benign tumors. Here are some potential implications and extensions: [0330] Tissue-Specific Targeting: The key to FibroTherapyTM is its ability to selectively target and remodel fibrotic tissue within the uterus. This tissue-specific targeting is achieved through the use of PyroCellsTM, which are genetically engineered to express specific molecules that interact with fibrotic tissue. This targeting strategy could potentially be adapted to other benign tumors that involve fibrosis or excessive extracellular matrix deposition.
  • FibroTherapyTM demonstrates the potential for personalized medicine in the treatment of benign tumors. By engineering PyroCellsTM to express molecules tailored to the specific characteristics of the target tissue, it's possible to customize the therapy for different benign tumor types. This approach allows for a high degree of specificity and precision in treatment.
  • Minimally Invasive Nature FibroTherapyTM is designed to be delivered through minimally invasive techniques, such as laparoscopy or hysteroscopy. This minimizes the need for open surgery and reduces the associated risks and recovery times.
  • FibroTherapyTM relies on the targeted action of PyroCellsTM within the tumor microenvironment, it may have a favorable safety profile 55 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO compared to more systemic treatments. This aspect could be explored in the context of other benign tumors, where minimizing off-target effects is crucial.
  • Clinical Research To explore the extension of FibroTherapyTM to other benign tumors, further research and clinical trials would be necessary.
  • FibroTherapyTM was initially developed for uterine leiomyomas (fibroids), its tissue-specific targeting and minimally invasive nature open possibilities for potential extensions to other benign tumors characterized by fibrosis or similar pathological features. These extensions could offer new treatment options for subjects with a range of benign tumor conditions, addressing unmet medical needs in multiple clinical settings.
  • the present disclosure provides compositions and methods for the treatment of cancer.
  • the cancer includes a form of acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), adrenocortical carcinoma, aids-related cancer, Kaposi sarcoma, AIDS-related lymphoma, primary CNS lymphoma, anal cancer, astrocytoma, atypical teratoid/rhabdoid tumor, central nervous system cancer, basal cell carcinoma of the skin, skin cancer, bile duct cancer, bladder cancer, bone cancer (includes Ewing sarcoma, osteosarcoma and malignant fibrous histiocytoma), brain tumor, breast cancer, bronchial tumors, non-Hodgkin lymphoma (including Burkitt lymphoma), carcinoid tumor, carcinoma of unknown primary origin, cardiac tumor, medulloblastoma and other CNS embryonal tumor, germ cell tumor, cervical cancer, childhood cancer, bile duct cancer (including cholangiocarcinoma), chordoma
  • ALL
  • PYRO1100-3WO cell Hodgkin lymphoma, hypopharyngeal cancer, intraocular melanoma, islet cell tumors, pancreatic neuroendocrine tumors, kidney (renal cell) cancer, Langerhans cell histiocytosis, laryngeal cancer, head and neck cancer, leukemia, lip and oral cavity cancer, liver cancer, lung cancer (including non-small cell, small cell, pleuropulmonary blastoma, and/or tracheobronchial tumor), lymphoma, male breast cancer, melanoma, intraocular melanoma, Merkel cell carcinoma, malignant mesothelioma, metastatic cancer, metastatic squamous neck cancer with occult primary, midline tract carcinoma with nut gene changes, mouth cancer, multiple endocrine neoplasia syndromes, multiple myeloma/plasma cell neoplasms, mycosis fungoides, myelodysplastic syndromes, myel
  • methods for treating, improving, achieving remission of, and/or reducing the risk of a cancer disclosed herein involves administering to a subject an immunotherapeutic composition in accordance with the present disclosure.
  • the immunotherapeutic composition may be administered as a pharmaceutical composition according to any appropriate route and regimen.
  • the route or regimen is one that correlates with a positive therapeutic benefit.
  • 57 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO [0338]
  • the exact amount immunotherapeutic composition administered may vary from patient to patient, depending on one or more factors as is known in the medical arts.
  • Such factors may include, but are not limited to, one or more factors such as species, age, general condition of the patient, the particular composition to be administered, its mode of administration, its mode of activity, the severity of disease, the activity of the specific immunotherapeutic composition employed, the specific pharmaceutical composition administered, the half-life of the composition after administration, body weight, sex, diet of the patient, time of administration, route of administration, rate of excretion of the specific immunotherapeutic composition employed, duration of the treatment, and any other therapeutic agents used in combination or coincidental with the specific immunotherapeutic composition to be administered.
  • factors such as species, age, general condition of the patient, the particular composition to be administered, its mode of administration, its mode of activity, the severity of disease, the activity of the specific immunotherapeutic composition employed, the specific pharmaceutical composition administered, the half-life of the composition after administration, body weight, sex, diet of the patient, time of administration, route of administration, rate of excretion of the specific immunotherapeutic composition employed, duration of the treatment, and any other therapeutic agents used in combination or coincidental with
  • compositions of the present disclosure are administered by oral (PO), intravenous (IV), intramuscular (IM), intra-arterial, intramedullary, intrathecal, subcutaneous (SQ), intraventricular, transdermal, interdermal, intradermal, rectal (PR), vaginal, intraperitoneal (IP), intragastric (IG), topical (e.g., by powders, ointments, creams, gels, lotions, and/or drops), mucosal, intranasal, buccal, enteral, intravitreal, sublingual, by intratracheal instillation, bronchial instillation, and/or inhalation, as an oral spray, nasal spray, aerosol, and/or through a portal vein catheter.
  • oral oral
  • IV intravenous
  • IM intramuscular
  • IM intra-arterial
  • intramedullary intrathecal
  • SQ subcutaneous
  • IP intraperitoneal
  • IG intragastric
  • topical e.g., by powders
  • an immunotherapeutic composition in accordance with the present disclosure and/or pharmaceutical compositions thereof may be administered intravenously, for example, by intravenous infusion.
  • an immunotherapeutic composition in accordance with the present disclosure and/or pharmaceutical compositions thereof may be administered by intramuscular injection.
  • an immunotherapeutic composition in accordance with the present disclosure and/or pharmaceutical compositions thereof may be administered by intratumoral injection.
  • an immunotherapeutic composition in accordance with the present disclosure and/or pharmaceutical compositions thereof may be administered by subcutaneous injection.
  • an immunotherapeutic composition in accordance with the present disclosure and/or pharmaceutical compositions thereof may be administered via portal vein catheter.
  • the disclosure encompasses the delivery of an immunotherapeutic composition in accordance with the present disclosure and/or 58 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No. PYRO1100-3WO pharmaceutical compositions thereof by any appropriate route taking into consideration likely advances in the art of drug delivery.
  • the desired dosage may be delivered only once.
  • the desired dosage may be delivered once per day, more than once per day, once every other day, once every third day, once every week, once every two weeks, once every three weeks, once every four weeks, once every two months, once every six months, or once every twelve months.
  • the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations). In certain aspects, the desired dosage may be delivered using one or more administrations during an initial period of time, followed by a period of time in which no dosage is administered.
  • an immunotherapeutic composition in accordance with the present disclosure may be utilized for prophylactic applications. In more aspects, prophylactic applications involve systems and methods for inhibiting progression of, and/or delaying the onset of a cancer in individuals susceptible to and/or displaying symptoms of the cancer.
  • an immunotherapeutic composition in accordance with the present disclosure is administered to a target cell in vivo.
  • an immunotherapeutic composition in accordance with the present disclosure is administered to a target cell ex vivo.
  • an immunotherapeutic composition in accordance with the present disclosure is administered to a target cell ex vivo, then the target cell is re-introduced into an organism.
  • the target cell is cultured into multiple progeny cells ex vivo before being re-introduced in an organism.
  • the organism is a human.
  • the target cell was originally derived from the organism to which it is re-introduced.
  • an immunotherapeutic composition in accordance with the present disclosure and and/or pharmaceutical compositions thereof are employed in combination therapies for treating or reducing the risk of a cancer.
  • administration can be in combination with one or more additional therapeutic agents.
  • the phrases “combination therapy,” “combined with,” “in combination,” and the like refer to the use of more than one medication or treatment simultaneously to increase the response.
  • an immunotherapeutic composition in accordance with the present disclosure and and/or pharmaceutical compositions thereof are administered concurrently with, prior to, or 59 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • an immunotherapeutic composition in accordance with the present disclosure and/or pharmaceutical compositions thereof are administered in combination together in a single composition or administered separately in different compositions.
  • the particular combination of therapies to employ in a combination regimen generally take into account compatibility of the desired therapeutics and/or procedures, and the desired therapeutic effect to be achieved.
  • the therapies employed may achieve a desired effect for the same purpose (e.g., an immunotherapeutic composition in accordance with the present disclosure which is useful for treating, and/or delaying the onset of a cancer may be administered concurrently with another therapeutic agent which is also useful for treating, and/or delaying the onset of the cancer), or they may achieve different effects.
  • the combination of therapies employed may achieve the same or a substantially similar desired effect for the same cancer; may achieve the same or a substantially similar desired effect for one or more different cancers; may achieve different desired effects for the same cancer; or may achieve different desired effects for one or more different cancers.
  • combination therapy may involve administrations of a plurality of immunotherapeutic compositions in accordance with the present disclosure.
  • combination therapy may involve administrations of a plurality of immunotherapeutic compositions that treat, improve, achieve remission of, and/or reduce the risk of a single type of cancer.
  • combination therapy can be a plurality of immunotherapeutic compositions that treat, improve, achieve remission of, and/or reduce the risk of multiple types of cancers.
  • an immunotherapeutic composition in accordance with the present disclosure is combined with at least one pharmaceutically acceptable excipient, in the form of a pharmaceutical composition.
  • pharmaceutical composition refers to a formulation containing an active ingredient, and optionally a pharmaceutically acceptable carrier, diluent or excipient.
  • active ingredient can interchangeably refer to an 60 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • active ingredient include, but are not limited to, chemical compound, drug, therapeutic agent, small molecule, and the like.
  • the active ingredient is an immunotherapeutic composition as disclosed herein.
  • pharmaceutically acceptable it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof, nor to the activity of the active ingredient of the formulation.
  • Pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology.
  • compositions for example as described J.P. Remington & A. Osol, Remington's Pharmaceutical Sciences, 16th edition (1980) or J.P. Remington & P. Beringer, Remington: The Science and Practice of Pharmacy, 21st Edition (2006), both of which are herein incorporated by reference with respect to the pharmaceutically acceptable carriers, excipients and stabilizers provided therein.
  • pharmaceutically acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and may include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol); low molecular weight peptides (less than about 10 amino acid residues); proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine,
  • carrier examples include, but are not limited to, liposome, nanoparticles, ointment, micelles, microsphere, microparticle, cream, emulsion, and gel.
  • excipients include, but are not limited to, anti-adherents such as magnesium stearate, binders such as saccharides and their derivatives (sucrose, lactose, starches, cellulose, sugar alcohols and the like) protein like gelatin and 61 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • PYRO1100-3WO synthetic polymers such as talc and silica, and preservatives such as antioxidants, vitamin A, vitamin E, vitamin C, retinyl palmitate, selenium, cysteine, methionine, citric acid, sodium sulfate and parabens.
  • diluents include, but are not limited to, water, alcohol, saline solution, glycol, mineral oil and dimethyl sulfoxide (DMSO).
  • the pharmaceutical compositions include one or more additional therapeutically or biologically active substances.
  • the pharmaceutical compositions are useful in medicine or the manufacture of medicaments.
  • the pharmaceutical compositions are useful in one or more of the therapeutic applications disclosed herein, for example, in an individual suffering from an autoimmune disorder.
  • the pharmaceutical compositions are formulated for administration to a human subject.
  • the pharmaceutical composition is in a sterile injectable form (e.g., a form that is suitable for subcutaneous injection or intravenous infusion).
  • the pharmaceutical composition is in a liquid dosage form that is suitable for injection.
  • the pharmaceutical composition is in a powder (e.g., lyophilized and/or sterilized), optionally under vacuum, which is reconstituted with an aqueous diluent (e.g., water; buffer; salt solution, and the like) prior to injection.
  • an aqueous diluent e.g., water; buffer; salt solution, and the like
  • the pharmaceutical composition is diluted and/or reconstituted in an aqueous diluent (e.g., water, sodium chloride solution, sodium acetate solution, benzyl alcohol solution, phosphate buffered saline, and the like).
  • an aqueous diluent e.g., water, sodium chloride solution, sodium acetate solution, benzyl alcohol solution, phosphate buffered saline, and the like.
  • the pharmaceutical composition is in a form that can be refrigerated and/or frozen.
  • the pharmaceutical composition is in a form that cannot be refrigerated and/or frozen.
  • the pharmaceutical composition is a reconstituted solution and/or liquid dosage form which can be stored for a certain period of time after reconstitution (e.g., 2 hours, 12 hours, 24 hours, 2 days, 5 days, 7 days, 10 days, 2 weeks, a month, two months, or longer).
  • preparatory methods for pharmaceutical compositions include bringing the active ingredient (e.g., an immunotherapeutic composition in accordance with the present disclosure) into association with one or more pharmaceutically acceptable excipients and then shaping and/or packaging the product into a desired single- or multi-dose unit.
  • a pharmaceutical composition in accordance with the disclosure may be prepared, packaged in bulk, packaged as a single unit dose, and/or packaged as a plurality of single unit doses.
  • a “unit dose” refers to a discrete amount of the pharmaceutical composition including a predetermined amount of the active ingredient. The amount of the active ingredient 62 ACTIVE ⁇ 1604904070.2 PATENT Attorney Docket No.
  • PYRO1100-3WO is generally equal to a dose that would be administered to a subject and/or a convenient fraction of such a dose such as, for example, one-half or one-third of such a dose.
  • the relative amounts of active ingredient, pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition in accordance with the disclosure may vary, depending upon the identity, size, and/or condition of the subject treated and/or depending upon the route by which the composition is to be administered. In certain aspects, for example, the composition may include between about 0.1% to 100% (w/w) of an active ingredient.
  • the present disclosure includes kits that are useful for carrying out the methods of the present disclosure.
  • the kit further includes instructions for administration.
  • the kits contain one or more immunotherapeutic compositions.
  • the kit includes a number of unit doses of a pharmaceutical composition containing an immunotherapeutic composition.
  • kits for use in accordance with the present disclosure include instructions (e.g., for administration, for storage, and the like), buffers and/or other reagents.
  • the kit includes (i) at least one immunotherapeutic composition, (ii) a syringe, needle, applicator, or the like for administration of the at least one immunotherapeutic composition to a subject, and (iii) instructions for use.
  • the kit includes a treatment schedule designating when the unit dosages are to be administered.
  • placebo dosages either in a form similar to or distinct from the dosages of the pharmaceutical compositions, are included.
  • kits include one or more containers so that certain of the individual components or reagents may be separately housed.
  • kits may include a means for enclosing the individual containers in relatively close confinement for commercial sale, e.g., a plastic box, in which instructions, packaging materials such as styrofoam, and the like, may be enclosed.
  • a means for enclosing the individual containers in relatively close confinement for commercial sale e.g., a plastic box, in which instructions, packaging materials such as styrofoam, and the like, may be enclosed.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La divulgation tire profit du potentiel thérapeutique de cellules autologues et allogéniques, en particulier de cellules cancéreuses et de lymphocytes T, qui ont été génétiquement modifiés ou mis en contact avec des protéines recombinantes spécifiques, des peptides ou des acides nucléiques codant pour ces entités. Ces cellules modifiées sont amorcées pour déclencher diverses réponses immunitaires et inflammatoires dans le microenvironnement tumoral, ce qui permet de soutenir la capacité naturelle du corps à reconnaître et détruire des cellules tumorales.
PCT/US2023/035802 2022-10-31 2023-10-24 Compositions d'immunothérapie et procédés d'utilisation WO2024097051A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263421050P 2022-10-31 2022-10-31
US63/421,050 2022-10-31
US202363437196P 2023-01-05 2023-01-05
US63/437,196 2023-01-05

Publications (1)

Publication Number Publication Date
WO2024097051A1 true WO2024097051A1 (fr) 2024-05-10

Family

ID=90931239

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/035802 WO2024097051A1 (fr) 2022-10-31 2023-10-24 Compositions d'immunothérapie et procédés d'utilisation

Country Status (1)

Country Link
WO (1) WO2024097051A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007035843A2 (fr) * 2005-09-21 2007-03-29 Dask Technologies, Llc Procédés et compositions pour fonctionnalité d'organes et de tissus
US20070264242A1 (en) * 2000-03-31 2007-11-15 Habib Fakhrai Genetically modified cells expressing a TGF beta inhibitor, the cells being lung cancer cells
WO2017147475A1 (fr) * 2016-02-27 2017-08-31 Hpvvax, Llc Méthode et composition pour traiter le cancer ou une lésion cutanée à l'aide d'un vaccin
WO2021146213A1 (fr) * 2020-01-13 2021-07-22 Gradalis, Inc. Procédés de traitement de cancers à l'aide de polynucléotide codant pour gm-csf et d'agents supplémentaires
US20220023338A1 (en) * 2018-05-15 2022-01-27 The Brigham And Women's Hospital, Inc. Compositions and methods related to tumor cell killers and vaccines
US20220257737A1 (en) * 2019-12-03 2022-08-18 Neuvogen, Inc. Tumor cell vaccines

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070264242A1 (en) * 2000-03-31 2007-11-15 Habib Fakhrai Genetically modified cells expressing a TGF beta inhibitor, the cells being lung cancer cells
WO2007035843A2 (fr) * 2005-09-21 2007-03-29 Dask Technologies, Llc Procédés et compositions pour fonctionnalité d'organes et de tissus
WO2017147475A1 (fr) * 2016-02-27 2017-08-31 Hpvvax, Llc Méthode et composition pour traiter le cancer ou une lésion cutanée à l'aide d'un vaccin
US20220023338A1 (en) * 2018-05-15 2022-01-27 The Brigham And Women's Hospital, Inc. Compositions and methods related to tumor cell killers and vaccines
US20220257737A1 (en) * 2019-12-03 2022-08-18 Neuvogen, Inc. Tumor cell vaccines
WO2021146213A1 (fr) * 2020-01-13 2021-07-22 Gradalis, Inc. Procédés de traitement de cancers à l'aide de polynucléotide codant pour gm-csf et d'agents supplémentaires

Similar Documents

Publication Publication Date Title
JP6884155B2 (ja) 癌治療のための併用免疫療法及びサイトカイン制御療法
Galluzzi et al. Classification of current anticancer immunotherapies
KR20220068240A (ko) 암 치료를 위한 암 요법과 사이토카인 조절 요법의 조합
CN112691120B (zh) 二价锰在制备免疫增强药物或抗肿瘤药物中的应用
US8652836B2 (en) Defective ribosomal products in blebs (DRibbles) and methods of use to stimulate an immune response
US9492518B2 (en) Tumor immunity
AU2018254626B2 (en) Oncolytic vaccinia virus and checkpoint inhibitor combination therapy
KR102646708B1 (ko) T 세포 반응을 촉진하는 방법
JP2021500932A (ja) 全身送達のためのプラットフォーム腫瘍溶解性ベクター
US20220233666A1 (en) Cancer vaccine
US20090010948A1 (en) Anti-tumor vaccines delivered by dendritic cells devoid of interleukin-10
AU2007314433B9 (en) DNA composition for eliciting an immune response against tumor-associated macrophages
Kato et al. RIG-I helicase-independent pathway in sendai virus-activated dendritic cells is critical for preventing lung metastasis of AT6. 3 prostate cancer
US20210085736A1 (en) Immuno-oncolytic modified vaccinia tian tan virus and methods of treating cancer
JP2017509652A (ja) 細胞性の細胞傷害性免疫応答を誘導または延長する方法における使用のための医薬
KR102606179B1 (ko) 암의 치료를 위한 abx196을 포함하는 조합물
Yan et al. Nano-adjuvants and immune agonists promote antitumor immunity of peptide amphiphiles
WO2024097051A1 (fr) Compositions d'immunothérapie et procédés d'utilisation
CA3028168C (fr) Compositions et procedes d'activation de cellules presentatrices d'antigene avec un poliovirus chimerique
CN110678192B (zh) 溶瘤痘苗病毒与免疫检查点抑制剂联合疗法
Bella-Carreño Evaluation of Modified Vaccinia virus Ankara based locoregional immunotherapy in peritoneal carcinomatosis models
WO2022011651A1 (fr) Procédé d'immunothérapie par administration ciblée de chimiokines et de cytokines par une cellule souche mésenchymateuse
Milling Priming systemic anti-tumor immunity via in situ immunomodulation of the tumor microenvironment
Gonzales Development of therapeutic vaccine strategies and pre-clinical animal tumor models for head and neck cancers
Gordy EFFICACY IN A MOUSE MELANOMA MODEL OF A DENDRITIC CELL-TARGETING THERAPEUTIC DNA VACCINE AND ENHANCEMENT OF ITS ACTIVITY BY CO-TREATMENT WITH ANTIBODIES TO NEUTRALIZE INTERLEUKIN-10