US20230340052A1 - Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof - Google Patents

Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof Download PDF

Info

Publication number
US20230340052A1
US20230340052A1 US18/172,938 US202318172938A US2023340052A1 US 20230340052 A1 US20230340052 A1 US 20230340052A1 US 202318172938 A US202318172938 A US 202318172938A US 2023340052 A1 US2023340052 A1 US 2023340052A1
Authority
US
United States
Prior art keywords
bite
car
diakine
icam
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/172,938
Inventor
John Mumm
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Deka Biosciences Inc
Original Assignee
Deka Biosciences 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 Deka Biosciences Inc filed Critical Deka Biosciences Inc
Priority to US18/172,938 priority Critical patent/US20230340052A1/en
Assigned to Deka Biosciences, Inc. reassignment Deka Biosciences, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MUMM, JOHN
Publication of US20230340052A1 publication Critical patent/US20230340052A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K38/20Interleukins [IL]
    • A61K38/2026IL-4
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5428IL-10
    • 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
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5406IL-4
    • 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
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes

Definitions

  • Cytokine Release Syndrome is a dose and treatment related toxicity resulting from Bispecific T cell Engager (BiTE) (Hosseini, 2020) activation of T cells and application of Chimeric Antigen Receptor T cells (CAR-T) to patients (Maude, 2014; Norelli, 2018).
  • CRS is defined by the uncontrolled induction of high levels of interleukin 6 (IL-6) and interleukin 1 beta (IL-1 ⁇ ) predominantly (Liu, 2018), Tumor Necrosis Factor alpha (TNF ⁇ ) (Chen, 2021) and interferon gamma (IFN ⁇ ) (Shimabukuro-Vornhagen, 2018) in the serum of treated patients.
  • IL-6 interleukin 6
  • IL-1 ⁇ interleukin 1 beta
  • TNF ⁇ Tumor Necrosis Factor alpha
  • IFN ⁇ interferon gamma
  • the inventor has found that treating monocytes with IL-10 or IL-4, IL-12, IL-15, IL-7 or any combination of the foregoing, or any half-life extended version thereof or any diakine comprising IL-10, IL-4, Il-12, IL-15, IL-7, or IL-2 that will engage cognate cytokine receptors on monocytes and directly inhibits the induction of proinflammatory cytokines by IL-2.
  • the application relates to a method of treating a monocyte with either IL-10 or IL-4, IL-12, IL-15, IL-7, half-life extended versions thereof, a combination of IL-10 and IL-4, IL-10 and IL-2, IL-10 and IL-7, IL-10 and IL-12, IL-10 and IL-15, or a fusion protein or diakine comprising at least two cytokines, wherein at least one of the at least two cytokines is IL-10 or IL-4, IL-12, Il-15, or IL-7 to reduce CRS associated with BiTE or CAR-T therapies.
  • monocytes are treated with IL-10, or IL-4, IL-12, IL-15, IL-7 or a half-life extended version thereof or a diakine comprising at least one of at least two cytokines is IL-10 or IL-4, IL-12, IL-15, or IL-7.
  • a patient will be treated with IL-10 or a half-life extended version thereof and a BiTE or CAR-T.
  • a patient will be treated with IL-4 or a half-life extended version thereof and a BiTE or CAR-T.
  • a patient is treated with a diakine comprising IL-10 and any one of IFN- ⁇ , IL-2, IL-4, IL-7, IL-12, IL-15, IL-21 or IL-27 in an amount sufficient to reduce CRS.
  • a patient is treated with a diakine comprising IL-4 and any one of IFN- ⁇ , IL-2, IL-7, IL-10, IL-12, IL-15, IL-21, or IL-27 in an amount sufficient to reduce CRS.
  • the application relates to a method of reducing BiTE or CAR-T associated CRS comprising administering to a subject thereof a BiTE or CAR-T therapeutic modality in combination with IL-10, IL-4, or any combination thereof, or a diakine comprising at least one of IL-10, IL-4, IL-2.
  • the method comprises administering to a patient in need thereof the BiTE or CAR-T therapy before, after or simultaneously with the IL-10 or IL-4, IL-12, IL-15, or IL-7, or half-life extended versions thereof, or a diakine comprising IL-10 or IL-4 combined with IFN- ⁇ , IL-2, IL-7, IL-12, IL-15, IL-21 or IL-27.
  • the application relates to a method of inhibiting the induction of proinflammatory cytokines in a patient undergoing BiTE or CAR-T therapy comprising administering to the patient undergoing said therapy a dose of IL-10 or IL-4 or half-life extended versions thereof, or a diakine comprising IL-10 or IL-4 in combination with IFN- ⁇ , IL-2, IL-7, IL-12, IL-15, IL-21 or IL-27 in an amount sufficient to suppress CRS caused by the proinflammatory cytokines.
  • FIG. 1 are graphs measuring the level of IL1 ⁇ , IFN ⁇ , TNF ⁇ , IFN ⁇ 2a, IL-12, and IL-6 in PBMC in response to increasing levels of IL-2 exposure.
  • FIG. 2 are graphs measuring the levels of IFNy, IL-6, and TNF ⁇ induction from PBMC to response to 1 ug/mL of anti-CD3 in the presence of a diakine, IL-10 and IL-2.
  • FIG. 3 mouse serum cytokines obtained from diakine exposure in vivo.
  • FIG. 4 non-human primate serum cytokine levels obtained from diakine exposure in vivo.
  • FIG. 5 graphs measuring cytotoxicity of CD8+ T cell exposed to diakine and BiTE.
  • FIG. 6 graphs measuring IFNg and TNFa levels in CD8+ T cells exposed to varying concentrations of BiTE.
  • FIG. 7 schematic diagram of proposed IL-2 mediated CRS circuit in response to BiTE or CAR-T therapy.
  • FIG. 8 combination of a diakine comprising IL-10 and IL-2 which targets EGFR in combination with CD3xCD19 BiTE enhances CD8+ T cell directed tumor cell cytolysis.
  • FIG. 9 combination of a diakine (DK2 10 EGFR) with CD3xCD19 BiTE exhibits enhanced cytolytic effector molecules and controlled CRS
  • FIG. 10 intracellular FACS analysis of a diakine (DK2 10 EGFR) and CD3xCD19 BiTE treatment of PBMC cultures containing Raji GFP+ tumor cells
  • Exemplary aspects are described herein in the context of a using IL-10, IL-4, IL-12, IL-15, IL-7, or half-life extended versions thereof, or a diakine comprising at least one of IL-10 or IL-4 in combination with IFN- ⁇ , IL-2, IL-7, IL-12, IL-15, or IL-27 in a method of suppressing, inhibiting, reducing or preventing CRS associated with BiTE or CAR-T therapeutic modalities.
  • the embodiments described herein employ conventional methods and techniques of molecular biology, biochemistry, pharmacology, chemistry, and immunology, which are well known to a person skilled in the art. See, e.g., Sambrook, et al., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.); Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C. Blackwell eds., Blackwell Scientific Publications); A. L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition).
  • IL-10 variants including but not limited to human, mouse, CMV and/or EBV forms of IL-10, as well as the assays for testing the IL-10 variants, diakines, and other are known assay methods.
  • the term “about”, refers to a deviance of between 0.0001-5% from the indicated number or range of numbers. In one embodiment, the term “about”, refers to a deviance of between 1-10% from the indicated number or range of numbers. In one embodiment, the term “about”, refers to a deviance of up to 25% from the indicated number or range of numbers. In a more specific embodiment, the term “about” refers to a difference of 1-25% in terms of nucleotide sequence homology or amino acid sequence homology when compared to a wild-type sequence.
  • agent as it relates to the various interleukins (IL), such “IL-10 agent” or “IL-4 agent” and the like, are intended to be construed broadly and include, for example, human and non-human forms of the interleukin polypeptides, including homologs, variants (including muteins), fragments thereof, and fusion proteins, as well as polypeptides having, for example, a leader sequence (e.g., the signal peptide), and modified versions of the foregoing.
  • the present disclosure also contemplates nucleic acid molecules encoding the foregoing, vectors and the like containing the nucleic acid molecules, and cells (e.g., transformed cells and host cells) that express the interleukin agents.
  • variant refers to biologically active derivatives of the reference molecule, that retain a desired activity, such as, for example, anti-inflammatory activity.
  • desired activity such as, for example, anti-inflammatory activity.
  • variant refers to a compound or compounds having a native polypeptide sequence and structure with one or more amino acid additions, substitutions (which may be conservative in nature), and/or deletions, relative to the native molecule.
  • an EBV IL-10 variant molecule is one that differs from wild-type EBV IL-10 by having one or more amino acid (or nucleotide sequence encoding the amino acid) additions, substitutions and/or deletions.
  • fusion protein refers to a combination or conjugation of two or more proteins or polypeptides that results in a novel arrangement of proteins that do not normally exist naturally.
  • the fusion protein is a result of covalent linkages of the two or more proteins or polypeptides.
  • the two or more proteins that make up the fusion protein may be arranged in any configuration from amino-terminal end (“NH 2 ”) to carboxy-terminal end (“COOH”).
  • homolog refers to the percent identity between at least two polynucleotide sequences or at least two polypeptide sequences. Sequences are homologous to each other when the sequences exhibit at least about 50%, preferably at least about 75%, more preferably at least about 80%-85%, preferably at least about 90%, and most preferably at least about 95%-98% sequence identity over a defined length of the molecules.
  • sequence identity refers to an exact nucleotide-by-nucleotide or amino acid-by-amino acid correspondence.
  • the sequence identity may range from 100% sequence identity to 50% sequence identity.
  • a percent sequence identity can be determined using a variety of methods including but not limited to a direct comparison of the sequence information between two molecules (the reference sequence and a sequence with unknown percent identity to the reference sequence) by aligning the sequences, counting the exact number of matches between the two aligned sequences, dividing by the length of the reference sequence, and multiplying the result by 100. Readily available computer programs can be used to aid in the identification of percent identity.
  • subject refers to a vertebrate, preferably a mammal.
  • Mammals include, but are not limited to, murine, rodent, simian, human, farm animals, sport animals, and certain pets.
  • administering includes routes of administration which allow the active ingredient of the application to perform their intended function.
  • a “therapeutically effective amount” or “effective amount” as it relates to, for example, administering the IL variants, fusion proteins, dual cytokine fusion proteins, or diakine thereof described herein, refers to an amount sufficient to promote certain biological activities. These might include, for example, suppression of myeloid cell function, enhanced Kupffer cell activity, and/or lack of any effect on CD8 + T cells or enhanced CD8 + T-cell activity as well as blockade of mast cell upregulation of Fc receptor or prevention of degranulation or to promote or enhance to effects of combination therapeutics (e.g., CAR-T therapies) or to suppress the induction of cytokines from monocytes or macorphages.
  • an “effective amount” will ameliorate or prevent a symptom or sign of the medical condition. Effective amount also means an amount sufficient to allow or facilitate diagnosis.
  • treat refers to a method of reducing the effects of a disease or condition. Treatment can also refer to a method of reducing the underlying cause of the disease or condition itself rather than just the symptoms.
  • the treatment can be any reduction from native levels and can be, but is not limited to, the complete ablation of the disease, condition, or the symptoms of the disease or condition.
  • half-life extended refers to a protein that includes one of more additional moiety or moieties (such as proteins or PEGylation) that extends and/or enhances the circulation time within a subject anywhere in the range of 1-100 fold longer than a protein in the absence of the additional moiety or moieties.
  • a half-life extended IL refers to an IL-10 or IL-4, IL-7, IL-12, or IL-15 conjugated to a scFv whereby the circulation time is extended in the range of 1-10 fold longer than the IL in the absence of the scFv.
  • a half-life extended version of an IL will adopt the configuration of formula I
  • DK diakine or “DK”, as used in this application, refers to a dual cytokine fusion protein comprising two monomers of a dimeric cytokine, which can be either a homodimer such as IL-10 or IL-10 variants, or a heterodimer such as IL-12 or IL-12 variants, that is fused together with a monomeric cytokine, such as IL-2, IL-4, IL-7, IL-15, IL-21, IL-28, IL-29, or with a dimeric cytokine, such as IL-12 or IL-10, whereby both of the dimeric cytokine and monomeric cytokine being fused onto a half-life extending antigen targeting domain.
  • a monomeric cytokine such as IL-2, IL-4, IL-7, IL-15, IL-21, IL-28, IL-29
  • a dimeric cytokine such as IL-12 or IL-10
  • diakines are described in detail in U.S. Pat. 11,292,822 (IL-10 based diakines) and co-pending U.S. Application 18/065,504 (dual dimeric cytokine based diakines), both of which are incorporated by reference in their entireties.
  • a diakine which may be used in a method in combination with a BiTE or CAR-T, is represented by formula II
  • dimeric cytokine may include IFN- ⁇ , IL-2, IL-4, IL-7, IL-10, IL-12, IL-15, IL-21, and IL-27.
  • the monomeric cytokine may include IL-6, IL-4, IL-1, IL-2, IL-3, IL-5, IL-7, IL-8, IL-9, IL-15, IL-21 IL-26, IL-27, IL-28, IL-29, GM-CSF, G-CSF, interferons- ⁇ , - ⁇ , - ⁇ , TGF- ⁇ , or tumor necrosis factors - ⁇ , - ⁇ , basic FGF, EGF, PDGF, IL-4, IL-11, or IL-13.
  • the VH and VL of the diakine is a scFv and may be derived from any monoclonal antibody but is preferably derived from an antibody that is capable of targeting a specific antigen.
  • the monoclonal antibody from which the scFv (as it applies to Formula I and II) may be derived is selected from EGFR; CD52; CD14; various immune check point targets, such as but not limited to PD-L1, PD-1, TIM3, BTLA, LAG3 or CTLA4; CD19; CD20; CD22; CD47; GD-2; VEGFR1; VEGFR2; HER2; PDGFR; EpCAM; ICAM (ICAM-1, -2, -3, -4, -5), VCAM, CD14, FAP ⁇ ; 5T4; Trop2; EDB-FN; TGF ⁇ Trap; MAdCAM, ⁇ 7 integrin subunit; ⁇ 4 ⁇ 7 integrin; ⁇ 4 integrin SR-A1; SR-A3
  • the scFv (as it applies to Formula I and II) is an engrafted scFv, whereby the VH and VL framework region is derived from a first antibody (such as an anti-ebola antibody) and the CDRs are obtained from a second antibody (such as but not limited to EGFR; CD52; CD14; various immune check point targets, such as but not limited to PD-L1, PD-1, TIM3, BTLA, LAG3 or CTLA4; CD19; CD20; CD22; CD47; GD-2; VEGFR1; VEGFR2; HER2; PDGFR; EpCAM; ICAM (ICAM-1, -2, -3, -4, -5), VCAM, CD14, FAP ⁇ ; 5T4; Trop2; EDB-FN; TGF ⁇ Trap; MAdCAM, ⁇ 7 integrin subunit; ⁇ 4 ⁇ 7 integrin; ⁇ 4 integrin SR-A1; SR-A3; SR-A
  • the scFv is derived from an anti-Ebola antibody where the VH and VL framework regions of an anti-ebola antibody are substituted or engrafted with 6 CDR regions from an antibody specific for EGFR; CD52; CD14; various immune check point targets, such as but not limited to PD-L1, PD-1, TIM3, BTLA, LAG3 or CTLA4; CD19; CD20; CD22; CD47; GD-2; VEGFR1; VEGFR2; HER2; PDGFR; EpCAM; ICAM (ICAM-1, -2, -3, -4, -5), VCAM, CD14, FAP ⁇ ; 5T4; Trop2; EDB-FN; TGF ⁇ Trap; MAdCAM, ⁇ 7 integrin subunit; ⁇ 4 ⁇ 7 integrin; ⁇ 4 integrin SR-A1; SR-A3; SR-A4; SR-A5; SR-A6; SR-B; dSR-C
  • the diakine which may be used in a method in combination with a BiTE or a CAR-T, is a diakine described in U.S. Pat. 11,292,822 (IL-10 based diakines) or co-pending U.S. Application 18/065,504 (dual dimeric cytokine based diakines), both of which are incorporated by reference in their entireties.
  • the diakine which may be used in a method in combination with a BiTE or a CAR-T, is protein represented by formula III
  • IL-10 monomer “second monoclonal antibody” “Z” Human IL-10 (Seq ID No: 1) EGFR 2 15 7 28 29 IFN-alpha 21 HER2 2 15 7 28 29 IFN-alpha 21 VEGFR2 2 15 7 28 29 IFN-alpha 21 PDGFR 2 15 7 28 29 IFN-alpha 21 GPC3 2 15 7 28 29 IFN-alpha 21 PD-L1 2 15 7 28 29 IFN-alpha 21 CD19 2 15 7 28 29 IFN-alpha 21 CD20 2 15 7 28 29 IFN-alpha 21 CD22 2 15 7 28 29 IFN-alpha 21 PSMA 2 15 7 28 29 IFN-alpha 21 CEA 2 15 7 28 29 IFN-alpha 21 BCMA 2 15 7 28 29 IFN-alpha 21 gpA33 2 15 7 28 29 IFN-alpha 21 CD33 2 15 7 28 29 IFN-alpha 21 DLL3 2 15 7 28 29 IFN-alpha 21 MUC17 2 15 7 28 29 IFN-alpha 21 CL
  • the diakine which may be used in a method in combination with a BiTE or CAR-T, is a diakine described in co-pending U.S. Application 18/065,504 in Tables 2a-2d and 3a-3d, which is incorporated by reference in its entirety.
  • the protein or nucleic acid molecules encoding dual cytokine fusion protein or DK may be formulated as a pharmaceutical composition comprising a therapeutically effective amount of the dual cytokine fusion protein and a pharmaceutical carrier and/or pharmaceutically acceptable excipients.
  • the pharmaceutical composition may be formulated with commonly used buffers, excipients, preservatives, stabilizers.
  • the pharmaceutical compositions comprising the dual cytokine fusion protein is mixed with a pharmaceutically acceptable carrier or excipient.
  • Various pharmaceutical carriers are known in the art and may be used in the pharmaceutical composition.
  • the carrier can be any compatible, non-toxic substance suitable for delivering the dual cytokine fusion protein compositions of the application to a patient.
  • Carriers may also include any poloxamers generally known to those of skill in the art, including, but not limited to, those having molecular weights of 2900 (L64), 3400 (P65), 4200 (P84), 4600 (P85), 11,400 (F88), 4950 (P103), 5900 (P104), 6500 (P105), 14,600 (F108), 5750 (P123), and 12,600 (F127). Carriers may also include emulsifiers, including, but not limited to, polysorbate 20, polysorbate 40, polysorbate 60, and polysorbate 80, to name a few.
  • Non-aqueous carriers such as fixed oils and ethyl oleate may also be used.
  • the carrier may also include additives such as substances that enhance isotonicity and chemical stability, e.g., buffers and preservatives, see, e.g., Remington’s Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary, Mack Publishing Company, Easton, Pa. (1984).
  • Formulations of therapeutic and diagnostic agents may be prepared by mixing with physiologically acceptable carriers, excipients, or stabilizers in the form of lyophilized powders, slurries, aqueous solutions or suspensions, for example.
  • the pharmaceutical composition will be formulated for administration to a patient in a therapeutically effective amount sufficient to provide the desired therapeutic result. Preferably, such amount has minimal negative side effects.
  • the amount of dual cytokine fusion protein administered will be sufficient to treat or prevent inflammatory diseases or condition.
  • the amount of dual cytokine fusion protein administered will be sufficient to treat or prevent immune diseases or disorders.
  • the amount of diakine or dual cytokine fusion protein administered will be sufficient to treat or prevent CRS mediated by BiTE or CAR-T therapy.
  • the amount administered may vary from patient to patient and will need to be determined by considering the subject’s or patient’s disease or condition, the overall health of the patient, method of administration, the severity of side-effects, and the like.
  • An effective amount for a particular patient may vary depending on factors such as the condition being treated, the overall health of the patient, the method route and dose of administration and the severity of side effects.
  • the appropriate dose administered to a patient is typically determined by a clinician using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment. Generally, the dose begins with an amount somewhat less than the optimum dose and it is increased by small increments thereafter until the desired or optimum effect is achieved relative to any negative side effects.
  • Important diagnostic measures include those of symptoms of, e.g., the inflammation or level of inflammatory cytokines produced.
  • the method for determining the dosing of the presently described dual cytokine fusion protein will be substantially similar to that described in U.S. Pat. 10,858,412.
  • the presently described dual cytokine fusion protein will have a dosing in the range of 0.01 mg/kg to 1 mg/kg, preferably 0.025 mg/kg to 0.5 mg/kg.
  • the dual cytokine fusion protein may be administered daily, three times a week, twice a week, weekly, bimonthly, or monthly. An effective amount of therapeutic will impact the level of CRS inhibited caused by the BiTE or CAR-T therapy.
  • the diakine will be dosed at a concentration of 0.1 ng/mL to 200 ng/mL, preferably, 10 ng/mL to 100 ng/mL. Generally, the addition of a diakine will lower the required dose for a BiTE or CAR-T modality.
  • compositions of the application can be administered orally or injected into the body.
  • Formulations for oral use can also include compounds to further protect the IL or DK molecules from proteases in the gastrointestinal tract. Injections are usually intramuscular, subcutaneous, intradermal or intravenous. Alternatively, intra-articular injection or other routes could be used in appropriate circumstances.
  • Parenterally administered dual cytokine fusion protein are preferably formulated in a unit dosage injectable form (solution, suspension, emulsion) in association with a pharmaceutical carrier and/or pharmaceutically acceptable excipients.
  • compositions of the application may be introduced into a patient’s body by implantable or injectable drug delivery system.
  • adoptive cell therapy such as adoptive T-cell therapy
  • methods may include autologous transfer (i.e., derived from the patient) or allogenic transfer (i.e. derived from another subject other than the patient to be treated).
  • the CAR-T or TCR-T cells are administered by methods known and conventionally practiced by those familiar with adaptive cell therapy.
  • the administration method includes, but is not limited to bolus infusion, intravenous or subcutaneous injections, intraocular injection, periocular injection, subretinal injection, intravitreal injection, trans-septal injection, subscleral injection, intrachoroidal injection, intracameral injection, subconjectval injection, subconjuntival injection, sub-Tenon’s injection, retrobulbar injection, peribulbar injection, or posterior juxtascleral delivery.
  • they are administered by parenteral, intrapulmonary, and intranasal, or intralesional or intrtumoral administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the recombinantly engineered CAR-T or TCR-T is administered as a single bolus administration, multiple bolus or continuous infusion.
  • CAR-T therapies include idecabtagene vicleucel, lisocabtagene maraleucel, brexucabtagene autoleucel, tisagenlecleucel, or axicabtagene ciloleucel.
  • the diakine and the CAR-T are administered in separate subsequent time periods, wherein, for example, the diakine (such as DK2 10 vegfr2 or DK2 10 EGFR or any of those referenced in Table 1, above) is administered prior to the administration of a recombinantly engineered CAR-T cell.
  • the diakine and the CAR-T are administered simultaneously.
  • the diakine is administered 1-3 days before the CAR-T therapy and then simultaneously administered along with the CAR-T, and/or 1-7 days following CAR-T administration.
  • the diakine may be administered once a day or week, or 2-3 times a week in combination or conjunction with the CAR-T.
  • the diakine is utilized in the expansion and/or thawing procedure of the CAR-T cells prior to administration.
  • the CAR-T Upon reconstituting CAR-T cells from cryopreserved stock, the CAR-T are typically rested in the presence of CAR-T beneficial cytokines (e.g., low dose IL-2).
  • the CAR-T cells may be primed or expanded from cryopreserved stocks in the presence of a diakine.
  • the CAR-T is expanded or primed in the presence of 0.001 to 300 ng/mL of a diakine, more preferably 0.01 to 200 ng/mL of a diakine.
  • the diakine and the BiTE are administered in separate subsequent time periods, wherein the diakine (e.g., DK2 10 CD20 or DK2 10 EGFR or DK2 10 HER2 or DK2 10 HER3) is administered 1-3 days before administering the BiTE (e.g., CD3xCD19 BiTE).
  • the diakine is administered 1-3 days before the BiTE and then simultaneously administered along with the BiTE, and/or 1-7 days following BiTE administration.
  • the diakine may be administered once a day or week, or 2-3 times a week in combination or conjunction with the BiTE.
  • a BiTE will generally follow the format of a bispecific antibody having an anti-CD3 and an anti-TAA fused together.
  • method combines an IL-10 or IL-4, or half-life extended versions thereof, or a diakine comprising IL-10 or IL-4 and IFN- ⁇ , IL-2, IL-7, IL-12, IL-15, IL-21 or IL-27, in conjunction with a BiTE, such as those that have bispecificity for CD3 with CD33, BCMA, CD19, CD20, CD22, PSMA, EGFR, DLL3, MUC17, CLDN18, CEA, HER2, HER3, EpCAM, gpA33, GPC3, GD2 5T4, VEGFR2, PDGFR, PDL1, or PD1 to name a few.
  • the BiTE is an anti-CD3 and anti-CD19, anti-CD20, anti-HER2, anti-HER3, anti-PSMA, or anti-BCMA .
  • CRS CRS is associated by concomitant increases in serum IL-6, IL-1 ⁇ , TNF ⁇ , IFN ⁇ / ⁇ , IL-12 and IL-23. While these cytokines are generally associated with CRS, it is not known whether one, two or all of these cytokines working together to lead to the toxic side effects associated with CRS. The inventor believes that a potential root cause of CRS is the fact than many patients with severe CRS induced by BiTE or CAR-T treatment develop vascular leak syndrome. Vascular or capillary leak syndrome is also predominantly observed as the dose limiting and often lethal toxicity associated with high dose IL-2 therapy.
  • PBMC peripheral blood mononuclear cells
  • IL-2 proinflammatory cytokines
  • the inventor believes that the similarities between the cytokines induced by treatment with IL-2 alone, and the apparent blockade of the induction of these secondary cytokines by a diakine comprising IL-10 (e.g., DK2 10 (EGFR)) or IL-4, IL-12, IL-15, IL-7, or any combination thereof will suppress or block IL-2 mediated CRS.
  • IL-10 e.g., DK2 10 (EGFR)
  • IL-4 IL-12
  • IL-15 IL-15
  • IL-7 IL-7
  • FIG. 2 Further investigation of whether coupling IL-10 to IL-2 indicates that the application of DK2 10 (EGFR) to mice prevents the induction of peripheral cytokines induced by IL-2 alone.
  • FIG. 3 Furthermore, the treatment of non-human primates with DK2 10 (EGFR) similarly does not lead to significant inductions of peripheral plasma cytokines.
  • FIG. 4 the treatment of non-human primates with DK2 10 (EGFR) similarly does not lead to significant inductions of peripheral plasma cytokines.
  • CD8+ T cells pre-exposure of CD8+ T cells to a diakine (e.g., DK2 10 (EGFR)) dramatically enhances the cells subsequent capacity to engage BiTE’s and effect cytolysis of tumor cells.
  • a diakine e.g., DK2 10 (EGFR)
  • EGFR DK2 10
  • the molecular circuit responsible for BiTE or CAR-T mediated CRS is a cascade of pro-inflammatory cytokine release, that is first triggered by CD4+ T cell engagement with BiTE or CAR-T, which subsequently triggers IL-2 secretion and inducement of monocyte/macrophage to undergo additional secretion of proinflammatory cytokines.
  • FIG. 7 from monocytes/macrophages.
  • DK2 10 (EGFR) has been previously shown to prime CD8+ T cell for subsequent tumor cytolysis in vitro and in vivo. See, U.S. Pat. 11,292,822.
  • DK2 10 (EGFR) which is a representative example of a diakine, is shown to both enhance BiTE mediated tumor cell cytolysis as well as suppress BiTE mediated CRS.
  • DK2 10 (EGFR) is shown to enhance BiTE mediated tumor cell cytotoxicity.
  • the combinatorial anti-tumor effects of DK2 10 (EGFR) with the CD19 BiTE is assessed in multiple rounds of exposure to target tumor cells.
  • CD8+ T cells are isolated from fresh donor Leukopaks via magnetic bead isolation per the manufacturer’s suggested protocol (Miltenyi).
  • the isolated CD8+ T cells are plated at 2.5 ⁇ 10 6 cells/well and exposed for 2 days in various concentrations (0 or 100 ng/mL) of DK2 10 (EGFR) in AIMV. Following the 2 days of exposure to the various concentrations of DK2 10 (EGFR), the CD8+ T cells are harvested, counted, washed, and finally resuspended in the corresponding concentration of DK2 10 (EGFR).
  • Raji cells which constitutively express Green Florescent Protein (GFP) are counted, washed and resuspended in varying concentrations (0 or 0.1 ng/mL) of CD19 BiTE.
  • the CD8+ cells (effector) and Raji-GFP cells (target) are then combined at a 10:1 effector to target ratio.
  • the mixture of effector and target cells, which are exposed to CD19 BiTE alone, DK2 10 (EGFR) alone, or the combination of CD19 BiTE and DK2 10 (EGFR) were monitored over 5 days using an IncuCyte® S3 Live-Cell Analysis System (Essen Bioscience/Sartorius).
  • CD3xCD19 BiTE when combined with DK2 10 (EGFR) enhances tumor cell cytotoxicity.
  • FIGS. 5 and 8 CD3XCD19 BiTE and DK2 10 (EGFR) are tested both alone and in combination using normal, healthy human donor derived CD8+ T cells.
  • FIG. 8 Effector cells went through multiple rounds (5 rounds of serial cytolysis) of exposure to target tumor cells (Raji-GFP), and cytotoxicity was measured via the disappearance of GFP.
  • Raji-GFP target tumor cells
  • In vitro treatment with the BiTE in combination with DK2 10 (EGFR) suggests that activation of T cells with DK2 10 (EGFR) enhances responses to BITE’s when assessing CD8+ T cell anti-Raji GFP+ responses.
  • PBMCs are isolated from Leukopaks collected from healthy donors using the Ficoll density gradient method. Equal volumes of HBSS and donor samples were transferred to conical tubes, individually. Ficoll was slowly added to create a bottom layer then samples were centrifuged at 400 xg for 30 minutes at 25° C. PBMCs were harvested from the top layer then washed twice using Aim V media (300 xg, 8 minutes). The isolated PBMCs were plated at 2 ⁇ 10 6 cells/well in various concentrations (0 or 100 ng/mL) of DK2 10 EGFR in AIMV then incubated for two days (37° C., 5% CO 2 ).
  • Raji tumor cells were counted, washed and resuspended in varying concentrations of CD19 BiTE (0, or 0.1 ng/mL final), with or without DK2 10 (EGFR) (100 ng/mL) using AIMV and allowed to prime for two-days.
  • the PBMCs assuming 10% CD8+ T cells (effector), and Raji cells (target) were combined at a 10:1 effector to target ratio. After 24-hour incubation, supernatants are harvested and cytokine secretion was measured via multiplexed capture assay (MSD) and ELISA.
  • MSD multiplexed capture assay
  • FIG. 9 Illustrated in FIG. 9 , the data suggests that the presence of DK2 10 (EGFR) under these conditions significantly promotes tumor cell lysis (induction of IFN-gamma, Granzyme B, and Perforin) while limiting the induction of BiTE mediated CRS at a non-functional concentration of BiTE (0.1 ng/mL). Represented data is taken at 24 hrs., from exposure 1. Longitudinal data indicates a reduction of CRS over time in these conditions.
  • FIG. 10 Illustrated in FIG. 9 , the data suggests that the presence of DK2 10 (EGFR) under these conditions significantly promotes tumor cell lysis (induction of IFN-gamma, Granzyme B, and Perforin) while limiting the induction of BiTE mediated CRS at a non-functional concentration of BiTE (0.1 ng/mL). Represented data is taken at 24 hrs., from exposure 1. Longitudinal data indicates a reduction of CRS over time in these conditions.
  • Hosseini I. (2020). Mitigating the risk of cytokine release syndrome in Phase I trial of CD20CD3 bispecific antibody mosunetuzumab in NHL impact of translational system modeling. Systems Biology and Applications .

Abstract

The disclosure provides for various methods including a method of reducing the severity of bispecific T cell engager (BiTE) or chimeric antigen receptor T cell (CAR-T) induced cytokine release syndrome (CRS) comprising administering to a patient in need thereof an amount of a composition comprising an interleukin 10 (IL-10) or an IL-10 agent, an interleukin 4 (IL-4) or an IL-4 agent, or combinations thereof.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. Provisional Application No. 63/312,697, filed Feb. 22, 2022, the disclosure of which is incorporated by reference in its entirety.
  • REFERENCE TO AN ELECTRONIC SEQUENCE LISTING
  • The contents of the electronic sequence listing (039451-00100-Sequence-Listing.xml; Size: 37,997 bytes; and Date of Creation: Feb. 22, 2023) are herein incorporated by reference in its entirety.
  • BACKGROUND
  • Cytokine Release Syndrome (CRS) is a dose and treatment related toxicity resulting from Bispecific T cell Engager (BiTE) (Hosseini, 2020) activation of T cells and application of Chimeric Antigen Receptor T cells (CAR-T) to patients (Maude, 2014; Norelli, 2018). CRS is defined by the uncontrolled induction of high levels of interleukin 6 (IL-6) and interleukin 1 beta (IL-1β) predominantly (Liu, 2018), Tumor Necrosis Factor alpha (TNFα) (Chen, 2021) and interferon gamma (IFNγ) (Shimabukuro-Vornhagen, 2018) in the serum of treated patients.
  • To date, the molecular circuit responsible for the induction of CRS is not known. Based on the inventor’s findings, it has been determined that the induction of CRS in BiTE and CAR-T patients is likely due to T Cell receptor (TCR):BiTE or CAR-T:Tumor Associated Antigen (TAA) clustering mediated activation of CD4+ T cells. This activation, in-turn, leads to the secretion of IL-2 (Brandl, 2007). The secretion of IL-2 then drives monocyte secretion of the proinflammatory cytokines associated with CRS (Bosco, 2000; Musso, 1992; Strieter, 1989).
  • SUMMARY OF VARIOUS EMBODIMENTS
  • The inventor has found that treating monocytes with IL-10 or IL-4, IL-12, IL-15, IL-7 or any combination of the foregoing, or any half-life extended version thereof or any diakine comprising IL-10, IL-4, Il-12, IL-15, IL-7, or IL-2 that will engage cognate cytokine receptors on monocytes and directly inhibits the induction of proinflammatory cytokines by IL-2.
  • In one aspect, the application relates to a method of treating a monocyte with either IL-10 or IL-4, IL-12, IL-15, IL-7, half-life extended versions thereof, a combination of IL-10 and IL-4, IL-10 and IL-2, IL-10 and IL-7, IL-10 and IL-12, IL-10 and IL-15, or a fusion protein or diakine comprising at least two cytokines, wherein at least one of the at least two cytokines is IL-10 or IL-4, IL-12, Il-15, or IL-7 to reduce CRS associated with BiTE or CAR-T therapies. In one embodiment, monocytes are treated with IL-10, or IL-4, IL-12, IL-15, IL-7 or a half-life extended version thereof or a diakine comprising at least one of at least two cytokines is IL-10 or IL-4, IL-12, IL-15, or IL-7. In another embodiment a patient will be treated with IL-10 or a half-life extended version thereof and a BiTE or CAR-T. In yet another embodiment, a patient will be treated with IL-4 or a half-life extended version thereof and a BiTE or CAR-T. In another embodiment, a patient is treated with a diakine comprising IL-10 and any one of IFN-α, IL-2, IL-4, IL-7, IL-12, IL-15, IL-21 or IL-27 in an amount sufficient to reduce CRS. In yet another embodiment a patient is treated with a diakine comprising IL-4 and any one of IFN-α, IL-2, IL-7, IL-10, IL-12, IL-15, IL-21, or IL-27 in an amount sufficient to reduce CRS.
  • In another aspect, the application relates to a method of reducing BiTE or CAR-T associated CRS comprising administering to a subject thereof a BiTE or CAR-T therapeutic modality in combination with IL-10, IL-4, or any combination thereof, or a diakine comprising at least one of IL-10, IL-4, IL-2. In one embodiment, the method comprises administering to a patient in need thereof the BiTE or CAR-T therapy before, after or simultaneously with the IL-10 or IL-4, IL-12, IL-15, or IL-7, or half-life extended versions thereof, or a diakine comprising IL-10 or IL-4 combined with IFN-α, IL-2, IL-7, IL-12, IL-15, IL-21 or IL-27.
  • In yet another aspect, the application relates to a method of inhibiting the induction of proinflammatory cytokines in a patient undergoing BiTE or CAR-T therapy comprising administering to the patient undergoing said therapy a dose of IL-10 or IL-4 or half-life extended versions thereof, or a diakine comprising IL-10 or IL-4 in combination with IFN-α, IL-2, IL-7, IL-12, IL-15, IL-21 or IL-27 in an amount sufficient to suppress CRS caused by the proinflammatory cytokines.
  • The above simplified summary of representative aspects serves to provide a basic understanding of the present disclosure. This summary is not an extensive overview of all contemplated aspects, and is intended to neither identify key or critical elements of all aspects nor delineate the scope of any or all aspects of the present disclosure. Its sole purpose is to present one or more aspects in a simplified form as a prelude to the more detailed description of the disclosure that follows. To the accomplishment of the foregoing, the one or more aspects of the present disclosure include the features described and exemplarily pointed out in the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 are graphs measuring the level of IL1β, IFNγ, TNFα, IFNα2a, IL-12, and IL-6 in PBMC in response to increasing levels of IL-2 exposure.
  • FIG. 2 are graphs measuring the levels of IFNy, IL-6, and TNFα induction from PBMC to response to 1 ug/mL of anti-CD3 in the presence of a diakine, IL-10 and IL-2.
  • FIG. 3 mouse serum cytokines obtained from diakine exposure in vivo.
  • FIG. 4 non-human primate serum cytokine levels obtained from diakine exposure in vivo.
  • FIG. 5 graphs measuring cytotoxicity of CD8+ T cell exposed to diakine and BiTE.
  • FIG. 6 graphs measuring IFNg and TNFa levels in CD8+ T cells exposed to varying concentrations of BiTE.
  • FIG. 7 schematic diagram of proposed IL-2 mediated CRS circuit in response to BiTE or CAR-T therapy.
  • FIG. 8 combination of a diakine comprising IL-10 and IL-2 which targets EGFR in combination with CD3xCD19 BiTE enhances CD8+ T cell directed tumor cell cytolysis.
  • FIG. 9 combination of a diakine (DK210 EGFR) with CD3xCD19 BiTE exhibits enhanced cytolytic effector molecules and controlled CRS
  • FIG. 10 intracellular FACS analysis of a diakine (DK210 EGFR) and CD3xCD19 BiTE treatment of PBMC cultures containing RajiGFP+ tumor cells
  • DETAILED DESCRIPTION
  • Exemplary aspects are described herein in the context of a using IL-10, IL-4, IL-12, IL-15, IL-7, or half-life extended versions thereof, or a diakine comprising at least one of IL-10 or IL-4 in combination with IFN-α, IL-2, IL-7, IL-12, IL-15, or IL-27 in a method of suppressing, inhibiting, reducing or preventing CRS associated with BiTE or CAR-T therapeutic modalities. Those of ordinary skill in the art will realize that the following description is illustrative only and is not intended to be in any way limiting. Other aspects will readily suggest themselves to those skilled in the art having the benefit of this disclosure. Reference will now be made in detail to implementations of the exemplary aspects as illustrated in the accompanying drawings. The same reference indicators will be used to the extent possible throughout the drawings and the following description to refer to the same or like items.
  • Although a number of methods and materials similar or equivalent to those described herein can be used in the practice of the various described embodiments, the preferred materials and methods are described herein.
  • Unless otherwise indicated, the embodiments described herein employ conventional methods and techniques of molecular biology, biochemistry, pharmacology, chemistry, and immunology, which are well known to a person skilled in the art. See, e.g., Sambrook, et al., Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.); Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C. Blackwell eds., Blackwell Scientific Publications); A. L. Lehninger, Biochemistry (Worth Publishers, Inc., current addition). Many of the general techniques for designing and fabricating diakines have been previously described in U.S. Application Publication No. 20220017587. This includes the types of IL-10 variants, including but not limited to human, mouse, CMV and/or EBV forms of IL-10, as well as the assays for testing the IL-10 variants, diakines, and other are known assay methods.
  • The following terms will be used to describe the various embodiments discussed herein, and are intended to be defined as indicated below.
  • As used herein in describing the various embodiments, the singular forms “a”, “an” and “the” include plural referents unless the content clearly dictates otherwise.
  • The term “about”, refers to a deviance of between 0.0001-5% from the indicated number or range of numbers. In one embodiment, the term “about”, refers to a deviance of between 1-10% from the indicated number or range of numbers. In one embodiment, the term “about”, refers to a deviance of up to 25% from the indicated number or range of numbers. In a more specific embodiment, the term “about” refers to a difference of 1-25% in terms of nucleotide sequence homology or amino acid sequence homology when compared to a wild-type sequence.
  • The term “agent” as it relates to the various interleukins (IL), such “IL-10 agent” or “IL-4 agent” and the like, are intended to be construed broadly and include, for example, human and non-human forms of the interleukin polypeptides, including homologs, variants (including muteins), fragments thereof, and fusion proteins, as well as polypeptides having, for example, a leader sequence (e.g., the signal peptide), and modified versions of the foregoing. The present disclosure also contemplates nucleic acid molecules encoding the foregoing, vectors and the like containing the nucleic acid molecules, and cells (e.g., transformed cells and host cells) that express the interleukin agents. The terms “variant,” “analog” and “mutein” refer to biologically active derivatives of the reference molecule, that retain a desired activity, such as, for example, anti-inflammatory activity. Generally, the terms “variant,” “variants,” “analog” and “mutein” as it relates to a polypeptide refers to a compound or compounds having a native polypeptide sequence and structure with one or more amino acid additions, substitutions (which may be conservative in nature), and/or deletions, relative to the native molecule. For example, the terms “IL-10 variant”, “variant IL-10,” “IL-10 variant molecule,” and grammatical variations and plural forms thereof are all intended to be equivalent terms that refer to a variant forms of IL-10 amino acid (or nucleic acid) sequence that differs from wild-type IL-10 form anywhere from 1-25% in sequence identity or homology. Thus, for example, an EBV IL-10 variant molecule is one that differs from wild-type EBV IL-10 by having one or more amino acid (or nucleotide sequence encoding the amino acid) additions, substitutions and/or deletions.
  • The term “fusion protein” refers to a combination or conjugation of two or more proteins or polypeptides that results in a novel arrangement of proteins that do not normally exist naturally. The fusion protein is a result of covalent linkages of the two or more proteins or polypeptides. The two or more proteins that make up the fusion protein may be arranged in any configuration from amino-terminal end (“NH2”) to carboxy-terminal end (“COOH”).
  • The term “homolog,” “homology,” “homologous” or “substantially homologous” refers to the percent identity between at least two polynucleotide sequences or at least two polypeptide sequences. Sequences are homologous to each other when the sequences exhibit at least about 50%, preferably at least about 75%, more preferably at least about 80%-85%, preferably at least about 90%, and most preferably at least about 95%-98% sequence identity over a defined length of the molecules.
  • The term “sequence identity” refers to an exact nucleotide-by-nucleotide or amino acid-by-amino acid correspondence. The sequence identity may range from 100% sequence identity to 50% sequence identity. A percent sequence identity can be determined using a variety of methods including but not limited to a direct comparison of the sequence information between two molecules (the reference sequence and a sequence with unknown percent identity to the reference sequence) by aligning the sequences, counting the exact number of matches between the two aligned sequences, dividing by the length of the reference sequence, and multiplying the result by 100. Readily available computer programs can be used to aid in the identification of percent identity.
  • The terms “subject,” “individual” or “patient” are used interchangeably herein and refer to a vertebrate, preferably a mammal. Mammals include, but are not limited to, murine, rodent, simian, human, farm animals, sport animals, and certain pets.
  • The term “administering” includes routes of administration which allow the active ingredient of the application to perform their intended function.
  • A “therapeutically effective amount” or “effective amount” as it relates to, for example, administering the IL variants, fusion proteins, dual cytokine fusion proteins, or diakine thereof described herein, refers to an amount sufficient to promote certain biological activities. These might include, for example, suppression of myeloid cell function, enhanced Kupffer cell activity, and/or lack of any effect on CD8+ T cells or enhanced CD8+ T-cell activity as well as blockade of mast cell upregulation of Fc receptor or prevention of degranulation or to promote or enhance to effects of combination therapeutics (e.g., CAR-T therapies) or to suppress the induction of cytokines from monocytes or macorphages. Thus, an “effective amount” will ameliorate or prevent a symptom or sign of the medical condition. Effective amount also means an amount sufficient to allow or facilitate diagnosis.
  • The term “treat,” “treating,” or “treatment” refers to a method of reducing the effects of a disease or condition. Treatment can also refer to a method of reducing the underlying cause of the disease or condition itself rather than just the symptoms. The treatment can be any reduction from native levels and can be, but is not limited to, the complete ablation of the disease, condition, or the symptoms of the disease or condition.
  • The term “half-life extended” as used in this application, refers to a protein that includes one of more additional moiety or moieties (such as proteins or PEGylation) that extends and/or enhances the circulation time within a subject anywhere in the range of 1-100 fold longer than a protein in the absence of the additional moiety or moieties. As it relates to IL, in one preferred embodiment, a half-life extended IL refers to an IL-10 or IL-4, IL-7, IL-12, or IL-15 conjugated to a scFv whereby the circulation time is extended in the range of 1-10 fold longer than the IL in the absence of the scFv. In another embodiment, a half-life extended version of an IL will adopt the configuration of formula I
  • Figure US20230340052A1-20231026-C00001
  • wherein
    • “Y” is any monomer from either a homodimeric or heterodimeric cytokine;
    • “X1” is a VL or VH region obtained from a first monoclonal antibody;
    • “X2” is a VH or VL region obtained from the first monoclonal antibody,
    wherein when X1 is a VL, X2 is a VH or when X1 is a VH, X2 is a VL and wherein the VH and VL together form a scFv.
  • The term “diakine” or “DK”, as used in this application, refers to a dual cytokine fusion protein comprising two monomers of a dimeric cytokine, which can be either a homodimer such as IL-10 or IL-10 variants, or a heterodimer such as IL-12 or IL-12 variants, that is fused together with a monomeric cytokine, such as IL-2, IL-4, IL-7, IL-15, IL-21, IL-28, IL-29, or with a dimeric cytokine, such as IL-12 or IL-10, whereby both of the dimeric cytokine and monomeric cytokine being fused onto a half-life extending antigen targeting domain. Representative diakines are described in detail in U.S. Pat. 11,292,822 (IL-10 based diakines) and co-pending U.S. Application 18/065,504 (dual dimeric cytokine based diakines), both of which are incorporated by reference in their entireties. In one embodiment, a diakine, which may be used in a method in combination with a BiTE or CAR-T, is represented by formula II
  • Figure US20230340052A1-20231026-C00002
  • wherein
    • “Y” is any monomer from either a homodimeric or heterodimeric cytokine;
    • “X1” is a VL or VH region obtained from a first monoclonal antibody;
    • “X2” is a VH or VL region obtained from the first monoclonal antibody,
    • wherein when X1 is a VL, X2 is a VH or when X1 is a VH, X2 is a VL and wherein the VH and VL together form a scFv;
    • “Z” is a second cytokine, wherein the second cytokine is any monomeric cytokine or another dimeric cytokine; and
    • “n” is an integer selected from 0-2.
  • In one embodiment, dimeric cytokine may include IFN-α, IL-2, IL-4, IL-7, IL-10, IL-12, IL-15, IL-21, and IL-27. In another embodiments, the monomeric cytokine may include IL-6, IL-4, IL-1, IL-2, IL-3, IL-5, IL-7, IL-8, IL-9, IL-15, IL-21 IL-26, IL-27, IL-28, IL-29, GM-CSF, G-CSF, interferons-α, -β, -γ, TGF-β, or tumor necrosis factors -α, -β, basic FGF, EGF, PDGF, IL-4, IL-11, or IL-13. The VH and VL of the diakine is a scFv and may be derived from any monoclonal antibody but is preferably derived from an antibody that is capable of targeting a specific antigen. The monoclonal antibody from which the scFv (as it applies to Formula I and II) may be derived is selected from EGFR; CD52; CD14; various immune check point targets, such as but not limited to PD-L1, PD-1, TIM3, BTLA, LAG3 or CTLA4; CD19; CD20; CD22; CD47; GD-2; VEGFR1; VEGFR2; HER2; PDGFR; EpCAM; ICAM (ICAM-1, -2, -3, -4, -5), VCAM, CD14, FAPα; 5T4; Trop2; EDB-FN; TGFβ Trap; MAdCAM, β7 integrin subunit; α4β7 integrin; α4 integrin SR-A1; SR-A3; SR-A4; SR-A5; SR-A6; SR-B; dSR-C1; SR-D1; SR-E1; SR-F1; SR-F2; SR-G; SR-H1; SR-H2; SR-I1; SR-J1; CD123; CD33; BCMA; PSA; PSMA; CEA; GPC3; BCMA; DLL3; MUC17; CLDN 18; gpA33; HIV or Ebola. In an embodiment, the scFv (as it applies to Formula I and II) is an engrafted scFv, whereby the VH and VL framework region is derived from a first antibody (such as an anti-ebola antibody) and the CDRs are obtained from a second antibody (such as but not limited to EGFR; CD52; CD14; various immune check point targets, such as but not limited to PD-L1, PD-1, TIM3, BTLA, LAG3 or CTLA4; CD19; CD20; CD22; CD47; GD-2; VEGFR1; VEGFR2; HER2; PDGFR; EpCAM; ICAM (ICAM-1, -2, -3, -4, -5), VCAM, CD14, FAPα; 5T4; Trop2; EDB-FN; TGFβ Trap; MAdCAM, β7 integrin subunit; α4β7 integrin; α4 integrin SR-A1; SR-A3; SR-A4; SR-A5; SR-A6; SR-B; dSR-C1; SR-D1; SR-E1; SR-F1; SR-F2; SR-G; SR-H1; SR-H2; SR-I1; SR-J1; CD123; CD33; BCMA; PSA; PSMA; CEA; GPC3; BCMA; DLL3; MUC17; CLDN 18; gpA33; HIV or Ebola). In one preferred embodiment, the scFv is derived from an anti-Ebola antibody where the VH and VL framework regions of an anti-ebola antibody are substituted or engrafted with 6 CDR regions from an antibody specific for EGFR; CD52; CD14; various immune check point targets, such as but not limited to PD-L1, PD-1, TIM3, BTLA, LAG3 or CTLA4; CD19; CD20; CD22; CD47; GD-2; VEGFR1; VEGFR2; HER2; PDGFR; EpCAM; ICAM (ICAM-1, -2, -3, -4, -5), VCAM, CD14, FAPα; 5T4; Trop2; EDB-FN; TGFβ Trap; MAdCAM, β7 integrin subunit; α4β7 integrin; α4 integrin SR-A1; SR-A3; SR-A4; SR-A5; SR-A6; SR-B; dSR-C1; SR-D1; SR-E1; SR-F1; SR-F2; SR-G; SR-H1; SR-H2; SR-I1; SR-J1; CD123; CD33; BCMA; PSA; PSMA; CEA; GPC3; BCMA; DLL3; MUC17; CLDN 18; gpA33; more preferably an anti-EGFR, anti-MAdCAM, anti-VEGFR1, anti-VEGFR2, anti-PDGFR, or anti-CD14, anti-CD19, anti-CD20, or anti-CD22.
  • In another embodiment, the diakine, which may be used in a method in combination with a BiTE or a CAR-T, is a diakine described in U.S. Pat. 11,292,822 (IL-10 based diakines) or co-pending U.S. Application 18/065,504 (dual dimeric cytokine based diakines), both of which are incorporated by reference in their entireties. In yet another embodiment, the diakine, which may be used in a method in combination with a BiTE or a CAR-T, is protein represented by formula III
  • Figure US20230340052A1-20231026-C00003
  • wherein
    • “IL-10” is a monomer;
    • “X1” is a VL or VH region from a first monoclonal antibody;
    • “X2” is a VH or VL region from the first monoclonal antibody;
    • wherein when X1 is a VL, X2 is a VH or when X1 is a VH, X2 is a VL;
    • wherein the first monoclonal antibody is an anti-ebola antibody or the scFv framework region obtained therefrom;
    • wherein the VL and VH from the anti-ebola antibody include 3 light chain CDRs and 3 heavy chain CDRs that are engrafted with 3 light chain CDRs and 3 heavy chain CDRs from a second monoclonal antibody;
    • “Z” is a cytokine other than IL-10;
    • “n” is an integer of 1; and
    wherein the following variables in Table 1 apply to formula III:
  • TABLE 1
    “IL-10” monomer “second monoclonal antibody” “Z”
    Human IL-10 (Seq ID No: 1) EGFR 2
    15
    7
    28
    29
    IFN-alpha
    21
    HER2 2
    15
    7
    28
    29
    IFN-alpha
    21
    VEGFR2 2
    15
    7
    28
    29
    IFN-alpha
    21
    PDGFR 2
    15
    7
    28
    29
    IFN-alpha
    21
    GPC3 2
    15
    7
    28
    29
    IFN-alpha
    21
    PD-L1 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD19 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD20 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD22 2
    15
    7
    28
    29
    IFN-alpha
    21
    PSMA 2
    15
    7
    28
    29
    IFN-alpha
    21
    CEA 2
    15
    7
    28
    29
    IFN-alpha
    21
    BCMA 2
    15
    7
    28
    29
    IFN-alpha
    21
    gpA33 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD33 2
    15
    7
    28
    29
    IFN-alpha
    21
    DLL3 2
    15
    7
    28
    29
    IFN-alpha
    21
    MUC17 2
    15
    7
    28
    29
    IFN-alpha
    21
    CLDN18 2
    15
    7
    28
    29
    IFN-alpha
    21
    GD2 2
    15
    7
    28
    29
    IFN-alpha
    21
    5T4 2
    15
    7
    28
    29
    IFN-alpha
    21
    HER3 2
    15
    7
    28
    29
    IFN-alpha
    21
    EBV IL-10 (Seq ID No: 5) EGFR 2
    15
    7
    28
    29
    IFN-alpha
    21
    HER2 2
    15
    7
    28
    29
    IFN-alpha
    21
    VEGFR2 2
    15
    7
    28
    29
    IFN-alpha
    21
    PDGFR 2
    15
    7
    28
    29
    IFN-alpha
    21
    GPC3 2
    15
    7
    28
    29
    IFN-alpha
    21
    PD-L1 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD19 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD20 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD22 2
    15
    7
    28
    29
    IFN-alpha
    21
    PSMA 2
    15
    7
    28
    29
    IFN-alpha
    21
    CEA 2
    15
    7
    28
    29
    IFN-alpha
    21
    BCMA 2
    15
    7
    28
    29
    IFN-alpha
    21
    gpA33 2
    15
    7
    28
    29
    IFN-alpha
    21
    CD33 2
    15
    7
    28
    29
    IFN-alpha
    21
    DLL3 2
    15
    7
    28
    29
    IFN-alpha
    21
    MUC17 2
    15
    7
    28
    29
    IFN-alpha
    21
    CLDN18 2
    15
    7
    28
    29
    IFN-alpha
    21
    GD2 2
    15
    7
    28
    29
    IFN-alpha
    21
    5T4 2
    15
    7
    28
    29
    IFN-alpha
    21
    HER3 2
    15
    7
    28
    29
    IFN-alpha
    21
  • In another embodiment, the diakine, which may be used in a method in combination with a BiTE or CAR-T, is a diakine described in co-pending U.S. Application 18/065,504 in Tables 2a-2d and 3a-3d, which is incorporated by reference in its entirety.
  • In another aspect, the protein or nucleic acid molecules encoding dual cytokine fusion protein or DK may be formulated as a pharmaceutical composition comprising a therapeutically effective amount of the dual cytokine fusion protein and a pharmaceutical carrier and/or pharmaceutically acceptable excipients. The pharmaceutical composition may be formulated with commonly used buffers, excipients, preservatives, stabilizers. The pharmaceutical compositions comprising the dual cytokine fusion protein is mixed with a pharmaceutically acceptable carrier or excipient. Various pharmaceutical carriers are known in the art and may be used in the pharmaceutical composition. For example, the carrier can be any compatible, non-toxic substance suitable for delivering the dual cytokine fusion protein compositions of the application to a patient. Examples of suitable carriers include normal saline, Ringer’s solution, dextrose solution, and Hank’s solution. Carriers may also include any poloxamers generally known to those of skill in the art, including, but not limited to, those having molecular weights of 2900 (L64), 3400 (P65), 4200 (P84), 4600 (P85), 11,400 (F88), 4950 (P103), 5900 (P104), 6500 (P105), 14,600 (F108), 5750 (P123), and 12,600 (F127). Carriers may also include emulsifiers, including, but not limited to, polysorbate 20, polysorbate 40, polysorbate 60, and polysorbate 80, to name a few. Non-aqueous carriers such as fixed oils and ethyl oleate may also be used. The carrier may also include additives such as substances that enhance isotonicity and chemical stability, e.g., buffers and preservatives, see, e.g., Remington’s Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary, Mack Publishing Company, Easton, Pa. (1984). Formulations of therapeutic and diagnostic agents may be prepared by mixing with physiologically acceptable carriers, excipients, or stabilizers in the form of lyophilized powders, slurries, aqueous solutions or suspensions, for example.
  • The pharmaceutical composition will be formulated for administration to a patient in a therapeutically effective amount sufficient to provide the desired therapeutic result. Preferably, such amount has minimal negative side effects. In one embodiment, the amount of dual cytokine fusion protein administered will be sufficient to treat or prevent inflammatory diseases or condition. In another embodiment, the amount of dual cytokine fusion protein administered will be sufficient to treat or prevent immune diseases or disorders. In still another embodiment, the amount of diakine or dual cytokine fusion protein administered will be sufficient to treat or prevent CRS mediated by BiTE or CAR-T therapy. The amount administered may vary from patient to patient and will need to be determined by considering the subject’s or patient’s disease or condition, the overall health of the patient, method of administration, the severity of side-effects, and the like.
  • An effective amount for a particular patient may vary depending on factors such as the condition being treated, the overall health of the patient, the method route and dose of administration and the severity of side effects. The appropriate dose administered to a patient is typically determined by a clinician using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment. Generally, the dose begins with an amount somewhat less than the optimum dose and it is increased by small increments thereafter until the desired or optimum effect is achieved relative to any negative side effects. Important diagnostic measures include those of symptoms of, e.g., the inflammation or level of inflammatory cytokines produced.
  • The method for determining the dosing of the presently described dual cytokine fusion protein will be substantially similar to that described in U.S. Pat. 10,858,412. Generally, the presently described dual cytokine fusion protein will have a dosing in the range of 0.01 mg/kg to 1 mg/kg, preferably 0.025 mg/kg to 0.5 mg/kg. The dual cytokine fusion protein may be administered daily, three times a week, twice a week, weekly, bimonthly, or monthly. An effective amount of therapeutic will impact the level of CRS inhibited caused by the BiTE or CAR-T therapy. In yet another embodiment, the diakine will be dosed at a concentration of 0.1 ng/mL to 200 ng/mL, preferably, 10 ng/mL to 100 ng/mL. Generally, the addition of a diakine will lower the required dose for a BiTE or CAR-T modality.
  • Compositions of the application can be administered orally or injected into the body. Formulations for oral use can also include compounds to further protect the IL or DK molecules from proteases in the gastrointestinal tract. Injections are usually intramuscular, subcutaneous, intradermal or intravenous. Alternatively, intra-articular injection or other routes could be used in appropriate circumstances. Parenterally administered dual cytokine fusion protein are preferably formulated in a unit dosage injectable form (solution, suspension, emulsion) in association with a pharmaceutical carrier and/or pharmaceutically acceptable excipients. In other embodiments, compositions of the application may be introduced into a patient’s body by implantable or injectable drug delivery system.
  • Those of skill in the art will recognize that adoptive cell therapy (such as adoptive T-cell therapy) is well known and practiced according to procedures previously described. See, e.g., U.S. Pat. No. 4,690,915. These methods may include autologous transfer (i.e., derived from the patient) or allogenic transfer (i.e. derived from another subject other than the patient to be treated).
  • The CAR-T or TCR-T cells are administered by methods known and conventionally practiced by those familiar with adaptive cell therapy. In one embodiment, the administration method includes, but is not limited to bolus infusion, intravenous or subcutaneous injections, intraocular injection, periocular injection, subretinal injection, intravitreal injection, trans-septal injection, subscleral injection, intrachoroidal injection, intracameral injection, subconjectval injection, subconjuntival injection, sub-Tenon’s injection, retrobulbar injection, peribulbar injection, or posterior juxtascleral delivery. In some embodiments, they are administered by parenteral, intrapulmonary, and intranasal, or intralesional or intrtumoral administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. In certain some embodiments, the recombinantly engineered CAR-T or TCR-T is administered as a single bolus administration, multiple bolus or continuous infusion. Generally known CAR-T therapies include idecabtagene vicleucel, lisocabtagene maraleucel, brexucabtagene autoleucel, tisagenlecleucel, or axicabtagene ciloleucel.
  • In one embodiment, the diakine and the CAR-T are administered in separate subsequent time periods, wherein, for example, the diakine (such as DK210vegfr2 or DK210EGFR or any of those referenced in Table 1, above) is administered prior to the administration of a recombinantly engineered CAR-T cell. In other embodiments, the diakine and the CAR-T are administered simultaneously. In other embodiments, the diakine is administered 1-3 days before the CAR-T therapy and then simultaneously administered along with the CAR-T, and/or 1-7 days following CAR-T administration. The diakine may be administered once a day or week, or 2-3 times a week in combination or conjunction with the CAR-T. In another aspect, the diakine is utilized in the expansion and/or thawing procedure of the CAR-T cells prior to administration. Upon reconstituting CAR-T cells from cryopreserved stock, the CAR-T are typically rested in the presence of CAR-T beneficial cytokines (e.g., low dose IL-2). In one aspect, the CAR-T cells may be primed or expanded from cryopreserved stocks in the presence of a diakine. In one aspect, the CAR-T is expanded or primed in the presence of 0.001 to 300 ng/mL of a diakine, more preferably 0.01 to 200 ng/mL of a diakine.
  • Likewise, the diakine and the BiTE are administered in separate subsequent time periods, wherein the diakine (e.g., DK210CD20 or DK210EGFR or DK210HER2 or DK210HER3) is administered 1-3 days before administering the BiTE (e.g., CD3xCD19 BiTE). In other embodiments, the diakine is administered 1-3 days before the BiTE and then simultaneously administered along with the BiTE, and/or 1-7 days following BiTE administration. The diakine may be administered once a day or week, or 2-3 times a week in combination or conjunction with the BiTE. A BiTE will generally follow the format of a bispecific antibody having an anti-CD3 and an anti-TAA fused together. In one embodiment, method combines an IL-10 or IL-4, or half-life extended versions thereof, or a diakine comprising IL-10 or IL-4 and IFN-α, IL-2, IL-7, IL-12, IL-15, IL-21 or IL-27, in conjunction with a BiTE, such as those that have bispecificity for CD3 with CD33, BCMA, CD19, CD20, CD22, PSMA, EGFR, DLL3, MUC17, CLDN18, CEA, HER2, HER3, EpCAM, gpA33, GPC3, GD2 5T4, VEGFR2, PDGFR, PDL1, or PD1 to name a few. In a preferred embodiment, the BiTE is an anti-CD3 and anti-CD19, anti-CD20, anti-HER2, anti-HER3, anti-PSMA, or anti-BCMA .
  • It is generally understood that CRS is associated by concomitant increases in serum IL-6, IL-1β, TNFα, IFNα/γ, IL-12 and IL-23. While these cytokines are generally associated with CRS, it is not known whether one, two or all of these cytokines working together to lead to the toxic side effects associated with CRS. The inventor believes that a potential root cause of CRS is the fact than many patients with severe CRS induced by BiTE or CAR-T treatment develop vascular leak syndrome. Vascular or capillary leak syndrome is also predominantly observed as the dose limiting and often lethal toxicity associated with high dose IL-2 therapy. In addition, the inventor has found that that treatment of human peripheral blood mononuclear cells (PBMC) or human whole blood cells with an increasing concentration of IL-2 leads to the induction of a panel of proinflammatory cytokines reminiscent of CRS. FIG. 1 .
  • Without being bound to any particular theory, the inventor believes that the similarities between the cytokines induced by treatment with IL-2 alone, and the apparent blockade of the induction of these secondary cytokines by a diakine comprising IL-10 (e.g., DK210 (EGFR)) or IL-4, IL-12, IL-15, IL-7, or any combination thereof will suppress or block IL-2 mediated CRS. Others have shown that the addition of tumor cells expressing specific tumor associated antigens (TAA) to PBMC with a titration range of an anti-TAA:anti-CD3 BiTE leads to the induction of a range of proinflammatory cytokines. (Fu, 2019). It also appears that BiTE stimulation temporally leads to the initial induction of TNFα, IL-2 and IL-4, followed by the other cytokines. (Brandl, 2007). The level of IFNy, IL-6, and TNFα induction from PBMCs in response to exposure to anti-CD3 in the presence of a diakine (DK210 EGFR), IL-10, and IL-2, suggest that the presence of DK210 (EGFR) prevents the induction of significant secondary proinflammatory cytokines associated with CRS, unlike the increasing concentration of IL-2. FIG. 2 . In addition, the fusion of IL-2 with the high affinity EBV IL-10 (internally termed DV07, Seq ID No: 5), which is known as DK210EGFR, prevents the induction of IL-2 mediated secondary cytokines both from PBMC and from anti-CD3 stimulated PBMC. FIG. 2 . Further investigation of whether coupling IL-10 to IL-2 indicates that the application of DK210(EGFR) to mice prevents the induction of peripheral cytokines induced by IL-2 alone. FIG. 3 . Furthermore, the treatment of non-human primates with DK210 (EGFR) similarly does not lead to significant inductions of peripheral plasma cytokines. FIG. 4 .
  • Moreover, pre-exposure of CD8+ T cells to a diakine (e.g., DK210 (EGFR)) dramatically enhances the cells subsequent capacity to engage BiTE’s and effect cytolysis of tumor cells. FIG. 5 . Furthermore, these cells appear to secrete similar levels of IFNy, but lower levels of TNFα compared to BiTE alone stimulated cells. FIG. 6 . Thus, the inventor believes that the molecular circuit responsible for BiTE or CAR-T mediated CRS is a cascade of pro-inflammatory cytokine release, that is first triggered by CD4+ T cell engagement with BiTE or CAR-T, which subsequently triggers IL-2 secretion and inducement of monocyte/macrophage to undergo additional secretion of proinflammatory cytokines. FIG. 7 from monocytes/macrophages.
  • Example 1: Diakine Enhances BiTE Mediated Tumor Cell Cytolysis
  • DK210 (EGFR) has been previously shown to prime CD8+ T cell for subsequent tumor cytolysis in vitro and in vivo. See, U.S. Pat. 11,292,822. Here, DK210 (EGFR), which is a representative example of a diakine, is shown to both enhance BiTE mediated tumor cell cytolysis as well as suppress BiTE mediated CRS.
  • Initially, DK210 (EGFR) is shown to enhance BiTE mediated tumor cell cytotoxicity. In this in vitro model, the combinatorial anti-tumor effects of DK210 (EGFR) with the CD19 BiTE is assessed in multiple rounds of exposure to target tumor cells.
  • CD8+ T cells are isolated from fresh donor Leukopaks via magnetic bead isolation per the manufacturer’s suggested protocol (Miltenyi). The isolated CD8+ T cells are plated at 2.5×106 cells/well and exposed for 2 days in various concentrations (0 or 100 ng/mL) of DK210 (EGFR) in AIMV. Following the 2 days of exposure to the various concentrations of DK210 (EGFR), the CD8+ T cells are harvested, counted, washed, and finally resuspended in the corresponding concentration of DK210 (EGFR). Concurrently, Raji cells, which constitutively express Green Florescent Protein (GFP), are counted, washed and resuspended in varying concentrations (0 or 0.1 ng/mL) of CD19 BiTE. The CD8+ cells (effector) and Raji-GFP cells (target) are then combined at a 10:1 effector to target ratio. The mixture of effector and target cells, which are exposed to CD19 BiTE alone, DK210 (EGFR) alone, or the combination of CD19 BiTE and DK210 (EGFR), were monitored over 5 days using an IncuCyte® S3 Live-Cell Analysis System (Essen Bioscience/Sartorius). In conjunction, additional plates were seeded with the same conditions as stated above to be used for subsequent and serial rounds of the cytotoxicity assay. Every 3 days media is aspirated from wells and fresh media, with appropriate concentrations of either DK210 (EGFR), CD19 BiTE, or the combination of the two, are added to the wells. Following the 5-day exposure, cells are harvested, counted, washed, and re-exposed to the similar conditions as stated previously. The percentage of (GFP) disappearance is measured as an indicator of cytotoxicity.
  • CD3xCD19 BiTE when combined with DK210 (EGFR) enhances tumor cell cytotoxicity. See, FIGS. 5 and 8 . CD3XCD19 BiTE and DK210 (EGFR) are tested both alone and in combination using normal, healthy human donor derived CD8+ T cells. FIG. 8 . Effector cells went through multiple rounds (5 rounds of serial cytolysis) of exposure to target tumor cells (Raji-GFP), and cytotoxicity was measured via the disappearance of GFP. In vitro treatment with the BiTE in combination with DK210 (EGFR) suggests that activation of T cells with DK210 (EGFR) enhances responses to BITE’s when assessing CD8+ T cell anti-RajiGFP+ responses.
  • Example 2: Diakine Reduces BiTE Mediated CRS
  • One of the current clinical challenges with BiTE’s is the significant induction of CRS (Zhou, 2021). Since diakine (e.g., DK210 (EGFR)) treatment of peripheral blood derived mononuclear cells (PBMC), tumor-bearing mice and non-human primates (see, FIGS. 1, 3, and 4 ) appears to prevent IL-2 mediated CRS, in vitro cultures comprising PBMC, RajiGFP+ cells, 0.1 ng/mL CD19 BiTE with or without 100 ng/mL DK210 (EGFR) are used to determine whether DK210 (EGFR) is capable of suppressing BiTE mediated CRS.
  • PBMCs are isolated from Leukopaks collected from healthy donors using the Ficoll density gradient method. Equal volumes of HBSS and donor samples were transferred to conical tubes, individually. Ficoll was slowly added to create a bottom layer then samples were centrifuged at 400 xg for 30 minutes at 25° C. PBMCs were harvested from the top layer then washed twice using Aim V media (300 xg, 8 minutes). The isolated PBMCs were plated at 2×106 cells/well in various concentrations (0 or 100 ng/mL) of DK210 EGFR in AIMV then incubated for two days (37° C., 5% CO2). Raji tumor cells were counted, washed and resuspended in varying concentrations of CD19 BiTE (0, or 0.1 ng/mL final), with or without DK210 (EGFR) (100 ng/mL) using AIMV and allowed to prime for two-days. The PBMCs, assuming 10% CD8+ T cells (effector), and Raji cells (target) were combined at a 10:1 effector to target ratio. After 24-hour incubation, supernatants are harvested and cytokine secretion was measured via multiplexed capture assay (MSD) and ELISA.
  • Illustrated in FIG. 9 , the data suggests that the presence of DK210 (EGFR) under these conditions significantly promotes tumor cell lysis (induction of IFN-gamma, Granzyme B, and Perforin) while limiting the induction of BiTE mediated CRS at a non-functional concentration of BiTE (0.1 ng/mL). Represented data is taken at 24 hrs., from exposure 1. Longitudinal data indicates a reduction of CRS over time in these conditions. FIG. 10 .
  • To better understand the cell types present in the PBMC + RajiGFP+ cultures that are affected by DK210 (EGFR), we assessed 48-hour cultures by intracellular fluorescence activated cell sorting (FACS). This analysis, suggests that the presence of DK210 (EGFR) polarizes both CD4+ and CD8+ T cells to predominantly express IFN-gamma, while reducing TNF-alpha and IL-2 production. MHC II positive cells, denoted as antigen-presenting cells (APC’s), also exhibit reduced TNF-alpha, IL-6 and IL-1-beta production, suggesting DK210 (EGFR) mediates pleiotropic cell type control of CRS associated with BiTE mediated T cell activation. FIG. 10 .
  • REFERENCES
  • BIBLIOGRAPHY Bosco, M. C. (2000). Il-2 Signaling in Human Monocytes Involves the Phosphorylation and Activation of p59hck1. Journal of Immunology.
  • Brandl, C. (2007). The effect of dexamethasone on polyclonal T cell activation and redirected taget cell lysis as induced by a CD19 CD3 bispecific single-chain antibody construct. Cancer Immunology Immunotherapy.
  • Chen, Y. (2021). Therapeutic Potential of TNFa and IL-1b Blockade for CRS/ICANS in CART Therapy via Ameliorating Endothelial Activation. Frontiers in Immunology.
  • Hosseini, I. (2020). Mitigating the risk of cytokine release syndrome in Phase I trial of CD20CD3 bispecific antibody mosunetuzumab in NHL impact of translational system modeling. Systems Biology and Applications.
  • Fu et al (1029). Therapeutic Bispecific T-Cell Engager Antibody Targeting the Transferrin Receptor. Frontiers in Immunology.
  • Liu, D. (2018). Cytokine release syndrome; grading, modeling, and new therapy. Journal of Hematology and Oncology.
  • Maude, S. L. (2014). Managing Cytokine Release Syndrome Associated with Novel T Cell-Engaging Therapies. Cancer Journal.
  • Musso, T. (1992). IL-2 Induces IL-6 Production in Human Monocytes. Journal of Immunology.
  • Norelli, M. (2018). Monocyte-derived IL-1 and IL-6 are differentially required for cytokine release syndrome and neurotoxicity due to CAR T cells. Nature Medicine.
  • Shimabukuro-Vornhagen, A. (2018). Cytokine release syndrome. Journal of ImmunoTherapy of Cancer. Strieter, R. M. (1989). Interleukin-2-induced Tumor Necrosis Factor-alpha Gene Expression in Human Alveolar Macrophages and Blood Monocytes. American Review Respiratory Disease.

Claims (20)

1. A method of reducing the severity of bispecific T cell engager (BiTE) or chimeric antigen receptor T cell (CAR-T) induced cytokine release syndrome (CRS) comprising administering to a patient in need thereof an amount of a composition comprising an interleukin 10 (IL-10) or an IL-10 agent, an interleukin 4 (IL-4) or an IL-4 agent, or combinations thereof.
2. The method according to claim 1, wherein the composition comprises a human IL-10 or viral IL-10, mutein, variants, fusion protein, and fragments thereof.
3. The method according to claim 2, wherein the human IL-10 is a sequence of SEQ ID No: 1.
4. The method according to claim 2, wherein viral IL-10 is an Epstein Barr viral (EBV) IL-10 of SEQ ID No: 5.
5. The method according to claim 1, wherein the IL-10 agent is a fusion protein comprising IL-10.
6. The method according to claim 1, wherein the IL-4 agent is a fusion protein comprising IL-4.
7. The method according to claim 6, wherein the fusion protein is a diakine comprising IL-10.
8. The method according to claim 7, wherein the fusion protein is a diakine comprising IL-4.
9. The method according to claim 7, wherein the diakine further comprises a scFv targeting domain that targets a receptor different from the BiTE or CAR-T.
10. The method according to claim 8, wherein the diakine further comprises a scFv targeting domain that targets a receptor different from the BiTE or CAR-T.
11. The method according to claim 10, wherein the scFv targets a receptor selected from EGFR, CD3, CD4, CD5, CD7, CD14, CD19, CD20, CD22, CD25, CD30, CD33, CD34, CD38, CD40, CD47, CD52, CD56, CD70, CD79B, CD117, CD123, CD138, CD147, BCMA, C-type lectin-like molecule-1 (CLL01), PD-L1, PD-1, TIM3, BTLA, latent membrane protein 1 (LMP-1), signal lymphocytic activation molecule F7 (SLAMF7), NY-ESO-1, transmembrane activator and CAML interactor (TACI), CS-1, CXCR4, NKG2D, B7-H3, LAG3, CTLA4, GD-2, VEGFR1, VEGFR2, HER2, HER3, PDGFR, EpCAM, mesothelin (MSO), PSCA, PSA, MUC1, Lewis-Y, GPC3, AXL, Claudin 18.2, GD2, CEA, ICAM-1, ICAM-2, ICAM-3, ICAM-4, ICAM-5, VCAM, FAPα, 5T4, Trop2, EDB-FN; TGFβ, Trap, MAdCAM, β7 integrin subunit, α4β7 integrin, α4 integrin, SR-A1, SR-A3, SR-A4, SR-A5, SR-A6, SR-B, dSR-C1, SR-D1, SR-E1, SR-F1, SR-F2, SR-G, SR-H1, SR-H2, SR-l1, SR-J1, HIV, or Ebola.
12. The method according to claim 11, wherein the scFv targets a receptor selected from EGFR, CD3, CD4, CD5, CD7, CD14, CD19, CD20, CD22, CD25, CD30, CD33, CD34, CD38, CD40, CD47, CD52, CD56, CD70, CD79B, CD117, CD123, CD138, CD147, BCMA, C-type lectin-like molecule-1 (CLL01), PD-L1, PD-1, TIM3, BTLA, latent membrane protein 1 (LMP-1), signal lymphocytic activation molecule F7 (SLAMF7), NY-ESO-1, transmembrane activator and CAML interactor (TACI), CS-1, CXCR4, NKG2D, B7-H3, LAG3, CTLA4, GD-2, VEGFR1, VEGFR2, HER2, HER3, PDGFR, EpCAM, mesothelin (MSO), PSCA, PSA, MUC1, Lewis-Y, GPC3, AXL, Claudin 18.2, GD2, CEA, ICAM-1, ICAM-2, ICAM-3, ICAM-4, ICAM-5, VCAM, FAPα, 5T4, Trop2, EDB-FN; TGFβ, Trap, MAdCAM, β7 integrin subunit, α4β7 integrin, α4 integrin, SR-A1, SR-A3, SR-A4, SR-A5, SR-A6, SR-B, dSR-C1, SR-D1, SR-E1, SR-F1, SR-F2, SR-G, SR-H1, SR-H2, SR-l1, SR-J1, HIV, or Ebola.
13. The method according to claim 9, wherein the diakine comprising IL-10 further comprises a second cytokine selected from IFN-α, IL-2, IL-4, IL-7, IL-12, IL-15, IL-21, IL-27.
14. The method according to claim 10, wherein the diakine comprising IL-4 further comprises a second cytokine selected from IFN-α, IL-2, IL-7, IL-10, IL-12, IL-15, IL-21, IL-27.
15. The method according to claim 1, wherein the BiTE or CAR-T therapy targets hematological or solid tumors, selected from CD19, CD20, or CD22.
16. The method according to claim 1, wherein the CAR-T targets are selected from TNFRSF17, IL3RA, SDC1, CD5, CD19, CD20, CD22, CD23, CD33, CD38, CD44, CD70, CD133, CD174, CD274, CD276, CEACAM6, GFRA1, ITGB6, MS4A1, TNFRSF8, NCAM1, ULBP1, ULBP2, IL1RAP, CEACAM5, MET, EGFR, EGFRvIII, ENPP1, FGFR4, EPCAM, EPHA2, ERBB2, GPC3, MSLN, Muc1, PDCD1, KDR, IL13RA2, FOLH1, FAP, CA9, FOLR1, L1CAM, ROR1, SLAMF7, GD2, PSCA, GPNMB, CSPG4, or TEM1.
17. The method according to claim 1, wherein the CAR-T therapy is idecabtagene vicleucel, lisocabtagene maraleucel, brexucabtagene autoleucel, tisagenlecleucel, or axicabtagene ciloleucel.
18. A method of inhibiting cytokine release syndrome (CRS) in a patient undergoing a bispecific T cell engager (BiTE) or chimeric antigen receptor T cell (CAR-T) therapy comprising administering to the patient in need thereof a composition comprising an interleukin 10 (IL-10) or an IL-10 agent.
19. A method of inhibiting cytokine release syndrome (CRS) in a patient undergoing a bispecific T cell engager (BiTE) or chimeric antigen receptor T cell (CAR-T) therapy comprising administering to the patient in need thereof a composition comprising an interleukin 4 (IL-4) or an IL-4 agent.
20. A method of inhibiting the induction of proinflammatory cytokines in a patient undergoing BiTE or CAR-T therapy comprising administering to the patient undergoing said therapy a composition comprising an interleukin 10 (IL-10) or an IL-10 agent or an interleukin 4 (IL-4) or an IL-4 agent.
US18/172,938 2022-02-22 2023-02-22 Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof Pending US20230340052A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/172,938 US20230340052A1 (en) 2022-02-22 2023-02-22 Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263312697P 2022-02-22 2022-02-22
US18/172,938 US20230340052A1 (en) 2022-02-22 2023-02-22 Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof

Publications (1)

Publication Number Publication Date
US20230340052A1 true US20230340052A1 (en) 2023-10-26

Family

ID=87766892

Family Applications (1)

Application Number Title Priority Date Filing Date
US18/172,938 Pending US20230340052A1 (en) 2022-02-22 2023-02-22 Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof

Country Status (2)

Country Link
US (1) US20230340052A1 (en)
WO (1) WO2023164503A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20240059748A1 (en) * 2020-07-20 2024-02-22 Deka Biosciences, Inc. Dual cytokine fusion proteins comprising il-10

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3302547A1 (en) * 2015-05-28 2018-04-11 Armo Biosciences, Inc. Pegylated interleukin-10 for use in treating cancer
TW202203917A (en) * 2020-04-13 2022-02-01 加拿大健康網路大學 Methods for treating cytokine release syndrome
JP2024517027A (en) * 2021-05-11 2024-04-18 シンガポール ヘルス サービシーズ ピーティーイー リミテッド Chimeric antigen receptor and modified cells containing the same

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20240059748A1 (en) * 2020-07-20 2024-02-22 Deka Biosciences, Inc. Dual cytokine fusion proteins comprising il-10
US20240076335A1 (en) * 2020-07-20 2024-03-07 Deka Biosciences, Inc. Dual cytokine fusion proteins comprising il-10
US20240076336A1 (en) * 2020-07-20 2024-03-07 Deka Biosciences, Inc. Dual cytokine fusion proteins comprising il-10

Also Published As

Publication number Publication date
WO2023164503A3 (en) 2023-10-12
WO2023164503A2 (en) 2023-08-31

Similar Documents

Publication Publication Date Title
Conlon et al. Cytokines in the treatment of cancer
Xiao et al. Biological drug and drug delivery-mediated immunotherapy
US20240091264A1 (en) Anti-b-cell maturation antigen chimeric antigen receptors with human domains
Zhang et al. A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity
Zheng et al. The use of supercytokines, immunocytokines, engager cytokines, and other synthetic cytokines in immunotherapy
JP7133241B2 (en) Fusion protein of IFN and anti-PD-L1 antibody and use thereof
EP3733716A1 (en) Fusion protein dimer using antibody fc region as backbone and use thereof
Curtsinger et al. Autocrine IFN-γ promotes naive CD8 T cell differentiation and synergizes with IFN-α to stimulate strong function
US20180147257A1 (en) Btn3a ectodomain proteins and methods of use
Zhang et al. Biological effects of IL-15 on immune cells and its potential for the treatment of cancer
US20210130430A1 (en) Activatable cytokine polypeptides and methods of use thereof
US20190300620A1 (en) Cd46-specific effector cells and uses thereof
US20220041724A1 (en) Plasmid constructs for treating cancer and methods of use
US20230340052A1 (en) Method of reducing bispecific t cell engager or chimeric antigen receptor t cell mediated cytokine release syndrome using interleukins-4, -10, or a fusion protein thereof
JP2022542461A (en) Treatment of immune evasion tumors
US20240059749A1 (en) Dual cytokine fusion proteins comprising il-10
JP2022525621A (en) Cytokine-based immune cells and their immunotherapeutic uses
Pieper et al. Radiation augments the local anti-tumor effect of in situ vaccine with CpG-oligodeoxynucleotides and anti-OX40 in immunologically cold tumor models
Chen et al. Sequential treatment with aT19 cells generates memory CAR-T cells and prolongs the lifespan of Raji-B-NDG mice
US20230287075A1 (en) Dual cytokine fusion proteins comprising multi-subunit cytokines
JP2023521174A (en) Binders and chimeric antigen receptors specific for interleukin-1 receptor accessory proteins
TWI734027B (en) Therapeutic combination and method for treating cancer
Park et al. Harnessing the power of IL-7 to boost T cell immunity in experimental and clinical immunotherapies
Yoon et al. Advanced T and Natural Killer Cell Therapy for Glioblastoma
US11970537B2 (en) Fusion protein dimer using antibody Fc region as backbone and use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: DEKA BIOSCIENCES, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MUMM, JOHN;REEL/FRAME:062780/0337

Effective date: 20230222

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED