WO2023141551A1 - Granulocyte-macrophage colony-stimulating factor-based dermatological pathology treatments - Google Patents

Granulocyte-macrophage colony-stimulating factor-based dermatological pathology treatments Download PDF

Info

Publication number
WO2023141551A1
WO2023141551A1 PCT/US2023/060966 US2023060966W WO2023141551A1 WO 2023141551 A1 WO2023141551 A1 WO 2023141551A1 US 2023060966 W US2023060966 W US 2023060966W WO 2023141551 A1 WO2023141551 A1 WO 2023141551A1
Authority
WO
WIPO (PCT)
Prior art keywords
skin
csf
patient
wound
activity
Prior art date
Application number
PCT/US2023/060966
Other languages
French (fr)
Inventor
Edwin ROCK
Ila JOSHI
Hillard LAZARUS
Original Assignee
Partner Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Partner Therapeutics, Inc. filed Critical Partner Therapeutics, Inc.
Publication of WO2023141551A1 publication Critical patent/WO2023141551A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • 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
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • 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
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • 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
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • 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
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/20Dermatological disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • This disclosure relates to, in part, treatment and/or mitigation of a dermatological pathology, as well as diagnostic, prognostic and patient selection methods.
  • Dermatological pathologies include skin diseases/disorders, nail diseases/disorders and hair/scalp diseases/disorders, comprising common rashes to severe and complex infections. These can occur due to a range of things, such as stress, infections, heat, allergens, system disorders and medications, with dermatitis being the most common skin disorder.
  • Common skin pathologies can include common inflammatory skin diseases such as eczema/dermatitis and psoriasis, as well as common cutaneous tumors such as seborrheic keratosis and actinic keratosis.
  • Common nail pathologies can include common nail diseases and abnormalities such as paronychia, nail fungus, lichen planus and pseudomonas nail infection among others.
  • Common hair/scalp pathologies can include seborrheic dermatitis, folliculitis, tinea capitis and contact dermatitis among others.
  • Atopic dermatitis can be a chronic condition that causes recurrent or relapsed inflamed skin, most frequently seen as patches on the face, neck, trunk or limbs. It can flare up sporadically.
  • Most dermatological diseases are not fully curable, current available therapies are based on the management of the symptoms associated with it.
  • Psoriasis is a common, multisystem inflammatory disease that manifests predominantly in the skin. Between 20% and 30% of patients with plaque psoriasis have moderate-to-severe forms that require systemic therapy. Although the pathogenesis of psoriasis is not fully understood, critical T-cell subsets and effector cytokines that mediate chronic inflammation observed in psoriasis have been identified. Further, mechanistic studies have demonstrated that therapeutic blockade of the cytokine interleukin (IL)-17A or its receptor can reverse the clinical, histologic, and molecular features of psoriasis. See Nestle FO et al. N Engl J Med. 2009.
  • T-cell activation is considered to be an important element in the pathogenesis of psoriasis, a human skin disease characterized by keratinocyte hyperproliferation, altered keratinocyte differentiation and inflammation of the dermis and epidermis. See Abernethy NJ et al. Immunol Cell Biol. 2000. 78(6): 596-602.
  • Psoriatic arthritis is a chronic, immune-mediated, inflammatory arthropathy that presents with inflammation of the joints and entheses, including those of the axial skeleton, and is associated with increased mortality from cardiovascular disease. Diagnosis is primarily based on clinical phenotype because of the diversity of the associated features, which can include skin, nail and hair/scalp disease, dactylitis, uveitis, and osteitis. Improved understanding of the pathogenesis of psoriatic arthritis has led to the development of a few effective biologies and small-molecular drugs targeting specific cytokines and signaling pathways, which has been shown to prevent disease progression and improve quality of life.
  • infections range from the uncomplicated impetigo to the potentially lethal necrotizing fasciitis, and can be caused by bacteria such as Staphylococcus aureus, Streptococcus pyogenes, Propionibacterium acnes, Erysipelothrix rhusiopathiae, coryneform bacteria; viruses including Herpes Simplex Virus, poxviruses, papillomaviruses (including, without limitation, Human Papilloma virus); fungi including dermatophytes such as Trichophyton, Microsporum, and Epidermophyton, and nondermatophytes such as yeast. See Aly R Medical Microbiology. 4 th edition. Chapter 98; Moubasher AH et al. J de Mycologie Medicale. 2017. 27: 166-179.
  • AK Actinic or solar keratosis
  • UV ultraviolet
  • 5- fluorouracil 5- fluorouracil
  • imiquimod and photodynamic therapy may have the best cosmetic outcomes.
  • AKs may be treated to improve appearance and relieve symptoms, but the keratinocytic dysplasia that gives rise to malignancy, and sometimes appears as an AK, may be what actually threatens patient health. See Siegel JA et al. Br J Dermatol. 2017. 177(2): 350-358.
  • a skin, nail or scalp wound is damage or disruption to its normal anatomical structure and function. This can range from a simple break in the epithelial integrity of the skin to a deeper wound, extending into subcutaneous tissue with damage to other structures such as tendons, muscles, vessels, nerves, parenchymal organs and even bone.
  • Wound healing is a normal feature of living animals and is a dynamic, interactive process which involves various types of cell and the extracellular matrix. The normal healing process can be described in terms of four programmed phases comprising 1 ) hemostasis, 2) inflammation, 3) proliferation and 4) remodeling. The speed and efficiency of the process of wound healing depends on various internal and external factors. See Velnar T et al. J Int Med Res. 2009. 37(5): 1528-42.
  • biomarker development and validation Since therapeutic response can vary on the basis of heterogeneous clinical and molecular phenotypes, a shift toward personalized or precision medicine approaches, including biomarker development and validation, has been thought to improve the management of many dermatological pathologies and skin diseases/disorders. Substantial progress in molecular immunology, coupled with an increased focus on translational research and personalized medicine, has resulted in a rapid expansion in the field of immune biomarkers in recent years. Such biomarkers might be used as an objective measure of normal versus pathogenic processes or indicator of pharmacological responses to therapeutic inventions. See Biomarkers Definitions Working Group. Clin Pharmacol Then 2001. 69(3): 89-95; Willis JCD and Lord GM. Nat Rev Immunol. 2015. 15:323-329; Renert-Yuval Y et al. J Allergy Clin Immunol. 2021. 147(4): 1174-1190.
  • CD86 Cluster of Differentiation-86
  • B7-2 Cluster of Differentiation-86
  • This dual role of CD86 can contribute to T cell activation and survival or T cell inhibition respectively.
  • PESI Psoriasis Area Severity Index
  • CD86 + monocytes located within psoriatic plaques were associated with enhanced epidermal proliferation. See Nguyes CTH et al. J Dermatol Sci. 2018. 90(2): 135-143. Further, targeting cutaneous dendritic cells with small interfering RNA (siRNA)-based CD86 therapy, CD86-siRNA, produced an amelioration in clinical manifestations of allergic skin diseases using a murine model. See Ritprajak P et al. Mol Then 2008. 16(7): 1323-30.
  • siRNA small interfering RNA
  • CD109 Cluster of Differentiation-109
  • a TGF-b co-receptor is a glycosyl phosphatidylinositol (GPI)-anchored protein, and is a co-receptor and negative regulator of TGF-[3 signaling.
  • CD109 has been shown to bind to the TGF-b1 isoform with high affinity, forms a complex with the TGF-b receptors and inhibits TGF-b signaling by increasing TGF-b receptor internalization and degradation.
  • CD109 can be released from the cell surface by cellular lipases such as phosphatidylinositol-specific phospholipase C (PI-PLC), and the soluble form of CD109 retains its ability to bind TGF- b1 and sequester it away from the TGF-b receptors.
  • PI-PLC phosphatidylinositol-specific phospholipase C
  • CD109 release from the cell surface in human keratinocytes has been postulated to induce molecular changes typically observed in psoriasis, potentially due to TGF-[3 receptor downregulation and decrease in TGF-[3 signaling. See Litvinov IV et al. Exp Dermatol. 2011. 20(8):627-32.
  • CD122 Cluster of Differentiation-122
  • IL-2Rb/IL-2RB interleukine-2 receptor subunit beta
  • IL-2Rb/IL-2RB interleukine-2 receptor subunit beta
  • T-cell activation is considered to be an important element in the pathogenesis of psoriasis, a human skin disease characterized by keratinocyte hyperproliferation, altered keratinocyte differentiation and inflammation of the dermis and epidermis.
  • psoriasis a human skin disease characterized by keratinocyte hyperproliferation, altered keratinocyte differentiation and inflammation of the dermis and epidermis.
  • fsn flaky skin
  • PPN peripheral lymph nodes
  • Granulocyte Macrophage - Colony Stimulating Factor is a hematological growth factor that regulates the production, migration, proliferation, differentiation and function of hematopoietic cells. It was first identified as being able to induce in vitro the proliferation and differentiation of bone marrow progenitors into granulocytes and macrophages. In response to inflammatory stimuli, GM-CSF is released by various cell types including T lymphocytes, macrophages, fibroblasts and endothelial cells. GM-CSF then activates and enhances the production and survival of neutrophils, eosinophils, and macrophages.
  • GM-CSF Native GM-CSF is usually produced near the site of action where it modulates in vitro proliferation, differentiation, and survival of hematopoietic progenitor cells, but is present in circulating blood in only picomolar concentrations (10’ 1 ° to 10’ 12 M).
  • GM-CSF has a wide range of functions across different tissues in its action on myeloid cells, and GM-CSF deletion/depletion approaches have indicated its potential as an important therapeutic target in several inflammatory and autoimmune disorders. See A Metcalf D. Immunol Cell Biology. 1987, 65:35-43; Gasson JC. Blood. 1991 , 77:1131 -1145; Shannon MF et al. Crit Rev Immunol.
  • GM- CSF granulocyte-macrophage colony-stimulating factor
  • rhu GM- CSF Recombinant human granulocyte-macrophage colony-stimulating factor
  • rhu GM- CSF has been approved by the FDA for the treatment of neutropenia, blood dyscrasias and malignancies like leukemia in combination with chemotherapies.
  • GM- CSF used for treatment of neutropenia and aplastic anemia following chemotherapy greatly reduces the risk of infection associated with bone marrow transplantation. Its utility in myeloid leukemia treatment and as a vaccine adjuvant is also well established. See Dorr RT. Clin Therapeutics. 1993. 15(1 ): 19-29; Armitage JO. Blood 1998, 92:4491 -4508; Kovacic JC et al. J Mol Cell Cardiol. 2007, 42:19-33; Jacobs PP et al. Microbial Cell Factories 2010, 9:93.
  • biomarkers can be used for the diagnosis, prognosis, or theranosis of dermatological pathologies and skin diseases/disorders. They can also be used to identify skin disorders and conditions that do not respond to monotherapy alone, and those that might benefit from combination therapies. Such combination therapies can potentially increase the percentage of patients who respond to treatments. Hence, there remains a need for new and more effective biomarkers and combination treatments of dermatological pathologies and skin diseases/disorders.
  • the present disclosure relates to a method for treating dermatological pathologies and skin diseases/disorders, comprising: administering an effective amount of a composition comprising GM-CSF to a patient in need thereof, wherein the patient is characterized by an increased expression of Cluster of Differentiation 86 (CD86).
  • CD86 Cluster of Differentiation 86
  • the present disclosure relates to a method for treating dermatological pathologies and skin diseases/disorders, comprising: administering an effective amount of a composition comprising GM-CSF to a patient in need thereof, wherein the patient is characterized by an increased expression of Cluster of Differentiation 109 (CD109).
  • CD109 Cluster of Differentiation 109
  • the present disclosure relates to a method for treating dermatological pathologies and skin diseases/disorders, comprising: administering an effective amount of a composition comprising GM-CSF to a patient in need thereof, wherein the patient is characterized by an increased expression of Cluster of Differentiation 122 (CD122).
  • CD122 Cluster of Differentiation 122
  • the present disclosure provides a method for treating dermatological pathologies and skin diseases/disorders, comprising: (a) identifying a patient undergoing or having undergone treatment with an agent for dermatological issues and presenting as failed, intolerant, resistant, or refractory to the treatment with a dermatological agent; (b) determining the presence, absence or amount of CD86 and/or CD109 and/or CD122 in a sample from the patient; and (c) administering an effective amount of a granulocytemacrophage colony-stimulating factor (GM-CSF) agent to a patient demonstrating an increased or high expression and/or activity of CD86 and/or CD109 and/or CD122 relative to a pre-treated and/or undiseased state.
  • GM-CSF granulocytemacrophage colony-stimulating factor
  • the present disclosure provides a method for selecting a patient for treatment with GM-CSF for a dermatological pathology and skin diseases based on the presence, absence or amount of CD86 and/or CD109 and/or CD122 CD86 and/or CD109 and/or CD122 in a sample from the patient.
  • FIG. 1 illustrates a graph depicting the kinetics of expression of CD86 induced by sargramostim (LEUKINE).
  • Monocytes and lymphocytes from a human donor were treated with sargramostim (LEUKINE) at various concentrations 0.33pM to 33.33pM.
  • Expression of CD86 was assessed on day 1 on both monocytes and lymphocytes.
  • the top curve is monocytes
  • the bottom curve is lymphocytes.
  • FIG. 2 illustrates graphs depicting the kinetics of expression of CD109 induced by sargramostim (LEUKINE).
  • Monocytes and lymphocytes from a human donor were treated with sargramostim (LEUKINE) at various concentrations 0.33pM to 33.33pM. Expression of CD109 was assessed on day 1 on both monocytes and lymphocytes.
  • the top curve is monocytes
  • the bottom curve is lymphocytes.
  • FIG. 3 illustrates graphs depicting the kinetics of expression of CD 122 induced by sargramostim (LEUKINE).
  • Monocytes and lymphocytes from a human donor were treated with sargramostim (LEUKINE) at various concentrations 0.33pM to 33.33pM.
  • Expression of CD122 was assessed on day 1 , day 2 and day 3 on both monocytes and lymphocytes.
  • the top curve is monocytes
  • the bottom curve is lymphocytes.
  • the present disclosure relates to, in part, to the use of GM-CSF as an effective treatment for specific dermatological pathologies and skin diseases/disorders, selected using CD86, CD109 and/or CD122 as a predictive marker of disease sequelae and responsiveness to current therapy, e.g. with a dermatological agent.
  • the present disclosure relates to improved treatments for dermatological pathologies and skin diseases/disorders in a patient that has failed, is intolerant, is resistant, or is refractory to the current treatment in use.
  • evaluation of the presence, absence, levels or activity of CD86, CD109 and/or CD122 informs or predicts the disease state in the and, without limitation, the administration of GM-CSF converts the patient that has failed, is intolerant, is resistant, or is refractory to current treatment(s) for the dermatological indication.
  • the GM-CSF modulates CD86, or cells expressing the same, to improve a patient’s treatment outcome.
  • the GM-CSF modulates CD109, or cells expressing the same, to improve a patient’s treatment outcome. In still another embodiment, the GM-CSF modulates CD122, or cells expressing the same, to improve a patient’s treatment outcome.
  • the disclosure provides methods for treating dermatological pathologies and skin diseases/disorders.
  • the dermatological pathology is a skin wound, skin condition or a skin disorder.
  • compositions of Immunomodulatory and/or Dermatological Agents are provided.
  • the additional immunomodulatory or dermatological agent is selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti-IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; non-steroidal o
  • the immunoregulatory agent is an antibody or antibody format which is selected from one or more of a monoclonal antibody, polyclonal antibody, antibody fragment, Fab, Fab', Fab'-SH, F(ab')2, Fv, single chain Fv, diabody, linear antibody, bispecific antibody, multispecific antibody, chimeric antibody, humanized antibody, human antibody, and fusion protein comprising the antigen-binding portion of an antibody.
  • compositions of GM-CSF Compositions of GM-CSF
  • GM-CSF used in the practice of the disclosure includes any pharmaceutically safe and effective GM-CSF, or any derivative thereof having the biological activity of GM-CSF.
  • the GM-CSF used in the practice of the present methods is rhu GM- CSF, such as sargramostim (LEUKINE).
  • Sargramostim is a biosynthetic, yeast-derived, recombinant human GM-CSF, having a single 127 amino acid glycoprotein that differs from endogenous human GM-CSF by having a leucine instead of a proline at position 23.
  • Other natural and synthetic GM-CSFs, and derivatives thereof having the biological activity of natural human GM-CSF may be equally useful in the practice of the disclosure.
  • the GM-CSF is produced or producible in bacteria, yeasts, plants, insect cells, and mammalian cells. In embodiments, the GM-CSF is produced or producible in Escherichia coli cells. In embodiments, the GM-CSF is produced or producible in yeast cells. In embodiments, the GM-CSF is produced or producible in Chinese hamster ovary cells (CHO). In embodiments, the GM-CSF is not produced in E. coli cells. In embodiments, the GM-CSF is produced in a cell that allows for glycosylation, e.g. yeast or CHO cells.
  • the GM-CSF has an amino acid sequence of SEQ ID NO: 1 , or a variant of at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98% identity thereto.
  • the GM-CSF has an amino acid sequence of SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO:4, or a variant of at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98% identity thereto.
  • the GM-CSF is one of, sargramostim, molgramostim, and regramostim.
  • the core of hGM-CSF consists of four helices that pack at angles.
  • Crystal structures and mutagenic analysis of rhGM-CSF (Rozwarski D A et al., Proteins 26:304-13, 1996) showed that, in addition to apolar side chains in the protein core, 10 buried hydrogen bonding residues involve intramolecular hydrogen bonding to main chain atoms that were better conserved than residues hydrogen bonding to other side chain atoms; 24 solvation sites were observed at equivalent positions in the two molecules in the asymmetric unit, and the strongest among these was located in clefts between secondary structural elements. Two surface clusters of hydrophobic side chains are located near the expected receptor binding regions.
  • the N-terminal helix of hGM-CSF governs high affinity binding to its receptor (Shanafelt A B et al. , EMBO J 10:4105-12, 1991 ) T ransduction of the biological effects of GM-CSF requires interaction with at least two cell surface receptor components, (one of which is shared with the cytokine IL-5).
  • the above study identified receptor binding determinants in GM-CSF by locating unique receptor binding domains on a series of human-mouse hybrid GM-CSF cytokines.
  • the interaction of GM-CSF with the shared subunit of their high affinity receptor complexes was governed by a very small part of the peptide chains. The presence of a few key residues in the N-terminal a-helix of was sufficient to confer specificity to the interaction.
  • the amino acid mutations are amino acid substitutions, and may include conservative and/or non-conservative substitutions.
  • “Conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved.
  • the 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1 ) hydrophobic: Met, Ala, Vai, Leu, lie; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gin; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe.
  • “conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide.
  • glycine and proline may be substituted for one another based on their ability to disrupt a-helices.
  • non-conservative substitutions are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1 ) to (6) shown above.
  • the substitutions may also include non-classical amino acids (e.g. selenocysteine, pyrrolysine, /V-formylmethionine [3-alanine, GABA and 5- Aminolevulinic acid, 4-aminobenzoic acid (PABA), D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2- amino butyric acid, y-Abu, s-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3- amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosme, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine,
  • non-classical amino acids
  • Modification of the amino acid sequences may be achieved using any known technique in the art e.g., site-directed mutagenesis or PCR based mutagenesis. Such techniques are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y., 1989 and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y., 1989. Without wishing to be bound by theory, the degree of glycosylation of biosynthetic GM-CSFs appears to influence half-life, distribution, and elimination. (Lieschke and Burgess, N. Engl. J. Med.
  • the present GM-CSF molecules are glycosylated.
  • the present methods relate to the utility of a predictive biomarkers to determine the use of GM-CSF in the treatment of dermatological pathologies and skin diseases/disorders.
  • the present disclosure relates to a method of treating a patient in need of therapy wherein the patient is characterized by having failed or being intolerant or refractory to a treatment with a dermatological agent.
  • an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring of a biomarker in a sample of the patient.
  • an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring a variety of patient parameters.
  • the patient sample may be analyzed using, e.g. immunohistochemical or immunofluorescence techniques may be used to evaluate the immune infiltrate, for example, immune subsets such as IL-17-producing CD4 + Th cells (Th17 cells), CD8 + T cells (cytotoxic T cells), and systemic or circulating intermediate monocytes.
  • polychromatic flow cytometry can be used to measure multiple surface and intracellular markers, allowing characterization of cell phenotype and activation state.
  • whole blood can be used to evaluate changes in cell count with therapy or changes in cytokine levels, for example TGF-b, IL-2, IL-12, IL-23 and/or TNFa.
  • cytokine levels for example TGF-b, IL-2, IL-12, IL-23 and/or TNFa.
  • deep sequencing techniques can be used to yield quantification of changes in individual cell clonotypes.
  • an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring the presence, absence, or amount of Cluster of Differentiation 86 (CD86) isotype in a sample of the patient.
  • CD86 Cluster of Differentiation 86
  • an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring the presence, absence, or amount of Cluster of Differentiation 109 (CD109) isotype in a sample of the patient.
  • CD109 Cluster of Differentiation 109
  • an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring the presence, absence, or amount of Cluster of Differentiation 122 (CD122) isotype in a sample of the patient.
  • CD122 Cluster of Differentiation 122
  • the present disclosure relates to a method for treating a dermatological pathology wherein CD86 is used as a biomarker for predicting or determining the need for treatment with GM-CSF.
  • the present disclosure relates to a method for treating a dermatological pathology wherein CD109 is used as a biomarker for predicting or determining the need for treatment with GM-CSF.
  • the present disclosure relates to a method for treating a dermatological pathology wherein CD122 is used as a biomarker for predicting or determining the need for treatment with GM-CSF.
  • the presence, absence or amount of CD86, CD109 and/or CD122 by detection of protein and/or nucleic acids in a sample of the patient.
  • the presence, absence or amount of CD86, CD109 and/or CD122 is determined by ELISA, immunohistochemical staining, western blotting, in-cell western, immunofluorescent staining, or fluorescent activating cell sorting (FACS) in a sample of the patient.
  • the method for determining the presence, absence or amount of CD86, CD109 and/or CD122 is a method of characterizing a patient or selecting a patient for the treatment comprising GM-CSF.
  • the method of determining CD86, CD109 and/or CD122 presence, absence, levels or activity for the purposes of patient selection employs a sample, the sample selected from a biopsy, tissue or body fluid.
  • the method of patient selection is undertaken using a sample of the patient, where the sample is selected from blood, skin sample or tissue sample, tissue biopsy, a formalin-fixed or paraffin- embedded tissue specimen, cytological sample, cultured cells, plasma, serum, pus, urine, perspiration, tears, mucus, sputum, saliva and/or other body fluids.
  • the present methods direct patient treatment decisions.
  • the method comprises the step of monitoring the expression and/or activity of CD86, CD109 and/or CD122 during the course of treatment.
  • the methods may detect a high or increased CD86, CD109 and/or CD122 expression or activity, and this is correlative with the patient having failed or being intolerant or refractory to a treatment with a dermatological agent.
  • this directs treatment of the patient with GM-CSF agents.
  • the patient with an increased or high expression or activity of CD86, CD 109 and/or CD122 receives, for example, a greater dose of GM-CSF, and/or additional dermatological therapies.
  • the GM-CSF agents potentiate treatment with a dermatological agent.
  • GM-CSF agents are used to modulate the patient’s immune system, e.g. by decreasing expression and/or activity of CD86, CD109 and/or CD122.
  • the present disclosure relates to a method for treating a dermatological pathology comprising: administering an effective amount of a composition GM-CSF to a patient in need thereof.
  • the present disclosure relates to a method for treating a dermatological pathology, comprising: administering an effective amount of a composition comprising GM-CSF in conjunction with a dermatological agent to a patient in need thereof, wherein the patient is characterized by the presence, absence or amount of CD86, CD109, and/or CD122 in a sample of the patient.
  • the present disclosure relates to methods for treating a dermatological pathology, comprising: (a) identifying a patient undergoing or having undergone treatment with an agent for dermatological issues and presenting as failed, intolerant, resistant, or refractory to the treatment with a dermatological agent; (b) determining the presence, absence or amount of CD86 and/or CD109 and/or CD122 in a sample from the patient; and (c) administering an effective amount of a granulocytemacrophage colony-stimulating factor (GM-CSF) agent to a patient demonstrating an increased or high expression and/or activity of CD86 and/or CD109 and/or CD122 relative to a pre-treated and/or undiseased state.
  • GM-CSF granulocytemacrophage colony-stimulating factor
  • the present disclosure relates to methods of treating a dermatological pathology, comprising: (a) selecting a patient having skin condition or skin disorders or skin wounds and one or more of (i) increased expression and/or activity of CD86 relative to a non-diseased state; (ii) increased expression and/or activity of CD109 relative to a non-diseased state; and/or (iii) increased expression and/or activity of CD122 relative to a non-diseased state and (b) administering an effective amount of a composition comprising GM-CSF to the patient.
  • the present disclosure relates to a method for treating a dermatological pathology, comprising: administering an effective amount of a composition comprising GM-CSF alone or in conjunction with a dermatological agent to a patient in need thereof, wherein the patient is characterized by as a partial responder or a nonresponder to a dermatological treatment.
  • the present disclosure relates to a method for treating cancer, comprising: administering an effective amount of a composition comprising GM-CSF alone or in conjunction with a dermatological agent to a patient in need thereof, wherein the patient is characterized by having failed or being intolerant or refractory to a treatment with a dermatological agent.
  • the method of treatment causes a decrease in the expression and/or activity of CD86. In some embodiments, the method of treatment causes a decrease in the expression and/or activity of CD109. In some embodiments, the method of treatment causes a decrease in the expression and/or activity of CD 122.
  • the method of treatment prevents, treats, and/or mitigates progression and/or development of the dermatological pathology in the patient.
  • the method of treatment improves the skin disorder, skin condition or skin wound in the patient.
  • the method of treatment improves skin renewal in the patient.
  • the method of treatment prevents or mitigates excessive skin cell reproduction in the patient.
  • the method of treatment reverses or prevents inflammation in the skin or systemically.
  • the method of treatment decreases or mitigates the activation of skin or systemic T lymphocytes.
  • the method of treatment modulates proliferation and/or activation of IL-17-producing CD4 + Th cells (Th17 cells) or CD8 + T cells (cytotoxic T cells).
  • the method decreases or mitigates the activation of skin or systemic monocytes.
  • the method of treatment modulates skin and/or systemic monocytes including circulating intermediate monocytes.
  • the method of treatment reverses or prevents excessive production and/or signaling of one or more inflammatory cytokines, such as TGF-b, IL-2, IL-12, IL-23 and/or TNFa.
  • one or more inflammatory cytokines such as TGF-b, IL-2, IL-12, IL-23 and/or TNFa.
  • the agent that stimulates the survival, proliferation and activation of neutrophils, macrophages and/or dendritic cells is administered at a time selected from (i) the same time as a dermatological agent; (ii) within about 1 hour, about 2 hours, about 4 hours, about 8 hours, about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 72 hours, or about 96 hours or about 1 week or about 2 weeks following administration of said dermatological agent; (iii) at least about 1 hour, about 2 hours, about 4 hours, about 8 hours, about 12 hours, about 24 hours, about 48 hours, about 36 hours, about 72 hours, or about 96 hours, or about 1 week or about 2 weeks prior to administration of the dermatological agent; and/or (iv) after at least an about 10%, about 20%, about 30%, about 40% or about 50% decrease in expression of an extracellular marker such as CD86, CD109 and/or CD122.
  • an extracellular marker such as CD86, CD109 and/or CD122.
  • the skin disorder selected from psoriasis, plaque psoriasis, eczema, rosacea, ichthyosis, vitiligo, hives, seborrheic dermatitis, cold sores, blisters, actinic keratosis, carbuncle, latex allergy, cellulitis, lupus rash, contact dermatitis, atopic dermatitis, wart, ringworm, pityriasis rosea, melasma, impetigo, infantile hemangiomas, actinic keratosis, flaky skin, damaged skin and/or chronic idiopathic urticaria (CIU).
  • CUA chronic idiopathic urticaria
  • the autoimmune skin disorder is selected from psoriasis, vitiligo, cutaneous lupus erythematosus, alopecia areata, and/or frontal fibrosing alopecia.
  • the skin wound comprising an ulcer, diabetes-related wound, bum, sunburn, chemical warfare agent (e.g.
  • Sulphur Mustard gas and/or chlorine gas skin burn or blisters, aging skin wound, corneal ulceration wound, inflammatory gastrointestinal wound, bowel inflammatory disease wound, Crohn's disease wound, ulcerative due to colitis, hemorrhoids, epidermolysis bullosa wound, skin blistering wound, psoriasis wound, animal skin wound, animal diabetes wound, retinopathy wound, oral wound (mucositis), vaginal mucositis, a wound due to periodontal disease, a laceration, a wound by surgical resection, and/or a wound after surgical adhesion.
  • the patient is afflicted with one or more of breakouts, red and painful fluid-filled blisters, itchy and raised welts, thick and scaly or crusty skin patches, red and painful lumps under the skin, itchy and inflamed wheals, yellow or white scaly patches, scaly and silvery well-defined patches, disc-shaped scaly rash, angioedema, skin patches with loss of melanin and/or a rash with raised borders.
  • the dermatological pathology is a skin disease or infection.
  • the skin disease or infection is selected from erysipelas, staphylococcal scalded skin syndrome, folliculitis, erysipeloid, pitted keratolysis, erythrasma, trichomycosis, trichosporonosis (including, without limitation, hepatic or pulmonary trichosporonosis, intertrigo, acute infectious eczematoid dermatitis, pseudofolliculitis, toe web infection, skin tuberculosis, mycobacterium marinum skin disease, mycobacterium ulcerans skin disease, actinomycetoma, actinomycosis, various viral skin infections, dermatophytoses, onychomycoses, chronic non-healing wounds, warts (including, without limitation, genital warts, perianal warts, vaginal warts, and/or various verruc
  • the method further comprises administering one or more additional therapeutic agents, selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti-IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; non-steroidal
  • compositions described herein can possess a sufficiently basic functional group, which can react with an inorganic or organic acid, or a carboxyl group, which can react with an inorganic or organic base, to form a pharmaceutically acceptable salt.
  • a pharmaceutically acceptable acid addition salt is formed from a pharmaceutically acceptable acid, as is well known in the art.
  • Such salts include the pharmaceutically acceptable salts listed in, for example, Journal of Pharmaceutical Science, 66, 2-19 (1977) and The Handbook of Pharmaceutical Salts; Properties, Selection, and Use. P. H. Stahl and C. G. Wermuth (eds.), Verlag, Zurich (Switzerland) 2002, which are hereby incorporated by reference in their entirety.
  • salts include, by way of non-limiting example, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, pamoate, phenylacetate, trifluoroacetate, acrylate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate,
  • Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxysubstituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N- methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-lower alkylamines), such as mono-; bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert- butylamine, or tri
  • compositions described herein are in the form of a pharmaceutically acceptable salt.
  • compositions comprising the compositions, e.g. GM-CSF and/or an additional therapeutic agent, e.g. the therapeutic agent described herein, and a pharmaceutically acceptable carrier or excipient.
  • the additional therapeutic agent comprises selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti- IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; nonsteroidal ointments including cri
  • compositions described herein can be administered to a patient as a component of a composition that comprises a pharmaceutically acceptable carrier or vehicle.
  • Such compositions can optionally comprise a suitable amount of a pharmaceutically acceptable excipient so as to provide the form for proper administration.
  • pharmaceutical excipients can be liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • the pharmaceutical excipients can be, for example, saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like.
  • auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used.
  • the pharmaceutically acceptable excipients are sterile when administered to a patient. Water is a useful excipient when any agent described herein is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid excipients, specifically for injectable solutions.
  • suitable pharmaceutical excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Any agent described herein, if desired, can also comprise minor amounts of wetting or emulsifying agents, or pH buffering agents. Other examples of suitable pharmaceutical excipients are described in Remington’s Pharmaceutical Sciences 1447-1676 (Alfonso R. Gennaro eds., 19th ed. 1995), incorporated herein by reference.
  • the present disclosure includes the described pharmaceutical compositions (and/or additional therapeutic agents) in various formulations.
  • Any inventive pharmaceutical composition (and/or additional therapeutic agents) described herein can take the form of solutions, suspensions, emulsion, drops, tablets, pills, pellets, capsules, capsules containing liquids, gelatin capsules, powders, sustained-release formulations, suppositories, emulsions, aerosols, sprays, suspensions, lyophilized powder, frozen suspension, desiccated powder, or any other form suitable for use.
  • the composition is in the form of a capsule.
  • the composition is in the form of a tablet.
  • the pharmaceutical composition is formulated in the form of a soft-gel capsule.
  • the pharmaceutical composition is formulated in the form of a gelatin capsule. In yet another embodiment, the pharmaceutical composition is formulated as a liquid.
  • the inventive pharmaceutical compositions can also include a solubilizing agent. Also, the agents can be delivered with a suitable vehicle or delivery device as known in the art. Combination therapies outlined herein can be co-delivered in a single delivery vehicle or delivery device.
  • compositions comprising the inventive pharmaceutical compositions (and/or additional therapeutic agents) of the present disclosure may conveniently be presented in unit dosage forms and may be prepared by any of the methods well known in the art of pharmacy. Such methods generally include the step of bringing the therapeutic agents into association with a carrier, which constitutes one or more accessory ingredients. Typically, the formulations are prepared by uniformly and intimately bringing the therapeutic agent into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into dosage forms of the desired formulation (e.g., wet or dry granulation, powder blends, etc., followed by tableting using conventional methods known in the art).
  • a carrier which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing the therapeutic agent into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into dosage forms of the desired formulation (e.g., wet or dry granulation, powder blends, etc., followed by tablet
  • any pharmaceutical compositions (and/or additional therapeutic agents) described herein is formulated in accordance with routine procedures as a composition adapted for a mode of administration described herein.
  • Routes of administration include, for example: topical, oral, intradermal, transdermal, subcutaneous, intramuscular, intraperitoneal, intravenous, intranasal, epidural, sublingual, intranasal, intracerebral, intravaginal, rectal, or by inhalation.
  • Administration can be local or systemic.
  • the administering is by topical route.
  • the administration is by dermal route.
  • the mode of administration can be left to the discretion of the practitioner, and depends in-part upon the site of the medical condition. In most instances, administration results in the release of any agent described herein onto or into the affected site.
  • the GM-CSF (and/or additional therapeutic agents) is administered via a topical route or dermal route.
  • the GM-CSF (and/or additional therapeutic agents) is administered to the skin.
  • the GM-CSF (and/or additional therapeutic agents) is administered via a cream, gel, ointment, lotion, or spray.
  • the GM-CSF (and/or additional therapeutic agents) is administered by topical or dermal route.
  • topical or dermal route of sargramostim produces low systemic exposure in the patient.
  • compositions (and/or additional therapeutic agents) described herein are formulated in accordance with routine procedures as a composition adapted for topical or dermal administration.
  • Compositions for topical delivery can be in the form of a cream, gel, ointment, lotion, spray, aqueous or oily suspensions, powders, or emulsions, for example.
  • Increased skin permeability and penetration may be achieved by non-invasive methods, for example, with the use of any nanocarriers combined with any pharmaceutical composition (and/or additional therapeutic agents) described herein.
  • the skin can act as a reservoir, and can be used to deliver the compositions (and/or additional therapeutic agents) described herein for more extended periods in a sustained manner.
  • compositions for oral delivery can be in the form of tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions, capsules, syrups, or elixirs, for example.
  • Orally administered compositions can comprise one or more agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of Wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation.
  • Dosage forms suitable for parenteral administration include, for example, solutions, suspensions, dispersions, emulsions, and the like. They may also be manufactured in the form of sterile solid compositions (e.g. lyophilized composition), which can be dissolved or suspended in sterile injectable medium immediately before use. They may contain, for example, suspending or dispersing agents known in the art.
  • Formulation components suitable for parenteral administration include a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as EDTA; buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl paraben
  • antioxidants such as ascorbic acid or sodium bisulfite
  • chelating agents such as EDTA
  • buffers such as acetates, citrates or phosphates
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS).
  • the carrier should be stable under the conditions of manufacture and storage, and should be preserved against microorganisms.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol), and suitable mixtures thereof.
  • compositions (and/or additional therapeutic agents) described herein can be administered by controlled-release or sustained-release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591 ,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; and 5,733,556, each of which is incorporated herein by reference in its entirety.
  • Such dosage forms can be useful for providing controlled- or sustained-release of one or more active ingredients using, for example, hydropropyl cellulose, hydropropylmethyl cellulose, polyvinylpyrrolidone, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled- or sustained-release formulations known to those skilled in the art, including those described herein, can be readily selected for use with the active ingredients of the agents described herein.
  • the disclosure thus provides single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled- or sustained-release.
  • Controlled- or sustained-release of an active ingredient can be stimulated by various conditions, including but not limited to, changes in pH, changes in temperature, stimulation by an appropriate wavelength of light, concentration or availability of enzymes, concentration or availability of water, or other physiological conditions or compounds.
  • a controlled-release system can be placed in proximity of the target area to be treated, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Other controlled-release systems discussed in the review by Langer, 1990, Science 249:1527-1533 may be used.
  • compositions preferably are sterile. Sterilization can be accomplished, for example, by filtration through sterile filtration membranes. Where the composition is lyophilized, filter sterilization can be conducted prior to or following lyophilization and reconstitution.
  • the actual dose of the composition to be administered according to the present disclosure will vary according to the particular dosage form, and the mode of administration. Many factors that may modify the action of the composition (e.g., body weight, gender, diet, time of administration, route of administration, rate of excretion, condition of the patient, drug combinations, genetic disposition and reaction sensitivities) can be taken into account by those skilled in the art. Administration can be carried out continuously or in one or more discrete doses within the maximum tolerated dose. Optimal administration rates for a given set of conditions can be ascertained by those skilled in the art using conventional dosage administration tests.
  • the expression of the various biomarkers provides guidance for the administration of GM-CSF at a specific dose, using a specific route, and/or determining the frequency and duration of therapy.
  • the GM-CSF is administered at a total dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg. In embodiments, the GM-CSF is administered at a total dose of about 250 pg.
  • the GM-CSF is administered at a dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg.
  • the GM-CSF is administered twice daily.
  • the GM-CSF is sargramostim, administered at a dose of about 125 pg, twice daily.
  • the pharmaceutical composition of the present disclosure is co-administered in conjunction with additional agent(s), for example an immunomodulatory or dermatological agent, such as a checkpoint inhibitor. Coadministration can be simultaneous or sequential.
  • additional agent(s) for example an immunomodulatory or dermatological agent, such as a checkpoint inhibitor.
  • Coadministration can be simultaneous or sequential.
  • the additional immunomodulatory or dermatological agent and the GM-CSF of the present disclosure are administered to a patient simultaneously.
  • the term “simultaneously” as used herein, means that the immunomodulatory or dermatological agent and the GM-CSF are administered with a time separation of no more than about 60 minutes, such as no more than about 30 minutes, no more than about 20 minutes, no more than about 10 minutes, no more than about 5 minutes, or no more than about 1 minute.
  • Administration of the immunomodulatory or dermatological agent and the GM-CSF can be by simultaneous administration of a single formulation (e.g., a formulation comprising the additional therapeutic agent and the GM-CSF composition) or of separate formulations (e.g., a first formulation including the immunomodulatory or dermatological agent and a second formulation including the GM-CSF composition).
  • a single formulation e.g., a formulation comprising the additional therapeutic agent and the GM-CSF composition
  • separate formulations e.g., a first formulation including the immunomodulatory or dermatological agent and a second formulation including the GM-CSF composition.
  • Co-administration does not require the therapeutic agents to be administered simultaneously, if the timing of their administration is such that the pharmacological activities of the immunomodulatory or dermatological agent and the GM- CSF overlap in time, thereby exerting a combined therapeutic effect.
  • the immunomodulatory or dermatological agent and the targeting moiety, the GM-CSF composition can be administered sequentially.
  • the term “sequentially” as used herein means that the immunomodulatory or dermatological agent and the GM-CSF are administered with a time separation of more than about 60 minutes.
  • the time between the sequential administration of the immunomodulatory or dermatological agent and the GM-CSF can be more than about 60 minutes, more than about 2 hours, more than about 5 hours, more than about 10 hours, more than about 1 day, more than about 2 days, more than about 3 days, more than about 1 week apart, more than about 2 weeks apart, or more than about one month apart.
  • the optimal administration times will depend on the rates of metabolism, excretion, and/or the pharmacodynamic activity of the additional therapeutic agent and the GM-CSF being administered. Either the immunomodulatory or dermatological agent or the GM-CSF composition may be administered first.
  • Co-administration also does not require the therapeutic agents to be administered to the patient by the same route of administration. Rather, each therapeutic agent can be administered by any appropriate route, for example, parenterally or non- parenterally.
  • the GM-CSF described herein acts synergistically when co-administered with the immunomodulatory or dermatological agent.
  • the targeting moiety, the GM-CSF composition and the immunomodulatory or dermatological agent may be administered at doses that are lower than the doses employed when the agents are used in the context of monotherapy.
  • the sample is selected from a biopsy, a tissue and/or a body fluid.
  • the sample is selected from blood, skin sample or tissue sample, tissue biopsy, a formalin-fixed or paraffin- embedded tissue specimen, cytological sample, cultured cells, plasma, serum, pus, urine, perspiration, tears, mucus, sputum, saliva and/or other body fluids.
  • SEQ ID NO: 1 is wild type GM-CSF:
  • SEQ ID NO: 2 is sargramostim:
  • SEQ ID NO: 3 is molgramostim: [00116] APARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVISEMFDL
  • SEQ ID NO: 4 is regramostim:
  • an “effective amount,” when used in connection with an agent effective for the treatment of a coronavirus infection is an amount that is effective for treating or mitigating a coronavirus infection.
  • a,” “an,” or “the” can mean one or more than one.
  • the term “about” when used in connection with a referenced numeric indication means the referenced numeric indication plus or minus up to 10% of that referenced numeric indication. For example, the language “about 50” covers the range of 45 to 55.
  • compositional percentages are by weight of the total composition, unless otherwise specified.
  • the word “include,” and its variants is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the materials, compositions, devices, and methods of this technology.
  • the terms “can” and “may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.
  • LUKINE sargramostim

Abstract

The present disclosure relates to the treatment of dermatological pathologies and skin/nail/scalp diseases/disorders/infections with granulocyte-macrophage colony stimulating factor.

Description

GRANULOCYTE-MACROPHAGE COLONY-STIMULATING FACTOR-BASED
DERMATOLOGICAL PATHOLOGY TREATMENTS
FIELD
[0001] This disclosure relates to, in part, treatment and/or mitigation of a dermatological pathology, as well as diagnostic, prognostic and patient selection methods.
CROSS-REFERENCE TO RELATED APPLICATION
[0002] This application claims the benefit of U.S. Provisional Application Nos. 63/301 ,707, filed January 21 , 2022; 63/347,766, filed June 1 , 2022; and 63/413,772, filed October 6, 2022, the entire contents of which are hereby incorporated by reference in their entirety.
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY
[0003] The instant application contains a sequence listing, which has been submitted in XML format via EFS-Web. The contents of the XML copy named “PNR-007PC/127114- 5007 Sequence Listing”; Date created: January 19, 2023; File size: 8,192 bytes in size, the contents of which are incorporated herein by reference in their entirety.
BACKGROUND
[0004] Dermatological pathologies include skin diseases/disorders, nail diseases/disorders and hair/scalp diseases/disorders, comprising common rashes to severe and complex infections. These can occur due to a range of things, such as stress, infections, heat, allergens, system disorders and medications, with dermatitis being the most common skin disorder. Common skin pathologies can include common inflammatory skin diseases such as eczema/dermatitis and psoriasis, as well as common cutaneous tumors such as seborrheic keratosis and actinic keratosis. Common nail pathologies can include common nail diseases and abnormalities such as paronychia, nail fungus, lichen planus and pseudomonas nail infection among others. Common hair/scalp pathologies can include seborrheic dermatitis, folliculitis, tinea capitis and contact dermatitis among others. [0005] Atopic dermatitis can be a chronic condition that causes recurrent or relapsed inflamed skin, most frequently seen as patches on the face, neck, trunk or limbs. It can flare up sporadically. Most dermatological diseases are not fully curable, current available therapies are based on the management of the symptoms associated with it.
[0006] Psoriasis is a common, multisystem inflammatory disease that manifests predominantly in the skin. Between 20% and 30% of patients with plaque psoriasis have moderate-to-severe forms that require systemic therapy. Although the pathogenesis of psoriasis is not fully understood, critical T-cell subsets and effector cytokines that mediate chronic inflammation observed in psoriasis have been identified. Further, mechanistic studies have demonstrated that therapeutic blockade of the cytokine interleukin (IL)-17A or its receptor can reverse the clinical, histologic, and molecular features of psoriasis. See Nestle FO et al. N Engl J Med. 2009. 361 : 496-509; Krueger JG et al. J Allergy Clin Immunol. 2012. 130: 145-154; Papp KA et al. N Engl J Med. 2012. 366: 1181-1189; Papp KA et al. J Invest Dermatol. 2012. 132: 2466-2469; Lowes MA et al. Annu Rev Immunol. 2014. 32: 227-255; Lonnberg AS et al. Clin Cosmet Investig Dermatol. 2014. 7: 251 -259; Blauvelt A. Expert Opin Drug Saf. 2016 Oct; 15(10): 1413-20. T-cell activation is considered to be an important element in the pathogenesis of psoriasis, a human skin disease characterized by keratinocyte hyperproliferation, altered keratinocyte differentiation and inflammation of the dermis and epidermis. See Abernethy NJ et al. Immunol Cell Biol. 2000. 78(6): 596-602.
[0007] Psoriatic arthritis is a chronic, immune-mediated, inflammatory arthropathy that presents with inflammation of the joints and entheses, including those of the axial skeleton, and is associated with increased mortality from cardiovascular disease. Diagnosis is primarily based on clinical phenotype because of the diversity of the associated features, which can include skin, nail and hair/scalp disease, dactylitis, uveitis, and osteitis. Improved understanding of the pathogenesis of psoriatic arthritis has led to the development of a few effective biologies and small-molecular drugs targeting specific cytokines and signaling pathways, which has been shown to prevent disease progression and improve quality of life. However, at least 40% of patients with psoriatic arthritis have only a partial response or fail to respond to such treatments. See Shah K et al. RMD Open. 2017. 3:e000588; Veale DJ and Fearon U. Lancet. 2018. 391 (10136): 2273-2284. [0008] Skin diseases/infections encompass a variety of conditions. Skin infections can be organized by their underlying etiology: bacterial, fungal, viral or parasitic causes. These infections range from the uncomplicated impetigo to the potentially lethal necrotizing fasciitis, and can be caused by bacteria such as Staphylococcus aureus, Streptococcus pyogenes, Propionibacterium acnes, Erysipelothrix rhusiopathiae, coryneform bacteria; viruses including Herpes Simplex Virus, poxviruses, papillomaviruses (including, without limitation, Human Papilloma virus); fungi including dermatophytes such as Trichophyton, Microsporum, and Epidermophyton, and nondermatophytes such as yeast. See Aly R Medical Microbiology. 4th edition. Chapter 98; Moubasher AH et al. J de Mycologie Medicale. 2017. 27: 166-179.
[0009]Actinic or solar keratosis (AK) is the most common precancer lesion that forms on skin damaged by chronic exposure to ultraviolet (UV) rays from the sun and/or indoor tanning. There is no current agreement on the most efficacious treatment, but 5- fluorouracil (5-Fll) has been shown to both prevent and treat AKs, and imiquimod and photodynamic therapy may have the best cosmetic outcomes. AKs may be treated to improve appearance and relieve symptoms, but the keratinocytic dysplasia that gives rise to malignancy, and sometimes appears as an AK, may be what actually threatens patient health. See Siegel JA et al. Br J Dermatol. 2017. 177(2): 350-358.
[0010]A skin, nail or scalp wound is damage or disruption to its normal anatomical structure and function. This can range from a simple break in the epithelial integrity of the skin to a deeper wound, extending into subcutaneous tissue with damage to other structures such as tendons, muscles, vessels, nerves, parenchymal organs and even bone. Wound healing is a normal feature of living animals and is a dynamic, interactive process which involves various types of cell and the extracellular matrix. The normal healing process can be described in terms of four programmed phases comprising 1 ) hemostasis, 2) inflammation, 3) proliferation and 4) remodeling. The speed and efficiency of the process of wound healing depends on various internal and external factors. See Velnar T et al. J Int Med Res. 2009. 37(5): 1528-42.
[0011] Even though wound healing is a normal feature of damage to the derma (including the epidermis and dermis), some wounds remain a challenging clinical problem and correct, efficient wound management is essential. In addition to a high number of acute wounds, there are also a large number of chronic, “hard-to-heal” wounds (chronic nonhealing wounds) associated with diseases and abnormalities that directly or indirectly culminate in damage of the cutaneous coverage, including arterial, venous, diabetic and pressure ulcers. The prevalence of these chronic wounds increases with age. Further, due to the complications that accompany acute wounds, when their healing does not progress in a timely and orderly manner, they can become chronic wounds, which tend to be more difficult to manage. Much effort has focused on understanding the physiology of healing and wound care with an emphasis on new therapeutic approaches and the continuing development of technologies for acute and long-term wound management. See Alonso JE et al. Surg Clin North Am 1996; 76: 879 - 903; Natarajan S et al. Am J Clin Dermatol. 2000. 1 : 269-275; Robson MC et al. Curr Probl Surg. 2001. 28: 72-140; Velnar T et al. J Int Med Res. 2009. 37(5): 1528-42.
[0012] Since therapeutic response can vary on the basis of heterogeneous clinical and molecular phenotypes, a shift toward personalized or precision medicine approaches, including biomarker development and validation, has been thought to improve the management of many dermatological pathologies and skin diseases/disorders. Substantial progress in molecular immunology, coupled with an increased focus on translational research and personalized medicine, has resulted in a rapid expansion in the field of immune biomarkers in recent years. Such biomarkers might be used as an objective measure of normal versus pathogenic processes or indicator of pharmacological responses to therapeutic inventions. See Biomarkers Definitions Working Group. Clin Pharmacol Then 2001. 69(3): 89-95; Willis JCD and Lord GM. Nat Rev Immunol. 2015. 15:323-329; Renert-Yuval Y et al. J Allergy Clin Immunol. 2021. 147(4): 1174-1190.
[0013] Cluster of Differentiation-86 (CD86), also known as B7-2 plays a key role in regulating T cell activation and tolerance. The engagement of CD86 with its receptors, CD28 or CTLA-4, can deliver critical second signal that can promote and sustain or downregulate T cell responses. This dual role of CD86 can contribute to T cell activation and survival or T cell inhibition respectively. See Greenwald RJ et al. Annu Rev Immunol. 2005. 23:515-48; Chen L and Flies DB. Nat Rev Immunol. 2013. 13(4):227-242. Upregulated expression of CD86 on circulating intermediate monocytes positively correlated with the clinical severity of psoriasis measured by Psoriasis Area Severity Index (PASI) scores. The CD86+ monocytes located within psoriatic plaques were associated with enhanced epidermal proliferation. See Nguyes CTH et al. J Dermatol Sci. 2018. 90(2): 135-143. Further, targeting cutaneous dendritic cells with small interfering RNA (siRNA)-based CD86 therapy, CD86-siRNA, produced an amelioration in clinical manifestations of allergic skin diseases using a murine model. See Ritprajak P et al. Mol Then 2008. 16(7): 1323-30.
[0014] Cluster of Differentiation-109 (CD109), a TGF-b co-receptor, is a glycosyl phosphatidylinositol (GPI)-anchored protein, and is a co-receptor and negative regulator of TGF-[3 signaling. CD109 has been shown to bind to the TGF-b1 isoform with high affinity, forms a complex with the TGF-b receptors and inhibits TGF-b signaling by increasing TGF-b receptor internalization and degradation. In addition, CD109 can be released from the cell surface by cellular lipases such as phosphatidylinositol-specific phospholipase C (PI-PLC), and the soluble form of CD109 retains its ability to bind TGF- b1 and sequester it away from the TGF-b receptors. Aberrant CD109 release from the cell surface in human keratinocytes has been postulated to induce molecular changes typically observed in psoriasis, potentially due to TGF-[3 receptor downregulation and decrease in TGF-[3 signaling. See Litvinov IV et al. Exp Dermatol. 2011. 20(8):627-32.
[0015] Cluster of Differentiation-122 (CD122), also known as interleukine-2 receptor subunit beta (IL-2Rb/IL-2RB) is part of the IL-2 receptor, which is involved in T cell- mediated immune responses. IL-2, which was the first cytokine that was molecularly cloned, was shown to be a T cell growth factor essential for the proliferation of T cells and the generation of effector and memory cells. See Abbas AK. Sci Immunol. 2018. 3(25): eaat1482.
[0016]T-cell activation is considered to be an important element in the pathogenesis of psoriasis, a human skin disease characterized by keratinocyte hyperproliferation, altered keratinocyte differentiation and inflammation of the dermis and epidermis. In mouse models of psoriasis (mice homozygous for the flaky skin (fsn) mutation), most CD8+ T cells in fsn/fsn peripheral lymph nodes (PLN) expressed high levels of CD122 as compared to control mice. Analysis of PLN cells from newborn fsn/fsn mice revealed that the high expression of CD122 on CD8+ cells was established by 2 weeks of age, just prior to the appearance of clinical skin disease, indicating that large numbers of T cells in these fsn/fsn mice were activated. See Abernethy NJ et al. Immunol Cell Biol. 2000. 78(6): 596-602. Targeting CD122, which has an important established role in both primary and secondary immune responses, has been further shown to reduce transplant rejection by targeting T-cell activation. Kelsey R. 2018. Nat Rev Nephrol. 2018 Dec;14(12):720.
[0017] Granulocyte Macrophage - Colony Stimulating Factor (GM-CSF) is a hematological growth factor that regulates the production, migration, proliferation, differentiation and function of hematopoietic cells. It was first identified as being able to induce in vitro the proliferation and differentiation of bone marrow progenitors into granulocytes and macrophages. In response to inflammatory stimuli, GM-CSF is released by various cell types including T lymphocytes, macrophages, fibroblasts and endothelial cells. GM-CSF then activates and enhances the production and survival of neutrophils, eosinophils, and macrophages. Native GM-CSF is usually produced near the site of action where it modulates in vitro proliferation, differentiation, and survival of hematopoietic progenitor cells, but is present in circulating blood in only picomolar concentrations (10’1° to 10’12 M). Several studies have shown that GM-CSF has a wide range of functions across different tissues in its action on myeloid cells, and GM-CSF deletion/depletion approaches have indicated its potential as an important therapeutic target in several inflammatory and autoimmune disorders. See A Metcalf D. Immunol Cell Biology. 1987, 65:35-43; Gasson JC. Blood. 1991 , 77:1131 -1145; Shannon MF et al. Crit Rev Immunol. 1997, 17:301 -323; Alexander WS. Int Rev Immunol. 1998, 16:651 -682; Barreda DR et al. Dev Comp Immunol. 2004, 28:509-554; Lee KMC et al. Immunotargets Then 2020. 9:225-240.
[0018] Recombinant human granulocyte-macrophage colony-stimulating factor (rhu GM- CSF) has been approved by the FDA for the treatment of neutropenia, blood dyscrasias and malignancies like leukemia in combination with chemotherapies. In the clinic, GM- CSF used for treatment of neutropenia and aplastic anemia following chemotherapy greatly reduces the risk of infection associated with bone marrow transplantation. Its utility in myeloid leukemia treatment and as a vaccine adjuvant is also well established. See Dorr RT. Clin Therapeutics. 1993. 15(1 ): 19-29; Armitage JO. Blood 1998, 92:4491 -4508; Kovacic JC et al. J Mol Cell Cardiol. 2007, 42:19-33; Jacobs PP et al. Microbial Cell Factories 2010, 9:93.
[0019] The identification of specific biomarkers can be used for the diagnosis, prognosis, or theranosis of dermatological pathologies and skin diseases/disorders. They can also be used to identify skin disorders and conditions that do not respond to monotherapy alone, and those that might benefit from combination therapies. Such combination therapies can potentially increase the percentage of patients who respond to treatments. Hence, there remains a need for new and more effective biomarkers and combination treatments of dermatological pathologies and skin diseases/disorders.
SUMMARY
[0020] Accordingly, in an aspect, the present disclosure relates to a method for treating dermatological pathologies and skin diseases/disorders, comprising: administering an effective amount of a composition comprising GM-CSF to a patient in need thereof, wherein the patient is characterized by an increased expression of Cluster of Differentiation 86 (CD86).
[0021] In an aspect, the present disclosure relates to a method for treating dermatological pathologies and skin diseases/disorders, comprising: administering an effective amount of a composition comprising GM-CSF to a patient in need thereof, wherein the patient is characterized by an increased expression of Cluster of Differentiation 109 (CD109).
[0022] In an aspect, the present disclosure relates to a method for treating dermatological pathologies and skin diseases/disorders, comprising: administering an effective amount of a composition comprising GM-CSF to a patient in need thereof, wherein the patient is characterized by an increased expression of Cluster of Differentiation 122 (CD122). In an aspect, the present disclosure provides a method for treating dermatological pathologies and skin diseases/disorders, comprising: (a) identifying a patient undergoing or having undergone treatment with an agent for dermatological issues and presenting as failed, intolerant, resistant, or refractory to the treatment with a dermatological agent; (b) determining the presence, absence or amount of CD86 and/or CD109 and/or CD122 in a sample from the patient; and (c) administering an effective amount of a granulocytemacrophage colony-stimulating factor (GM-CSF) agent to a patient demonstrating an increased or high expression and/or activity of CD86 and/or CD109 and/or CD122 relative to a pre-treated and/or undiseased state.
[0023] In an aspect, the present disclosure provides a method for selecting a patient for treatment with GM-CSF for a dermatological pathology and skin diseases based on the presence, absence or amount of CD86 and/or CD109 and/or CD122 CD86 and/or CD109 and/or CD122 in a sample from the patient.
BRIEF DESCRIPTION OF DRAWINGS
[0024] FIG. 1 illustrates a graph depicting the kinetics of expression of CD86 induced by sargramostim (LEUKINE). Monocytes and lymphocytes from a human donor were treated with sargramostim (LEUKINE) at various concentrations 0.33pM to 33.33pM. Expression of CD86 was assessed on day 1 on both monocytes and lymphocytes. In each graph of FIG. 1 , the top curve is monocytes, and the bottom curve is lymphocytes.
[0025] FIG. 2 illustrates graphs depicting the kinetics of expression of CD109 induced by sargramostim (LEUKINE). Monocytes and lymphocytes from a human donor were treated with sargramostim (LEUKINE) at various concentrations 0.33pM to 33.33pM. Expression of CD109 was assessed on day 1 on both monocytes and lymphocytes. In each graph of FIG. 2, the top curve is monocytes, and the bottom curve is lymphocytes.
[0026] FIG. 3 illustrates graphs depicting the kinetics of expression of CD 122 induced by sargramostim (LEUKINE). Monocytes and lymphocytes from a human donor were treated with sargramostim (LEUKINE) at various concentrations 0.33pM to 33.33pM. Expression of CD122 was assessed on day 1 , day 2 and day 3 on both monocytes and lymphocytes. In each graph of FIG. 3, the top curve is monocytes, and the bottom curve is lymphocytes.
DETAILED DESCRIPTION
[0027] The present disclosure relates to, in part, to the use of GM-CSF as an effective treatment for specific dermatological pathologies and skin diseases/disorders, selected using CD86, CD109 and/or CD122 as a predictive marker of disease sequelae and responsiveness to current therapy, e.g. with a dermatological agent.
[0028] In aspects, the present disclosure relates to improved treatments for dermatological pathologies and skin diseases/disorders in a patient that has failed, is intolerant, is resistant, or is refractory to the current treatment in use. For instance, in embodiments, evaluation of the presence, absence, levels or activity of CD86, CD109 and/or CD122 informs or predicts the disease state in the and, without limitation, the administration of GM-CSF converts the patient that has failed, is intolerant, is resistant, or is refractory to current treatment(s) for the dermatological indication. In one embodiment, the GM-CSF modulates CD86, or cells expressing the same, to improve a patient’s treatment outcome. In another embodiment, the GM-CSF modulates CD109, or cells expressing the same, to improve a patient’s treatment outcome. In still another embodiment, the GM-CSF modulates CD122, or cells expressing the same, to improve a patient’s treatment outcome.
[0029] Accordingly, in one aspect, the disclosure provides methods for treating dermatological pathologies and skin diseases/disorders. In embodiments, the dermatological pathology is a skin wound, skin condition or a skin disorder.
Compositions of Immunomodulatory and/or Dermatological Agents
[0030] In some embodiments, the additional immunomodulatory or dermatological agent is selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti-IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; non-steroidal ointments including crisborole; retinoids; immunosuppressants including azathioprine and methotrexate; enzyme inhibitors such as apremilast; nonsedating antihistamines; H2 antagonists including dapsone or omalizumab; and propranolol.
[0031] In some embodiments, the immunoregulatory agent is an antibody or antibody format which is selected from one or more of a monoclonal antibody, polyclonal antibody, antibody fragment, Fab, Fab', Fab'-SH, F(ab')2, Fv, single chain Fv, diabody, linear antibody, bispecific antibody, multispecific antibody, chimeric antibody, humanized antibody, human antibody, and fusion protein comprising the antigen-binding portion of an antibody.
Compositions of GM-CSF
[0032] GM-CSF used in the practice of the disclosure includes any pharmaceutically safe and effective GM-CSF, or any derivative thereof having the biological activity of GM-CSF. In an embodiment, the GM-CSF used in the practice of the present methods is rhu GM- CSF, such as sargramostim (LEUKINE). Sargramostim is a biosynthetic, yeast-derived, recombinant human GM-CSF, having a single 127 amino acid glycoprotein that differs from endogenous human GM-CSF by having a leucine instead of a proline at position 23. Other natural and synthetic GM-CSFs, and derivatives thereof having the biological activity of natural human GM-CSF, may be equally useful in the practice of the disclosure.
[0033] In embodiments, the GM-CSF is produced or producible in bacteria, yeasts, plants, insect cells, and mammalian cells. In embodiments, the GM-CSF is produced or producible in Escherichia coli cells. In embodiments, the GM-CSF is produced or producible in yeast cells. In embodiments, the GM-CSF is produced or producible in Chinese hamster ovary cells (CHO). In embodiments, the GM-CSF is not produced in E. coli cells. In embodiments, the GM-CSF is produced in a cell that allows for glycosylation, e.g. yeast or CHO cells.
[0034] In embodiments, the GM-CSF has an amino acid sequence of SEQ ID NO: 1 , or a variant of at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98% identity thereto. In embodiments, the GM-CSF has an amino acid sequence of SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO:4, or a variant of at least about 90%, or at least about 93%, or at least about 95%, or at least about 97%, or at least about 98% identity thereto. In embodiments, the GM-CSF is one of, sargramostim, molgramostim, and regramostim.
[0035] Without wishing to be bound by theory, the core of hGM-CSF consists of four helices that pack at angles. Crystal structures and mutagenic analysis of rhGM-CSF (Rozwarski D A et al., Proteins 26:304-13, 1996) showed that, in addition to apolar side chains in the protein core, 10 buried hydrogen bonding residues involve intramolecular hydrogen bonding to main chain atoms that were better conserved than residues hydrogen bonding to other side chain atoms; 24 solvation sites were observed at equivalent positions in the two molecules in the asymmetric unit, and the strongest among these was located in clefts between secondary structural elements. Two surface clusters of hydrophobic side chains are located near the expected receptor binding regions. Mutagenesis of residues on the helix A/helix C face confirmed the importance of certain Glu, Gly, and Gin residues. These residues are therefore not to be substituted in the functional substitution variants of hGM-CSF for use in the present disclosure and these helices are to be retained in a functional fragments or deletion variants of hGM-CSF for use in this disclosure. Further, in embodiments, one of ordinary skill can reference UniProtKB entry P04141 for structure information to inform the identity of variants.
[0036] The N-terminal helix of hGM-CSF governs high affinity binding to its receptor (Shanafelt A B et al. , EMBO J 10:4105-12, 1991 ) T ransduction of the biological effects of GM-CSF requires interaction with at least two cell surface receptor components, (one of which is shared with the cytokine IL-5). The above study identified receptor binding determinants in GM-CSF by locating unique receptor binding domains on a series of human-mouse hybrid GM-CSF cytokines. The interaction of GM-CSF with the shared subunit of their high affinity receptor complexes was governed by a very small part of the peptide chains. The presence of a few key residues in the N-terminal a-helix of was sufficient to confer specificity to the interaction.
[0037] In some embodiments, the amino acid mutations are amino acid substitutions, and may include conservative and/or non-conservative substitutions.
[0038] “Conservative substitutions” may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved. The 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1 ) hydrophobic: Met, Ala, Vai, Leu, lie; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gin; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe.
[0039] As used herein, “conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide. In addition, glycine and proline may be substituted for one another based on their ability to disrupt a-helices.
[0040] As used herein, “non-conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1 ) to (6) shown above.
[0041] In various embodiments, the substitutions may also include non-classical amino acids (e.g. selenocysteine, pyrrolysine, /V-formylmethionine [3-alanine, GABA and 5- Aminolevulinic acid, 4-aminobenzoic acid (PABA), D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2- amino butyric acid, y-Abu, s-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3- amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosme, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, [3-alanine, fluoro-amino acids, designer amino acids such as [3 methyl amino acids, C a-methyl amino acids, N a-methyl amino acids, and amino acid analogs in general).
[0042] Modification of the amino acid sequences may be achieved using any known technique in the art e.g., site-directed mutagenesis or PCR based mutagenesis. Such techniques are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y., 1989 and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y., 1989. Without wishing to be bound by theory, the degree of glycosylation of biosynthetic GM-CSFs appears to influence half-life, distribution, and elimination. (Lieschke and Burgess, N. Engl. J. Med. 327:28-35, 1992; Dorr, R. T., Clin. Then 15:19-29, 1993; Horgaard et al., Eur. J. Hematol. 50:32-36, 1993). In embodiments, the present GM-CSF molecules are glycosylated.
Biomarker
[0043] In aspects, the present methods relate to the utility of a predictive biomarkers to determine the use of GM-CSF in the treatment of dermatological pathologies and skin diseases/disorders. [0044] In one aspect, the present disclosure relates to a method of treating a patient in need of therapy wherein the patient is characterized by having failed or being intolerant or refractory to a treatment with a dermatological agent. In other aspects, an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring of a biomarker in a sample of the patient.
[0045] In embodiments, an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring a variety of patient parameters. In embodiments, the patient sample may be analyzed using, e.g. immunohistochemical or immunofluorescence techniques may be used to evaluate the immune infiltrate, for example, immune subsets such as IL-17-producing CD4+ Th cells (Th17 cells), CD8+ T cells (cytotoxic T cells), and systemic or circulating intermediate monocytes. In embodiments, polychromatic flow cytometry can be used to measure multiple surface and intracellular markers, allowing characterization of cell phenotype and activation state. In embodiments, whole blood can be used to evaluate changes in cell count with therapy or changes in cytokine levels, for example TGF-b, IL-2, IL-12, IL-23 and/or TNFa. In embodiments, deep sequencing techniques can be used to yield quantification of changes in individual cell clonotypes.
[0046] In embodiments, an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring the presence, absence, or amount of Cluster of Differentiation 86 (CD86) isotype in a sample of the patient.
[0047] In embodiments, an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring the presence, absence, or amount of Cluster of Differentiation 109 (CD109) isotype in a sample of the patient.
[0048] In embodiments, an assessment of the patient having failed or being intolerant or refractory to a treatment with a dermatological agent comprises measuring the presence, absence, or amount of Cluster of Differentiation 122 (CD122) isotype in a sample of the patient. [0049] In embodiments, the present disclosure relates to a method for treating a dermatological pathology wherein CD86 is used as a biomarker for predicting or determining the need for treatment with GM-CSF.
[0050] In embodiments, the present disclosure relates to a method for treating a dermatological pathology wherein CD109 is used as a biomarker for predicting or determining the need for treatment with GM-CSF.
[0051] In embodiments, the present disclosure relates to a method for treating a dermatological pathology wherein CD122 is used as a biomarker for predicting or determining the need for treatment with GM-CSF.
[0052] In embodiments, the presence, absence or amount of CD86, CD109 and/or CD122 by detection of protein and/or nucleic acids in a sample of the patient.
[0053] In embodiments, the presence, absence or amount of CD86, CD109 and/or CD122 is determined by ELISA, immunohistochemical staining, western blotting, in-cell western, immunofluorescent staining, or fluorescent activating cell sorting (FACS) in a sample of the patient.
[0054] In embodiments, the method for determining the presence, absence or amount of CD86, CD109 and/or CD122 is a method of characterizing a patient or selecting a patient for the treatment comprising GM-CSF.
[0055] In embodiments, the method of determining CD86, CD109 and/or CD122 presence, absence, levels or activity for the purposes of patient selection employs a sample, the sample selected from a biopsy, tissue or body fluid.
[0056] In embodiments, the method of patient selection is undertaken using a sample of the patient, where the sample is selected from blood, skin sample or tissue sample, tissue biopsy, a formalin-fixed or paraffin- embedded tissue specimen, cytological sample, cultured cells, plasma, serum, pus, urine, perspiration, tears, mucus, sputum, saliva and/or other body fluids.
[0057] In embodiments, the present methods direct patient treatment decisions. For instance, in embodiments, the method comprises the step of monitoring the expression and/or activity of CD86, CD109 and/or CD122 during the course of treatment. In embodiments, the methods may detect a high or increased CD86, CD109 and/or CD122 expression or activity, and this is correlative with the patient having failed or being intolerant or refractory to a treatment with a dermatological agent. In such embodiments, without limitation, this directs treatment of the patient with GM-CSF agents. In embodiments, the patient with an increased or high expression or activity of CD86, CD 109 and/or CD122 receives, for example, a greater dose of GM-CSF, and/or additional dermatological therapies.
[0058] In embodiments, the GM-CSF agents, as described herein, potentiate treatment with a dermatological agent. In embodiments, GM-CSF agents, as described herein, are used to modulate the patient’s immune system, e.g. by decreasing expression and/or activity of CD86, CD109 and/or CD122.
Methods of Treatment
[0059] In one aspect, the present disclosure relates to a method for treating a dermatological pathology comprising: administering an effective amount of a composition GM-CSF to a patient in need thereof.
[0060] In another aspect, the present disclosure relates to a method for treating a dermatological pathology, comprising: administering an effective amount of a composition comprising GM-CSF in conjunction with a dermatological agent to a patient in need thereof, wherein the patient is characterized by the presence, absence or amount of CD86, CD109, and/or CD122 in a sample of the patient.
[0061] In some aspects, the present disclosure relates to methods for treating a dermatological pathology, comprising: (a) identifying a patient undergoing or having undergone treatment with an agent for dermatological issues and presenting as failed, intolerant, resistant, or refractory to the treatment with a dermatological agent; (b) determining the presence, absence or amount of CD86 and/or CD109 and/or CD122 in a sample from the patient; and (c) administering an effective amount of a granulocytemacrophage colony-stimulating factor (GM-CSF) agent to a patient demonstrating an increased or high expression and/or activity of CD86 and/or CD109 and/or CD122 relative to a pre-treated and/or undiseased state. [0062] In further aspects, the present disclosure relates to methods of treating a dermatological pathology, comprising: (a) selecting a patient having skin condition or skin disorders or skin wounds and one or more of (i) increased expression and/or activity of CD86 relative to a non-diseased state; (ii) increased expression and/or activity of CD109 relative to a non-diseased state; and/or (iii) increased expression and/or activity of CD122 relative to a non-diseased state and (b) administering an effective amount of a composition comprising GM-CSF to the patient.
[0063] In embodiments, the present disclosure relates to a method for treating a dermatological pathology, comprising: administering an effective amount of a composition comprising GM-CSF alone or in conjunction with a dermatological agent to a patient in need thereof, wherein the patient is characterized by as a partial responder or a nonresponder to a dermatological treatment.
[0064] In embodiments, the present disclosure relates to a method for treating cancer, comprising: administering an effective amount of a composition comprising GM-CSF alone or in conjunction with a dermatological agent to a patient in need thereof, wherein the patient is characterized by having failed or being intolerant or refractory to a treatment with a dermatological agent.
[0065] In embodiments, the method of treatment causes a decrease in the expression and/or activity of CD86. In some embodiments, the method of treatment causes a decrease in the expression and/or activity of CD109. In some embodiments, the method of treatment causes a decrease in the expression and/or activity of CD 122.
[0066] In embodiments, the method of treatment prevents, treats, and/or mitigates progression and/or development of the dermatological pathology in the patient. In some embodiments, the method of treatment improves the skin disorder, skin condition or skin wound in the patient. In some embodiments, the method of treatment improves skin renewal in the patient. In other embodiments, the method of treatment prevents or mitigates excessive skin cell reproduction in the patient. In still other embodiments, the method of treatment reverses or prevents inflammation in the skin or systemically.
[0067] In embodiments, the method of treatment decreases or mitigates the activation of skin or systemic T lymphocytes. In some embodiments, the method of treatment modulates proliferation and/or activation of IL-17-producing CD4+ Th cells (Th17 cells) or CD8+ T cells (cytotoxic T cells). In some embodiments, the method decreases or mitigates the activation of skin or systemic monocytes. In some embodiments, the method of treatment modulates skin and/or systemic monocytes including circulating intermediate monocytes.
[0068] In embodiments, the method of treatment reverses or prevents excessive production and/or signaling of one or more inflammatory cytokines, such as TGF-b, IL-2, IL-12, IL-23 and/or TNFa.
[0069] In some embodiments, the agent that stimulates the survival, proliferation and activation of neutrophils, macrophages and/or dendritic cells is administered at a time selected from (i) the same time as a dermatological agent; (ii) within about 1 hour, about 2 hours, about 4 hours, about 8 hours, about 12 hours, about 24 hours, about 36 hours, about 48 hours, about 72 hours, or about 96 hours or about 1 week or about 2 weeks following administration of said dermatological agent; (iii) at least about 1 hour, about 2 hours, about 4 hours, about 8 hours, about 12 hours, about 24 hours, about 48 hours, about 36 hours, about 72 hours, or about 96 hours, or about 1 week or about 2 weeks prior to administration of the dermatological agent; and/or (iv) after at least an about 10%, about 20%, about 30%, about 40% or about 50% decrease in expression of an extracellular marker such as CD86, CD109 and/or CD122.
[0070] In some embodiments, the skin disorder selected from psoriasis, plaque psoriasis, eczema, rosacea, ichthyosis, vitiligo, hives, seborrheic dermatitis, cold sores, blisters, actinic keratosis, carbuncle, latex allergy, cellulitis, lupus rash, contact dermatitis, atopic dermatitis, wart, ringworm, pityriasis rosea, melasma, impetigo, infantile hemangiomas, actinic keratosis, flaky skin, damaged skin and/or chronic idiopathic urticaria (CIU). In other embodiments, the autoimmune skin disorder is selected from psoriasis, vitiligo, cutaneous lupus erythematosus, alopecia areata, and/or frontal fibrosing alopecia. In still other embodiments, the skin wound comprising an ulcer, diabetes-related wound, bum, sunburn, chemical warfare agent (e.g. Sulphur Mustard gas and/or chlorine gas), skin burn or blisters, aging skin wound, corneal ulceration wound, inflammatory gastrointestinal wound, bowel inflammatory disease wound, Crohn's disease wound, ulcerative due to colitis, hemorrhoids, epidermolysis bullosa wound, skin blistering wound, psoriasis wound, animal skin wound, animal diabetes wound, retinopathy wound, oral wound (mucositis), vaginal mucositis, a wound due to periodontal disease, a laceration, a wound by surgical resection, and/or a wound after surgical adhesion. In some embodiments, the patient is afflicted with one or more of breakouts, red and painful fluid-filled blisters, itchy and raised welts, thick and scaly or crusty skin patches, red and painful lumps under the skin, itchy and inflamed wheals, yellow or white scaly patches, scaly and silvery well-defined patches, disc-shaped scaly rash, angioedema, skin patches with loss of melanin and/or a rash with raised borders.
[0071] In embodiments, the dermatological pathology is a skin disease or infection. In embodiments, the skin disease or infection is selected from erysipelas, staphylococcal scalded skin syndrome, folliculitis, erysipeloid, pitted keratolysis, erythrasma, trichomycosis, trichosporonosis (including, without limitation, hepatic or pulmonary trichosporonosis, intertrigo, acute infectious eczematoid dermatitis, pseudofolliculitis, toe web infection, skin tuberculosis, mycobacterium marinum skin disease, mycobacterium ulcerans skin disease, actinomycetoma, actinomycosis, various viral skin infections, dermatophytoses, onychomycoses, chronic non-healing wounds, warts (including, without limitation, genital warts, perianal warts, vaginal warts, and/or various verrucas including, without limitation, vulgaris, palmaris, plantaris, and/or periungual), and invasive fungal disease (IFD).
[0072] In embodiments, the method further comprises administering one or more additional therapeutic agents, selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti-IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; non-steroidal ointments including crisborole; retinoids; immunosuppressants including azathioprine and methotrexate; enzyme inhibitors such as apremilast; non-sedating antihistamines; H2 antagonists including dapsone or omalizumab; and propranolol. Pharmaceutically Acceptable Salts and Excipients
[0073] The compositions described herein can possess a sufficiently basic functional group, which can react with an inorganic or organic acid, or a carboxyl group, which can react with an inorganic or organic base, to form a pharmaceutically acceptable salt. A pharmaceutically acceptable acid addition salt is formed from a pharmaceutically acceptable acid, as is well known in the art. Such salts include the pharmaceutically acceptable salts listed in, for example, Journal of Pharmaceutical Science, 66, 2-19 (1977) and The Handbook of Pharmaceutical Salts; Properties, Selection, and Use. P. H. Stahl and C. G. Wermuth (eds.), Verlag, Zurich (Switzerland) 2002, which are hereby incorporated by reference in their entirety.
[0074] Pharmaceutically acceptable salts include, by way of non-limiting example, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, camphorsulfonate, pamoate, phenylacetate, trifluoroacetate, acrylate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methylbenzoate, o-acetoxybenzoate, naphthalene- 2-benzoate, isobutyrate, phenylbutyrate, a-hydroxybutyrate, butyne-1 ,4-dicarboxylate, hexyne-1 ,4-dicarboxylate, caprate, caprylate, cinnamate, glycollate, heptanoate, hippurate, malate, hydroxymaleate, malonate, mandelate, mesylate, nicotinate, phthalate, teraphthalate, propiolate, propionate, phenylpropionate, sebacate, suberate, p-bromobenzenesulfonate, chlorobenzenesulfonate, ethylsulfonate, 2- hydroxyethylsulfonate, methylsulfonate, naphthalene-1 -sulfonate, naphthalene-2- sulfonate, naphthalene-1 ,5-sulfonate, xylenesulfonate, and tartarate salts.
[0075] The term “pharmaceutically acceptable salt” also refers to a salt of the compositions of the present disclosure having an acidic functional group, such as a carboxylic acid functional group, and a base. Suitable bases include, but are not limited to, hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxysubstituted mono-, di-, or tri-alkylamines, dicyclohexylamine; tributyl amine; pyridine; N- methyl, N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-OH-lower alkylamines), such as mono-; bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert- butylamine, or tris-(hydroxymethyl)methylamine, N,N-di-lower alkyl-N-(hydroxyl-lower alkyl)-amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine or tri-(2- hydroxyethyl)amine; N-methyl-D-glucamine; and amino acids such as arginine, lysine, and the like.
[0076] In some embodiments, the compositions described herein are in the form of a pharmaceutically acceptable salt.
Pharmaceutical Compositions and Formulations
[0077] In various embodiments, the present disclosure pertains to pharmaceutical compositions comprising the compositions, e.g. GM-CSF and/or an additional therapeutic agent, e.g. the therapeutic agent described herein, and a pharmaceutically acceptable carrier or excipient.
[0078] In embodiments, the additional therapeutic agent comprises selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti- IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; nonsteroidal ointments including crisborole; retinoids; immunosuppressants including azathioprine and methotrexate; enzyme inhibitors such as apremilast; non-sedating antihistamines; H2 antagonists including dapsone or omalizumab; and propranolol, and pharmaceutically acceptable salts, acids or derivatives of any of the above.
[0079]Any pharmaceutical compositions described herein can be administered to a patient as a component of a composition that comprises a pharmaceutically acceptable carrier or vehicle. Such compositions can optionally comprise a suitable amount of a pharmaceutically acceptable excipient so as to provide the form for proper administration.
[0080] In various embodiments, pharmaceutical excipients can be liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. The pharmaceutical excipients can be, for example, saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like. In addition, auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used. In one embodiment, the pharmaceutically acceptable excipients are sterile when administered to a patient. Water is a useful excipient when any agent described herein is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid excipients, specifically for injectable solutions. Suitable pharmaceutical excipients also include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. Any agent described herein, if desired, can also comprise minor amounts of wetting or emulsifying agents, or pH buffering agents. Other examples of suitable pharmaceutical excipients are described in Remington’s Pharmaceutical Sciences 1447-1676 (Alfonso R. Gennaro eds., 19th ed. 1995), incorporated herein by reference.
[0081] The present disclosure includes the described pharmaceutical compositions (and/or additional therapeutic agents) in various formulations. Any inventive pharmaceutical composition (and/or additional therapeutic agents) described herein can take the form of solutions, suspensions, emulsion, drops, tablets, pills, pellets, capsules, capsules containing liquids, gelatin capsules, powders, sustained-release formulations, suppositories, emulsions, aerosols, sprays, suspensions, lyophilized powder, frozen suspension, desiccated powder, or any other form suitable for use. In one embodiment, the composition is in the form of a capsule. In another embodiment, the composition is in the form of a tablet. In yet another embodiment, the pharmaceutical composition is formulated in the form of a soft-gel capsule. In a further embodiment, the pharmaceutical composition is formulated in the form of a gelatin capsule. In yet another embodiment, the pharmaceutical composition is formulated as a liquid. [0082] Where necessary, the inventive pharmaceutical compositions (and/or additional therapeutic agents) can also include a solubilizing agent. Also, the agents can be delivered with a suitable vehicle or delivery device as known in the art. Combination therapies outlined herein can be co-delivered in a single delivery vehicle or delivery device.
[0083] The formulations comprising the inventive pharmaceutical compositions (and/or additional therapeutic agents) of the present disclosure may conveniently be presented in unit dosage forms and may be prepared by any of the methods well known in the art of pharmacy. Such methods generally include the step of bringing the therapeutic agents into association with a carrier, which constitutes one or more accessory ingredients. Typically, the formulations are prepared by uniformly and intimately bringing the therapeutic agent into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into dosage forms of the desired formulation (e.g., wet or dry granulation, powder blends, etc., followed by tableting using conventional methods known in the art).
[0084] In various embodiments, any pharmaceutical compositions (and/or additional therapeutic agents) described herein is formulated in accordance with routine procedures as a composition adapted for a mode of administration described herein.
[0085] Routes of administration include, for example: topical, oral, intradermal, transdermal, subcutaneous, intramuscular, intraperitoneal, intravenous, intranasal, epidural, sublingual, intranasal, intracerebral, intravaginal, rectal, or by inhalation. Administration can be local or systemic. In some embodiments, the administering is by topical route. In another embodiment, the administration is by dermal route. The mode of administration can be left to the discretion of the practitioner, and depends in-part upon the site of the medical condition. In most instances, administration results in the release of any agent described herein onto or into the affected site.
[0086] In specific embodiments, the GM-CSF (and/or additional therapeutic agents) is administered via a topical route or dermal route.
[0087] In specific embodiments, the GM-CSF (and/or additional therapeutic agents) is administered to the skin. [0088] In specific embodiments, the GM-CSF (and/or additional therapeutic agents) is administered via a cream, gel, ointment, lotion, or spray.
[0089] In specific embodiments, the GM-CSF (and/or additional therapeutic agents) is administered by topical or dermal route. Without wishing to be bound by theory, topical or dermal route of sargramostim produces low systemic exposure in the patient.
[0090] In one embodiment, the pharmaceutical compositions (and/or additional therapeutic agents) described herein are formulated in accordance with routine procedures as a composition adapted for topical or dermal administration. Compositions for topical delivery can be in the form of a cream, gel, ointment, lotion, spray, aqueous or oily suspensions, powders, or emulsions, for example. Increased skin permeability and penetration may be achieved by non-invasive methods, for example, with the use of any nanocarriers combined with any pharmaceutical composition (and/or additional therapeutic agents) described herein. The skin can act as a reservoir, and can be used to deliver the compositions (and/or additional therapeutic agents) described herein for more extended periods in a sustained manner.
[0091] Compositions for oral delivery can be in the form of tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions, capsules, syrups, or elixirs, for example. Orally administered compositions can comprise one or more agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of Wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation.
[0092] Dosage forms suitable for parenteral administration (e.g. intravenous, intramuscular, intraperitoneal, subcutaneous and intra-articular injection and infusion) include, for example, solutions, suspensions, dispersions, emulsions, and the like. They may also be manufactured in the form of sterile solid compositions (e.g. lyophilized composition), which can be dissolved or suspended in sterile injectable medium immediately before use. They may contain, for example, suspending or dispersing agents known in the art. Formulation components suitable for parenteral administration include a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as EDTA; buffers such as acetates, citrates or phosphates; and agents for the adjustment of tonicity such as sodium chloride or dextrose.
[0093] For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). The carrier should be stable under the conditions of manufacture and storage, and should be preserved against microorganisms. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol), and suitable mixtures thereof.
[0094] Any inventive pharmaceutical compositions (and/or additional therapeutic agents) described herein can be administered by controlled-release or sustained-release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591 ,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; and 5,733,556, each of which is incorporated herein by reference in its entirety. Such dosage forms can be useful for providing controlled- or sustained-release of one or more active ingredients using, for example, hydropropyl cellulose, hydropropylmethyl cellulose, polyvinylpyrrolidone, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled- or sustained-release formulations known to those skilled in the art, including those described herein, can be readily selected for use with the active ingredients of the agents described herein. The disclosure thus provides single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled- or sustained-release.
[0095] Controlled- or sustained-release of an active ingredient can be stimulated by various conditions, including but not limited to, changes in pH, changes in temperature, stimulation by an appropriate wavelength of light, concentration or availability of enzymes, concentration or availability of water, or other physiological conditions or compounds.
[0096] In another embodiment, a controlled-release system can be placed in proximity of the target area to be treated, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled-release systems discussed in the review by Langer, 1990, Science 249:1527-1533) may be used.
[0097] Pharmaceutical formulations preferably are sterile. Sterilization can be accomplished, for example, by filtration through sterile filtration membranes. Where the composition is lyophilized, filter sterilization can be conducted prior to or following lyophilization and reconstitution.
Administration and Dosage
[0098] It will be appreciated that the actual dose of the composition to be administered according to the present disclosure will vary according to the particular dosage form, and the mode of administration. Many factors that may modify the action of the composition (e.g., body weight, gender, diet, time of administration, route of administration, rate of excretion, condition of the patient, drug combinations, genetic disposition and reaction sensitivities) can be taken into account by those skilled in the art. Administration can be carried out continuously or in one or more discrete doses within the maximum tolerated dose. Optimal administration rates for a given set of conditions can be ascertained by those skilled in the art using conventional dosage administration tests.
[0099] In embodiments, the expression of the various biomarkers provides guidance for the administration of GM-CSF at a specific dose, using a specific route, and/or determining the frequency and duration of therapy.
[00100] In embodiments, the GM-CSF is administered at a total dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg. In embodiments, the GM-CSF is administered at a total dose of about 250 pg.
[00101] In embodiments, the GM-CSF is administered at a dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg.
[00102] In embodiments, the GM-CSF is administered twice daily.
[00103] In embodiments, the GM-CSF is sargramostim, administered at a dose of about 125 pg, twice daily.
Combination Therapy and Additional Therapeutic Agents [00104] In various embodiments, the pharmaceutical composition of the present disclosure is co-administered in conjunction with additional agent(s), for example an immunomodulatory or dermatological agent, such as a checkpoint inhibitor. Coadministration can be simultaneous or sequential.
[00105] In one embodiment, the additional immunomodulatory or dermatological agent and the GM-CSF of the present disclosure are administered to a patient simultaneously. The term “simultaneously” as used herein, means that the immunomodulatory or dermatological agent and the GM-CSF are administered with a time separation of no more than about 60 minutes, such as no more than about 30 minutes, no more than about 20 minutes, no more than about 10 minutes, no more than about 5 minutes, or no more than about 1 minute. Administration of the immunomodulatory or dermatological agent and the GM-CSF can be by simultaneous administration of a single formulation (e.g., a formulation comprising the additional therapeutic agent and the GM-CSF composition) or of separate formulations (e.g., a first formulation including the immunomodulatory or dermatological agent and a second formulation including the GM-CSF composition).
[00106] Co-administration does not require the therapeutic agents to be administered simultaneously, if the timing of their administration is such that the pharmacological activities of the immunomodulatory or dermatological agent and the GM- CSF overlap in time, thereby exerting a combined therapeutic effect. For example, the immunomodulatory or dermatological agent and the targeting moiety, the GM-CSF composition can be administered sequentially. The term “sequentially” as used herein means that the immunomodulatory or dermatological agent and the GM-CSF are administered with a time separation of more than about 60 minutes. For example, the time between the sequential administration of the immunomodulatory or dermatological agent and the GM-CSF can be more than about 60 minutes, more than about 2 hours, more than about 5 hours, more than about 10 hours, more than about 1 day, more than about 2 days, more than about 3 days, more than about 1 week apart, more than about 2 weeks apart, or more than about one month apart. The optimal administration times will depend on the rates of metabolism, excretion, and/or the pharmacodynamic activity of the additional therapeutic agent and the GM-CSF being administered. Either the immunomodulatory or dermatological agent or the GM-CSF composition may be administered first.
[00107] Co-administration also does not require the therapeutic agents to be administered to the patient by the same route of administration. Rather, each therapeutic agent can be administered by any appropriate route, for example, parenterally or non- parenterally.
[00108] In some embodiments, the GM-CSF described herein acts synergistically when co-administered with the immunomodulatory or dermatological agent. In such embodiments, the targeting moiety, the GM-CSF composition and the immunomodulatory or dermatological agent may be administered at doses that are lower than the doses employed when the agents are used in the context of monotherapy.
Samples
[00109] In embodiments, the sample is selected from a biopsy, a tissue and/or a body fluid.
[00110] In embodiments, the sample is selected from blood, skin sample or tissue sample, tissue biopsy, a formalin-fixed or paraffin- embedded tissue specimen, cytological sample, cultured cells, plasma, serum, pus, urine, perspiration, tears, mucus, sputum, saliva and/or other body fluids.
Sequences
[00111] SEQ ID NO: 1 is wild type GM-CSF:
[00112] APARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVISEMFDL QEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQIITFESFKE NLKDFLLVIPFDCWEPVQE.
[00113] SEQ ID NO: 2 is sargramostim:
[00114] APARSPSPSTQPWEHVNAIQEALRLLNLSRDTAAEMNETVEVISEMFDL
QEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQIITFESFKE NLKDFLLVIPFDCWEPVQE.
[00115] SEQ ID NO: 3 is molgramostim: [00116] APARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVISEMFDL
QEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQIITFESFKE
NLKDFLLVIPFDCWEPVQE
[00117] SEQ ID NO: 4 is regramostim:
[00118] APARSPSPSTQPWEHVNAIQEARRLLNLSRDTAAEMNETVEVISEMFDL
QEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASHYKQHCPPTPETSCATQ TTFESFKE NLKDFLLVIPFDCWEPVQE
Definitions
[00119] The following definitions are used in connection with the invention disclosed herein. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of skill in the art to which this invention belongs.
[00120] An “effective amount,” when used in connection with an agent effective for the treatment of a coronavirus infection is an amount that is effective for treating or mitigating a coronavirus infection.
[00121] As used herein, “a,” “an,” or “the” can mean one or more than one. Further, the term “about” when used in connection with a referenced numeric indication means the referenced numeric indication plus or minus up to 10% of that referenced numeric indication. For example, the language “about 50” covers the range of 45 to 55.
[00122] As referred to herein, all compositional percentages are by weight of the total composition, unless otherwise specified. As used herein, the word “include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the materials, compositions, devices, and methods of this technology. Similarly, the terms “can” and “may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.
[00123] Although the open-ended term “comprising,” as a synonym of terms such as including, containing, or having, is used herein to describe and claim the invention, the present invention, or embodiments thereof, may alternatively be described using alternative terms such as “consisting of” or “consisting essentially of.”
EXAMPLES
Example 1 : Expression and Kinetics of CD86, CD109 and CD122 on Human Monocytes
[00124] Peripheral blood mononuclear cells from healthy volunteers were cultured overnight in the presence of with sargramostim (LEUKINE) at various concentrations from 0.05pM to 10pM. At completion of the culture period, the cells were harvested and stained for the expression of CD86, CD109, or CD122. The cells were co-stained for CD14 expression for the identification of monocytes. Expression of the various surface receptors was normalized to the level with no LEUKINE added and plotted versus the varying concentrations of LEUKINE. The procedure was replicated with distinct donors (n = 5), and error bars for the expression levels are shown. Treatment with sargramostim (LEUKINE) decreased the expression of CD86 (FIG. 1 ), CD109 (FIG. 2) and CD122 (FIG. 3) on monocytes but not lymphocytes.
EQUIVALENTS
[00125] Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
INCORPORATION BY REFERENCE
[00126] All patents and publications referenced herein are hereby incorporated by reference in their entireties.
[00127] As used herein, all headings are simply for organization and are not intended to limit the disclosure in any manner. The content of any individual section may be equally applicable to all sections.

Claims

CLAIMS What is claimed is:
1. A method of selecting a patient for treatment with an effective amount of a granulocyte-macrophage colony-stimulating factor (GM-CSF) agent for a dermatological pathology and/or skin diseases/disorders, comprising: determining the presence, absence or amount of Cluster of Differentiation 86 (CD86) in a sample from the patient, wherein the patient is suitable for the treatment if demonstrating an increased or high expression and/or activity of CD86 relative to a pre-treated and/or undiseased patient.
2. A method of selecting a patient for treatment with an effective amount of a granulocyte-macrophage colony-stimulating factor (GM-CSF) agent for a dermatological pathology and/or skin diseases/disorders, comprising: determining the presence, absence or amount of Cluster of Differentiation 109 (CD109) in a sample from the patient, wherein the patient is suitable for the treatment if demonstrating an increased or high expression and/or activity of CD109 relative to a pre-treated and/or undiseased patient.
3. A method of selecting a patient for treatment with an effective amount of a granulocyte-macrophage colony-stimulating factor (GM-CSF) agent for a dermatological pathology and/or skin diseases/disorders, comprising: determining the presence, absence or amount of Cluster of Differentiation 122 (CD122) in a sample from the patient, wherein the patient is suitable for the treatment if demonstrating an increased or high expression and/or activity of CD122 relative to a pre-treated and/or undiseased patient.
4. The method of claims 1 -3, wherein the patient is treated with an additional agent(s) comprising: administering an effective amount of drug(s) to treat a dermatological pathology and/or skin diseases/disorders.
5. A method for treating or preventing a dermatological pathology and/or skin diseases/disorders, comprising:
(a) identifying a patient undergoing or having undergone treatment with an agent for dermatological issues and presenting as failed, intolerant, resistant, or refractory to the treatment with a dermatological agent; (b) determining the presence, absence or amount of CD86 and/or CD109 and/or CD122 in a sample from the patient; and
(c) administering an effective amount of a granulocyte-macrophage colonystimulating factor (GM-CSF) agent to a patient demonstrating an increased or high expression and/or activity of CD86 and/or CD109 and/or CD122 relative to a pretreated and/or undiseased state.
6. The method of claims 1-5, wherein the dermatological pathology and/or skin diseases/disorders is a skin wound, skin condition or skin disorder or skin diseases or infections or nail infection/disease/disorders or hair/scalp infection/disease/disorders.
7. The method of claims 1-6, wherein the patient is characterized by having a skin wound, the skin wound comprising an ulcer, diabetes-related wound, burn, sunburn, chemical warfare agent (optionally Sulphur Mustard gas and/or chlorine gas), skin burn or blisters, aging skin wound, corneal ulceration wound, inflammatory gastrointestinal wound, bowel inflammatory disease wound, Crohn's disease wound, ulcerative colitis, hemorrhoids, epidermolysis bullosa wound, skin blistering wound, psoriasis wound, animal skin wound, animal diabetes wound, retinopathy wound, oral wound (mucositis), vaginal mucositis, a wound due to periodontal disease, a laceration, a wound by surgical resection, and/or a wound after surgical adhesion.
8. The method of claims 1-6, wherein the patient is characterized by having a skin/nail/scalp condition or skin/nail/scalp disorders or skin/nail/scalp diseases or infections optionally selected from damaged skin, flaky skin, psoriasis, plaque psoriasis, eczema, rosacea, ichthyosis, vitiligo, hives, seborrheic dermatitis, cold sores, blisters, actinic keratosis, carbuncle, latex allergy, cellulitis, lupus rash, contact dermatitis, atopic dermatitis, wart, ringworm, pityriasis rosea, melasma, impetigo, infantile hemangiomas, actinic keratosis, chronic idiopathic urticaria (Clll), erysipelas, staphylococcal scalded skin syndrome, folliculitis, erysipeloid, pitted keratolysis, erythrasma, trichomycosis, trichosporonosis (optionally hepatic or pulmonary trichosporonosis, intertrigo, acute infectious eczematoid dermatitis, pseudofolliculitis, toe web infection, skin tuberculosis, mycobacterium marinum skin disease, mycobacterium ulcerans skin disease, actinomycetoma, actinomycosis, various viral skin infections, dermatophytoses, onychomycoses, chronic non-healing wounds, warts (optionally genital warts, perianal warts, vaginal warts, and various verrucas), invasive fungal disease (IFD), paronychia, nail fungus, lichen planus, pseudomonas nail infection, and tinea capitis among others.
9. The method of claims 1 -6, wherein the dermatological pathology is an autoimmune skin disorder optionally selected from psoriasis, vitiligo, cutaneous lupus erythematosus, alopecia areata, and/or frontal fibrosing alopecia.
10. The method of any of the claims 1-9, wherein the patient is afflicted with a chronic or acute skin condition or disorder.
11. The method of any of the claims 1-6, wherein the patient is further afflicted with psoriatic arthritis.
12. The method of any one of claims 1 -11 , wherein the patient is afflicted with one or more of breakouts, red and painful fluid-filled blisters, itchy and raised welts, thick and scaly or crusty skin patches, red and painful lumps under the skin, itchy and inflamed wheals, yellow or white scaly patches, scaly and silvery well-defined patches, discshaped scaly rash, angioedema, skin patches with loss of melanin and/or a rash with raised borders.
13. The method of any one of claims 1 -5, wherein the presence, absence, or amount of CD86, CD109 and/or CD122 is determined by detection of protein and/or nucleic acids.
14. The method of claim 13, wherein the presence, absence, or amount of CD86, CD109 and/or CD122 is determined by ELISA, immunohistochemical staining, western blotting, in-cell western, immunofluorescent staining, or fluorescent activating cell sorting (FACS).
15. The method of claims 1 -5, wherein the biological sample is or comprises blood, skin sample or tissue sample, tissue biopsy, a formalin-fixed or paraffin- embedded tissue specimen, cytological sample, cultured cells, plasma, serum, pus, urine, perspiration, tears, mucus, sputum, saliva and/or other body fluids.
16. The method of any one of claims 1 -15, wherein the method prevents, treats, and/or mitigates progression and/or development of the dermatological pathology in the patient.
17. The method of any one of claims 1 -16, wherein the method improves skin renewal in the patient.
18. The method of any one of claims 1-17, wherein the method prevents or mitigates excessive skin cell reproduction in the patient.
19. The method of any one of the claims 1-18, wherein the method reverses or prevents inflammation in the skin or systemically.
20. The method of claim 19, wherein the method decreases or mitigates the activation of skin or systemic T lymphocytes.
21. The method of claim 20, where the T lymphocytes are IL-17-producing CD4+ Th cells (Th17 cells).
22. The method of claim 20, where the T lymphocytes are CD8+ T cells (cytotoxic T cells).
23. The method of claim 19, wherein the method decreases or mitigates the activation of skin and/or systemic monocytes.
24. The method of claim 23, wherein the monocytes are circulating intermediate monocytes.
25. The method of claim 23 or 24, wherein the method decreases the expression and/or activity of CD86.
26. The method of claims 19-25, wherein the method reverses or prevents excessive production and/or signaling of one or more inflammatory cytokines.
27. The method of claim 26, wherein the inflammatory cytokine is TGF-b.
28. The method of claim 26 or 27, wherein the method causes a decrease in the expression and/or activity of a TGF-b co-receptor such as CD109.
29. The method of claim 26, wherein the inflammatory cytokine is IL-2.
30. The method of claim 26 or 29, wherein the method causes a decrease in the expression and/or activity of the IL-2 receptor beta chain, CD122.
31 . The method of any one of claims 1-30, wherein the method provides guidance on dose, route, frequency and duration of therapy.
32. The method of any one of claims 1 -31 , wherein the GM-CSF has an amino acid sequence of SEQ ID NO: 1 , or a variant of about 90%, or about 93%, or about 95%, or about 97%, or about 98% identity thereto.
33. The method of any one of claims 1 -32, wherein the GM-CSF has an amino acid sequence of one of SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO:4, or a variant of about 90%, or about 93%, or about 95%, or about 97%, or about 98% identity thereto.
34. The method of any one of claims 1 -33, wherein the GM-CSF is one of sargramostim, molgramostim, and regramostim.
35. The method of claim 34, wherein the GM-CSF is sargramostim.
36. The method of any one of claims 1 -35, wherein the GM-CSF is administered at a total dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg.
37. The method of claim 36, wherein the GM-CSF is administered at a total dose of about 250 pg.
38. The method of any one of claims 36 or 37, wherein the GM-CSF is administered twice daily.
39. The method of claim 38, wherein the GM-CSF is sargramostim, administered at a dose of about 125 pg, twice daily.
40. The method of any one of claims 1 -39, wherein the GM-CSF is administered to via an oral, intradermal, transdermal, subcutaneous, intramuscular, intraperitoneal, intravenous, intranasal, epidural, sublingual, intranasal, intracerebral, intravaginal, rectal, or by inhalation.
41 . The method of any one of claims 1 -39, wherein the GM-CSF is administered via a topical route or dermal route.
42. The method of any one of the claims 1-41 , wherein the GM-CSF is administered via a cream, gel, ointment, lotion, or spray.
43. The method of any one of claims 1-42, wherein the method further comprises administering one or more additional therapeutic agents, selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti-IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; non-steroidal ointments including crisborole; retinoids; immunosuppressants including azathioprine and methotrexate; enzyme inhibitors such as apremilast; non-sedating antihistamines; H2 antagonists including dapsone or omalizumab; and propranolol.
44. A method for treating a dermatological pathology and/or skin diseases/disorders, comprising:
(a) selecting a patient having skin condition or skin disorders or skin wounds or nail infection/disease/disorders or hair/scalp infection/disease/disorders and one or more of
(i) increased expression and/or activity of CD86 relative to a non-diseased state;
(ii) increased expression and/or activity of CD109 relative to a non-diseased state; and/or
(iii) increased expression and/or activity of CD122 relative to a nondiseased state and
(b) administering an effective amount of a composition comprising GM-CSF to the patient.
45. The method of claim 44, wherein the method further comprises the step of monitoring the expression and/or activity of CD86 during the course of treatment.
46. The method of claim 45, wherein an increased expression and/or activity of CD86 directs continued administration of GM-CSF.
47. The method of claim 45, wherein decreased expression and/or activity of CD86 directs discontinuation of administration of GM-CSF.
48. The method of claim 44, wherein the method further comprises the step of monitoring the expression and/or activity of CD109 during the course of treatment.
49. The method of claim 48, wherein an increased expression and/or activity of CD109 directs continued administration of GM-CSF.
50. The method of claim 48, wherein a decreased expression and/or activity of CD109 directs discontinuation of administration of GM-CSF.
51. The method of claim 44, wherein the method further comprises the step of monitoring the expression and/or activity I of CD122 during the course of treatment.
52. The method of claim 51 , wherein an increased expression and/or activity of CD122 directs continued administration of GM-CSF.
53. The method of claim 51 , wherein a decreased expression and/or activity of CD122 directs discontinuation of administration of GM-CSF.
54. The method of claims 44-53, wherein the dose of GM-CSF administered to a patient is dependent on the expression and/or activity of CD86 and/or CD109 and/or CD122.
55. The method of any one of claims 44-54, wherein the expression and/or activity of CD86, CD109, and/or CD122 are assayed in a biological sample from the patient.
56. The method of any one of claims 44-55, wherein the biological sample comprises blood, skin sample or tissue sample, tissue biopsy, a formalin-fixed or paraffin- embedded tissue specimen, cytological sample, cultured cells, plasma, serum, pus, urine, perspiration, tears, mucus, sputum, saliva, and/or other body fluids.
57. The method of any one of claims 44-56, wherein the skin disorder or skin condition or skin wound or skin disease or infection or nail infection/disease/disorders or hair/scalp infection/disease/disorders is selected from
(i) a skin disorder or skin condition selected from psoriasis, plaque psoriasis, eczema, rosacea, ichthyosis, vitiligo, hives, seborrheic dermatitis, cold sores, blisters, actinic keratosis, carbuncle, latex allergy, cellulitis, lupus rash, contact dermatitis, atopic dermatitis, wart, ringworm, pityriasis rosea, melasma, impetigo, infantile hemangiomas, actinic keratosis, flaky skin, damaged skin and/or chronic idiopathic urticaria (Clll);
(ii) an autoimmune skin disorder selected from psoriasis, vitiligo, cutaneous lupus erythematosus, alopecia areata, and/or frontal fibrosing alopecia; and/or
(iii) a skin wound comprising an ulcer, diabetes-related wound, burn, sunburn, chemical warfare agent (optionally Sulphur Mustard gas and/or chlorine gas), skin burn or blisters, aging skin wound, corneal ulceration wound, inflammatory gastrointestinal wound, bowel inflammatory disease wound, Crohn's disease wound, ulcerative due to colitis, hemorrhoids, epidermolysis bullosa wound, skin blistering wound, psoriasis wound, animal skin wound, animal diabetes wound, retinopathy wound, oral wound (mucositis), vaginal mucositis, a wound due to periodontal disease, a laceration, a wound by surgical resection, and/or a wound after surgical adhesion.
(iv) a skin infection selected from erysipelas, staphylococcal scalded skin syndrome, folliculitis, erysipeloid, pitted keratolysis, erythrasma, trichomycosis, trichosporonosis (optionally selected from hepatic or pulmonary trichosporonosis, dermatophytosis, intertrigo, acute infectious eczematoid dermatitis, pseudofolliculitis, toe web infection, skin tuberculosis, mycobacterium marinum skin disease, mycobacterium ulcerans skin disease, actinomycetoma, actinomycosis, various viral skin infections, dermatophytoses, onychomycoses, chronic non-healing wounds, warts (optionally selected from genital warts, perianal warts, vaginal warts, and various verrucas), invasive fungal disease (IFD) paronychia, nail fungus, lichen planus, pseudomonas nail infection, and tinea capitis.
58. The method of claim 57, wherein the patient is afflicted with a chronic or acute dermatological pathology.
59. The method of claim 57, wherein the skin disorder is psoriasis or plaque psoriasis.
60. The method of any one of claims 44-59, wherein the patient is afflicted with one or more of breakouts, red and painful fluid-filled blisters, itchy and raised welts, thick and scaly or crusty skin patches, red and painful lumps under the skin, itchy and inflamed wheals, yellow or white scaly patches, scaly and silvery well-defined patches, discshaped scaly rash, angioedema, skin patches with loss of melanin and/or a rash with raised borders.
61 . The method of any one of claims 44-60, wherein the method prevents, treats, and/or mitigates progression and/or development of the dermatological pathology in the patient.
62. The method of any one of claims 44-61 , wherein the method improves the skin disorder, skin condition or skin wound in the patient.
63. The method of any one of claims 44-62, wherein the method reverses or prevents excessive production of one or more inflammatory cytokines.
64. The method of claim 63, wherein the inflammatory cytokine is one or more of TGF- b, IL-2, IL-12, IL-23 and/or TNFa.
65. The method of any one of claims 44-64, wherein the method causes a decrease in the sequelae of a dermatological pathology in the patient relative to before treatment.
66. The method of any one of claims 44-65, wherein the GM-CSF has an amino acid sequence of SEQ ID NO: 1 , or a variant of about 90%, or about 93%, or about 95%, or about 97%, or about 98% identity thereto.
67. The method of any one of claims 44-66, wherein the GM-CSF has an amino acid sequence of one of SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, or a variant of about 90%, or about 93%, or about 95%, or about 97%, or about 98% identity thereto.
68. The method of any one of claims 44-67, wherein the GM-CSF is one of sargramostim, molgramostim, and regramostim.
69. The method of claim 68, wherein the GM-CSF is sargramostim.
70. The method of any one of claims 44-69, wherein the GM-CSF is administered at a total dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg.
71. The method of claim 70, wherein the GM-CSF is administered at a total dose of about 250 pg.
72. The method of any one of claims 44-71 , wherein the GM-CSF is administered at a dose of about 125 pg, about 150 pg, or about 200 pg, or about 250 pg, or about 300 pg, or about 350 pg.
73. The method of any one of claims 44-72, wherein the GM-CSF is administered twice daily.
74. The method of claim 73, wherein the GM-CSF is sargramostim, administered at a dose of about 125 pg, twice daily.
75. The method of any one of claims 44-74, wherein the GM-CSF is administered via a topical route or dermal route.
76. The method of any one of claims 65-74, wherein the GM-CSF is administered via an intravenous route.
77. The method of claim 76, wherein the GM-CSF is administered via a cream, gel, ointment, lotion, or spray.
78. The method of any one of claims 43-77, wherein the method further comprises administering one or more additional therapeutic agents, selected from corticosteroids including hydrocortisone, triamcinolone, fluocinonide, and clobetasol; biologies such as TNF-a antagonists including etanercept, adalimumab, infliximab; anti-IL12/23 agents including ustekinumab; IL-17 inhibitors including secukinumab, ixekizumab, brodalumab; JAK receptor inhibitors including tofacitinib, baricitinib; PDE4 inhibitors including apremilast; antibacterials including mupirocin and clindamycin; anthralin; antivirals such as oral and intravenous acyclovir, famciclovir, and valacyclovir; antifungals such as clotrimazole and ketoconazole; benzoyl peroxide; coal tar; non-steroidal ointments including crisborole; retinoids; immunosuppressants including azathioprine and methotrexate; enzyme inhibitors such as apremilast; non-sedating antihistamines; H2 antagonists including dapsone or omalizumab; and propranolol.
PCT/US2023/060966 2022-01-21 2023-01-20 Granulocyte-macrophage colony-stimulating factor-based dermatological pathology treatments WO2023141551A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202263301707P 2022-01-21 2022-01-21
US63/301,707 2022-01-21
US202263347766P 2022-06-01 2022-06-01
US63/347,766 2022-06-01
US202263413772P 2022-10-06 2022-10-06
US63/413,772 2022-10-06

Publications (1)

Publication Number Publication Date
WO2023141551A1 true WO2023141551A1 (en) 2023-07-27

Family

ID=87349159

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/060966 WO2023141551A1 (en) 2022-01-21 2023-01-20 Granulocyte-macrophage colony-stimulating factor-based dermatological pathology treatments

Country Status (1)

Country Link
WO (1) WO2023141551A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110008394A1 (en) * 2008-03-14 2011-01-13 Basf Beauty Care Solutions France S.A.S. Active substances that increase the activation threshold of immune cells
US8673859B2 (en) * 2007-03-20 2014-03-18 New York University GM-CSF cosmeceutical compositions and methods of use thereof
US20150190470A1 (en) * 2012-08-01 2015-07-09 Tetralogic Pharmaceuticals Corporation Combination therapy
US20170306416A1 (en) * 2014-10-08 2017-10-26 Felipe Bedoya Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2022015960A2 (en) * 2020-07-17 2022-01-20 Icahn School Of Medicine At Mount Sinai Biomarkers and classifier of psoriasis and methods of treatment

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8673859B2 (en) * 2007-03-20 2014-03-18 New York University GM-CSF cosmeceutical compositions and methods of use thereof
US20110008394A1 (en) * 2008-03-14 2011-01-13 Basf Beauty Care Solutions France S.A.S. Active substances that increase the activation threshold of immune cells
US20150190470A1 (en) * 2012-08-01 2015-07-09 Tetralogic Pharmaceuticals Corporation Combination therapy
US20170306416A1 (en) * 2014-10-08 2017-10-26 Felipe Bedoya Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2022015960A2 (en) * 2020-07-17 2022-01-20 Icahn School Of Medicine At Mount Sinai Biomarkers and classifier of psoriasis and methods of treatment

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NGUYEN CHUYEN THI HONG, KAMBE NAOTOMO, YAMAZAKI FUMIKAZU, UEDA-HAYAKAWA IKUKO, KISHIMOTO IZUMI, OKAMOTO HIROYUKI: "Up-regulated expression of CD86 on circulating intermediate monocytes correlated with disease severity in psoriasis", JOURNAL OF DERMATOLOGICAL SCIENCE, ELSEVIER, AMSTERDAM, NL, vol. 90, no. 2, 1 May 2018 (2018-05-01), AMSTERDAM, NL , pages 135 - 143, XP093081770, ISSN: 0923-1811, DOI: 10.1016/j.jdermsci.2018.01.005 *
SCHUTT CHARLES: "Effects of GM-CSF on Dendritic Cells and Regulatory T cells in Parkinson arkinson’s Disease P s Disease Patients and Models of P atients and Models of Parkinson arkinson’s Disease s Disease ", DOCTORAL DISSERTATION, UNIVERSITY OF NEBRASKA MEDICAL CENTER, 15 December 2017 (2017-12-15), XP093081776, Retrieved from the Internet <URL:https://digitalcommons.unmc.edu/etd/236/#:~:text=In%20conclusion%2C%20GM%2DCSF%20induces,mediators%20and%20induction%20of%20Tregs.> [retrieved on 20230913] *

Similar Documents

Publication Publication Date Title
Cai et al. A critical role of the IL-1β–IL-1R signaling pathway in skin inflammation and psoriasis pathogenesis
Han et al. IL-38 ameliorates skin inflammation and limits IL-17 production from γδ T cells
Gu et al. The immunobiology of systemic sclerosis
Yang et al. Mitochondrial Ca2+ and membrane potential, an alternative pathway for Interleukin 6 to regulate CD4 cell effector function
EP3416634A1 (en) Immunomodulatory agents and methods of use thereof
US20170198039A1 (en) Methods for treating cancer
JP2013231052A (en) Pharmaceutical composition and kit for treatment of inflammatory disease and disorder
JP2704880B2 (en) Angiogenin inhibitors
Antiga et al. The role of etanercept on the expression of markers of T helper 17 cells and their precursors in skin lesions of patients with psoriasis vulgaris
WO2023141551A1 (en) Granulocyte-macrophage colony-stimulating factor-based dermatological pathology treatments
US11168112B2 (en) C1q and HMGB1 fusion proteins and uses thereof
Lee et al. IL-1 receptor antagonist (IL-1Ra)-Fc ameliorate autoimmune arthritis by regulation of the Th17 cells/Treg balance and arthrogenic cytokine activation
US20090191191A1 (en) Methods for the Modulation of IL-13
WO2017201448A1 (en) Animal models for psoriasis and screening methods
US20150307619A1 (en) Use of C-C Chemokine Receptor Type 7 (CCR7) Inhibitors
CA2568384A1 (en) Immune response suppressor and treatment of multiple sclerosis
JPH1052282A (en) Angiogenin suppressing agent
US20200355684A1 (en) Asprv1 as a neutrophil-specific marker and therapeutic target for inflammatory diseases
Camisa Pathogenesis of psoriasis
US20240025986A1 (en) Therapy for glaucoma and optic neuropathy by targeting colony stimulating factors
EP4052727A1 (en) Immune response suppressor
WO2023178323A9 (en) Therapeutic inhibition of cd38 to treat hidradenitis suppurativa
US20130296250A1 (en) Method for treatment of psoriasis
Bogaert et al. Congenital Defects of Phagocytes
Benedetti et al. Research Strands in Dermatology and Gastroenterology Units of Department of Clinical and Molecular Sciences in Polytechnic Marche University

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23743938

Country of ref document: EP

Kind code of ref document: A1