WO2020201444A1 - Repurposing small molecules for senescence-related diseases and disorders - Google Patents

Repurposing small molecules for senescence-related diseases and disorders Download PDF

Info

Publication number
WO2020201444A1
WO2020201444A1 PCT/EP2020/059436 EP2020059436W WO2020201444A1 WO 2020201444 A1 WO2020201444 A1 WO 2020201444A1 EP 2020059436 W EP2020059436 W EP 2020059436W WO 2020201444 A1 WO2020201444 A1 WO 2020201444A1
Authority
WO
WIPO (PCT)
Prior art keywords
senolytic
treatment
therapy
subject
disorders
Prior art date
Application number
PCT/EP2020/059436
Other languages
French (fr)
Inventor
Süleyman Selim ÇINAROGLU
Emel TIMUCIN
Ugar SEZERMAN
Gunseli Bayram AKCAPINAR
Yavuz Ahiska
Original Assignee
Eternans Ltd
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 Eternans Ltd filed Critical Eternans Ltd
Publication of WO2020201444A1 publication Critical patent/WO2020201444A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • A61K31/015Hydrocarbons carbocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • Senescence and apoptosis are among mechanisms that, when activated, restrict tumor growth. Through apoptosis, damaged cells are cleared from the organism while senescent cells remain alive in the organism though permanently restricted from entering the cell cycle. Senescence may be associated with an increase in metabolic activity. In the majority of cases, senescent cells develop a defined, but heterogeneous, secretory profile termed as senescence- associated secretory phenotype (SASP). The SASP entails release of pro-inflammatory cytokines and chemokines, tissue-damaging proteases, factors that can affect stem and progenitor cell function, haemostatic factors, and growth factors, among others. Senescent cells that express the SASP can have substantial local and systemic pathogenic effects.
  • SASP senescence- associated secretory phenotype
  • the SASP secretion comprises a range of different proteins, including several proteins known to play a role in aging and age-related diseases, including matrix metalloproteases such as MMP3, growth factors, chemokines such as CCL2 and CLL11, and prominent interleukins (ILs) such as ILl, IL6, and IL8. Above a certain threshold, such factors can significantly impair tissue function.
  • the chronic SASP secretion by senescent cells may impair the functioning of neighboring cells. As such, senescent cells are thought to be major contributors of inflammation. Theoretically, low, but chronic, levels of inflammation are drivers of age-related decline in function. Consistent with this theory, senescence and SASP are elevated in a number of fast-aging mouse models and, where tested, senescence clearance delays their decline in health.
  • senescent cells that display resistance to apoptosis and accumulate with age are targets of anti-aging research.
  • the main aim of such research has been to discover the molecular pathways that direct cells to senescence but not apoptosis and, eventfully, to develop agents that interfere with these pathways so that administration of such agents will induce apoptosis in a senescent cell in a safe and predictable manner.
  • the forkhead box (FOX) protein family comprises the FOX class O subfamily (FoxO) that has 4 mammalian members: Forkhead box protein 01 (FoxOl) (FKHR, FoxOl A), Forkhead box protein 03 (Fox03) (FKHRL1, Fox03A), Forkhead box protein 04 (Fox04) (AFX, AFXl, MLLT7), and Forkhead box protein 06 (Fox06).
  • FoxOs are transcription factors that play important roles in suppression of tumors.
  • Fox04 has a pivotal role in the cells’ direction to either senescence or apoptosis. Interfering with this key molecule Fox04 represents efficient way of blocking senescence and steering the cells’ fate towards apoptosis.
  • Senescence has been characterized to be a complex phenomenon that can be activated by distinct signals.
  • genotoxic activation of senescence has been characterized by the formation of DNA-SCARS (DNA Segments with Chromatin Alterations Reinforcing Senescence).
  • DNA-SCARS DNA Segments with Chromatin Alterations Reinforcing Senescence
  • Fox04 is recruited to DNA- SCARS that also includes tumor protein 53 (p53) as a major component.
  • Oncogenic BRAF mutation at V600E are encountered in ⁇ 7% of all human tumors with particularly enhanced occurrence in melanoma ( ⁇ 70%). Additionally, melanoma cells were found to generally have an elevated number of DNA-SCARS containing FOX04.
  • SCAPs Senescent Cell Anti- apoptotic Pathways
  • Dasatinib which acts on Dependence receptor/Src kinase/tyrosine kinase and target Primary human and mouse preadipocytes (adipose-derived stem cells), Quercetin (which acts on Bcl-2 family, p53/p21/serpine, & PI3K/AKT and target HUVECs, mouse bone marrow-derived mesenchymal stem cells), Navitoclax (which ascts on ABT263 and target MR-90 Cells, HUVECs), Piperlongumin (which acts on A1331852/ A1155463 and target IMR-90 Cells, HUVECs), and Fisetin (which acts on
  • One aspect of the present invention relates to a novel method of use for the 12 small molecules which are listed in the Table 1 and shown in Figures 1-12 as Senolytic Agents for treatment of senescence-associated diseases. These listed small molecules are normally used for other purposes other than to treat senescence-associated diseases and disorders. After extensive in-silico analysis, these small molecules were discovered to target the CR3 domain of Fox04, thus said small molecules are selected to be suitable to target senescent cells by blocking Fox04-p53 complex, and induce them to apoptosis.
  • Another aspect of the present invention relates to novel chemical structures which are derived from chemical scaffolds shown in the Figure 2. Not intending to be bound by theory, an extensive in silico analysis suggested that these selected 12 small molecules are able to interfere with SCAP by targeting the CR3 domain of Fox04.
  • the novel derivatives of said small molecules disclosed herein with a similar scaffold will still be able to integrfere with CR3 domain of Fox04 thereby interfering with SCAP. From here on the novel Senolytic Agents definition hereafter shall include the small molecules listed in Table 1 and their novel derivatives.
  • the novel Senolytic Agents of the present invention do not have to be continuously present to exert their effects. Brief disruption of Fox04 expressed pro-survival pathways is adequate to kill senescent cells.
  • the inventive treatment specifies that novel Senolytic Agents may be administered intermittently.
  • the novel Senolytic Agents trigger apoptosis of the senescent cells which in turn stimulate overall rejuvenation in a safe manner by reactivating the stem cell.
  • a method for selectively killing senescent cells and for treating senescence-associated diseases and disorders by administering the novel Senolytic Agents of the present invention to minimize the interaction between Fox04 and p53.
  • the method of use of the novel Senolytic Agents may comprise three different dose regimes and their variations, wherein each dose regime is expressed by distinct therapeutically effective amount and distinct administration frequencies. Each said dose regime is developed to treat different types of senescence-associated diseases and disorders. Killing senescent cells reduces the inflammatory SASP, and therefore reduces the chronic inflammation in the metabolism. Further, said treatment stimulates overall rejuvenation in a safe manner.
  • the diseases and disorders treatable with said novel Senolytic Agents include all diseases with inflammatory origin including diabetes, cardiovascular diseases, pulmonary diseases, osteoarthritis; senescence-associated ophthalmic diseases and disorders; and senescence-associated dermatological diseases and disorders.
  • diseases with inflammatory origin including diabetes, cardiovascular diseases, pulmonary diseases, osteoarthritis; senescence-associated ophthalmic diseases and disorders; and senescence-associated dermatological diseases and disorders.
  • the applicable regimes and individualization of the treatment for said diseases are described under corresponding headings within this application.
  • the Senolytic Small Molecules include those listed in Table 1 which are effective to induce apoptosis of senescent cells in a subject, such as a mammal, by inhibiting the action of Fox04 on p53.
  • the Senolytic Small Molecules may be designed to effectively block the CR3 domain of Fox04 from interfering with the DNA binding function of p53 that is phosphorylated at Serine 46, particularly, from interfering with the bulky FH domain of Fox04.
  • the Senolytic Small Molecules may be rationally designed to minimize their interaction with the DBD of p53, other FoxOs, and the DNA duplex containing a FoxO consensus binding.
  • a method for selectively inducing apoptosis of senescent cells and/or for treating a senescence-associated disease or disorder comprises administering one or more of the Senolytic Small Molecules which minimize the interaction between Fox04 and p53.
  • the method may comprise administering one or more of the Senolytic Small Molecule(s) via a treatment regime as disclosed herein or some variation thereof.
  • treatment regimes may vary as to administration frequency and/or dosing (for example, as to a dosage that will be therapeutically effective). Particular treatment regimes may be specifically developed to treat different types of senescence-associated diseases or disorders.
  • the diseases and disorders treatable via the Senolytic Small Molecules may include, but not limited to, all diseases with inflammatory origin including diabetes, cardiovascular diseases, pulmonary diseases, osteoarthritis; senescence-associated ophthalmic diseases and disorders; and senescence- associated dermatological diseases and disorders.
  • the applicable regimes and individualization of the treatment for said diseases are presented under corresponding headings within this application.
  • the Senolytic Small Molecules may not have to be continuously present to exert an effect.
  • brief disruption of pro-survival pathways, such as by administration of a Senolytic Small Molecule is adequate to kill senescent cells.
  • the Senolytic Small Molecules are suitable to be administered intermittently.
  • Figure 1 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 22863770. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 2 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 377845189. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 3 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 5390317. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 4 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 95566015. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 5 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 8402312. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 6 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 70664738. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 7 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 25725416. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 8 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 150421585. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 9 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 13513540. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 10 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 22074898. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 11 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 150421592. The snapshots were obtained from the last frame of the 200 ns MD simulations.
  • Figure 12 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 95608759. The snapshots were obtained from the last frame of the 200 ns MD simulations
  • FIG 13 A illustrates the dose in which the Senolytic Peptides shall be administered following guidance provided in the Impulse Regime.
  • the Therapeutically Effective Dose and frequency of administration may vary within this regime.
  • the Impulse Regime presents an optional follow up treatment course in which the Therapeutically Effective Dose may be increased or decreased.
  • Figure 13A depicts at least six different considerations: (1) Treatment Cycle (comprised of subject’s initial assessment, administration of Therapeutically Effective Dose and final assessment); (2) Therapeutically Effective Dose delivered in 1, 2, or 3 days; (3) Optional further treatment cycle(s) may be required if senescent cell reduction is unsatisfactory; (4) Senescence Clearance Interval 2-4 weeks; (5) Initial senescence assessment via biopsy samples; and (6) Final senescence assessment via biopsy samples.
  • Figure 13B illustrates the dose in which the Senolytic Peptides shall be administered following guidance provided in the Sustained Regime.
  • the Therapeutically Effective Dose and frequency of administration may vary within this regime.
  • the Sustained Regime presents an optional follow up treatment course in which the Therapeutically Effective Dose may be increased or decreased.
  • Figure 13B depicts: (1) Treatment Cycle (comprised of subject’s initial assessment, administration of Therapeutically Effective Dose and final assessment); (2) Therapeutically Effective Dose delivered in 1, 2, or 3 weeks; (3) Optional further treatment cycle(s) may be required if senescent cell reduction is unsatisfactory.
  • Therapeutically Effective Dose may be readjusted; (4) Senescence Clearance Interval 2-4 weeks; (5) Initial senescence assessment via biopsy samples; and (6) Final senescence assessment via biopsy samples.
  • Figure 13C illustrates the dose in which the Senolytic Peptides shall be administered following guidance provided in the Gentle Regimes.
  • the Therapeutically Effective Dose and frequency of administration may vary within this regime.
  • the Gentle Regime presents an optional follow up treatment course in which the Therapeutically Effective Dose may be increased or decreased.
  • Figure 13C depicts: (1) Treatment Cycle (comprised of subject’s initial assessment, administration of Therapeutically Effective Dose and final assessment); (2) Therapeutically Effective Dose delivered in 3 or 3 weeks; (3) Optional further treatment cycle(s) may be required if senescent cell reduction is unsatisfactory.
  • Therapeutically Effective Dose may be readjusted; (4) Senescence Clearance Interval 2-4 weeks; (5) Initial senescence assessment via biopsy samples; and (6) Final senescence assessment via biopsy samples.
  • Table 1 shows the binding affinities and the ligand affinities of the small molecules disclosed as Senolytic Agents in the application which are able to inhibit the CR3 domain of Foxo4, thereby inducing p53 dependent apoptosis.
  • Adjuvant refers to, but is not limited to, chemicals, small molecule, peptides such as the Senolytic Agent(s) including the Senolytic Small Molecule(s) or methods that are used prior to, in combination with or following the primary therapy which includes but not limited to surgery in order to enhance or modify the effect of the primary therapy, lower the risk of cancer recurrence and increase patient survival.
  • adjuvant therapy refers to, but not limited to, the therapy or therapies that are used prior to, in combination with; concomitant or concurrent therapy or following the primary therapy in order to enhance or modify the effect of the primary therapy which includes but not limited to surgery, lower the risk of cancer recurrence and increase patient survival.
  • the adjuvant therapy includes, but is not limited to, chemotherapy, radiation therapy, hormone therapy, targeted therapy, biological therapy or other novel therapies that will be recognized by those skilled in the art.
  • administering refers to a method of giving a dosage of a compound or composition to subject, such as a vertebrate or invertebrate, including a mammal, a bird, a fish, or an amphibian, via a suitable mode of administration, for example, intra-respiratory, topical, oral, intravenous, intraperitoneal, intramuscular, buccal, rectal, sublingual and/or intrathecal.
  • the preferred mode of administration can vary depending on various factors, such as the components being administered, the tissue site being targeted (e.g., a tissue in which the disease or disorder resides, is present, or is manifested which may be a tumor), the particular disease or disorder involved, and the severity of the disease or disorder.
  • Autoimmune disease(s) or disorder(s), refers to autoimmune diseases or disorders such, but not limited to, osteoporosis, psoriasis, oral mucositis, rheumatoid arthritis, inflammatory bowel disease, eczema, kyphosis, herniated intervertebral disc, and the pulmonary diseases, COPD, and idiopathic pulmonary fibrosis.
  • Bio sample refers to a biological sample which is obtained from a subject by invasive, non-invasive or minimally invasive methods, for example, a blood sample, a serum sample, a plasma sample, a biopsy specimen, body fluids (for example, lung lavage, ascites, mucosal washings, synovial fluid, vitreous fluid, or spinal fluid), bone marrow, lymph nodes, tissue explant, skin tissue sample, vaginal tissue, organ culture, or any other tissue or cell preparation from a subject.
  • body fluids for example, lung lavage, ascites, mucosal washings, synovial fluid, vitreous fluid, or spinal fluid
  • bone marrow lymph nodes
  • tissue explant skin tissue sample, vaginal tissue, organ culture, or any other tissue or cell preparation from a subject.
  • Cancer refers to, but is not limited to, cancers which are solid tumors or liquid tumors.
  • Solid tumors may include, for example, prostate cancer, testicular cancer, breast cancer, brain cancer (including glioblastoma) , pancreatic cancer, colon cancer, colorectal cancer, thyroid cancer, stomach cancer, lung cancer, ovarian cancer, Kaposi's sarcoma, skin cancer (including squamous cell skin cancer), renal cancer, head and neck cancers, throat cancer, squamous carcinomas (e.g., that form on the moist mucosal linings of the nose, mouth, throat, etc.
  • laryngeal and hypopharyngeal cancers such as laryngeal and hypopharyngeal cancers), bladder cancer, osteosarcoma (bone cancer), cervical cancer, endometrial cancer, esophageal cancer, liver cancer, hepatocellular carcinoma, and kidney cancer and further including the metastasis of melanoma cells, prostate cancer cells, testicular cancer cells, breast cancer cells, brain cancer cells, pancreatic cancer cells, colon cancer cells, thyroid cancer cells, stomach cancer cells, lung cancer cells, ovarian cancer cells, Kaposi's sarcoma cells, skin cancer cells, renal cancer cells, head or neck cancer cells, throat cancer cells, squamous carcinoma cells, bladder cancer cells, osteosarcoma cells, cervical cancer cells, endometrial cancer cells, esophageal cancer cells, liver cancer cells, or kidney cancer cells.
  • laryngeal and hypopharyngeal cancers bladder cancer
  • osteosarcoma bone cancer
  • cervical cancer endometrial cancer
  • Liquid tumors may include, for example, cancers occurring in blood, bone marrow, and lymph nodes and include generally, leukemias (myeloid and lymphocytic) including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), and hairy cell leukemia lymphomas (e.g., Hodgkin lymphoma), and melanoma, including multiple myeloma).
  • leukemias myeloid and lymphocytic
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • hairy cell leukemia lymphomas e.g., Hodgkin lymphoma
  • melanoma including multiple myeloma
  • CSC cancer stem cells
  • CSCs refers to, but is not limited to, cancer cells that are tumorigenic or tumor initiating.
  • CSCs possess properties of normal stem cells such as self-renewal to generate more CSCs and differentiation into different cell types to generate non- CSCs and increase tumor burden.
  • CSCs may be a distinct population of cells within a cancer that result in cancer progression, relapse, and/or metastasis.
  • CSCs may exhibit stem cell plasticity, where the cell-state transitions between a non-CSC and a CSC.
  • Cardiovascular disease(s) refers to, but is not limited to, angina, arrhythmia, atherosclerosis, cardiomyopathy, congestive heart failure, coronary artery disease (CAD), carotid artery disease, endocarditis, heart attack (coronary thrombosis, myocardial infarction [MI]), high blood pressure/hypertension, aortic aneurysm, brain aneurysm, cardiac fibrosis, cardiac diastolic dysfunction, hypercholesterolemia/hyperlipidemia, mitral valve prolapse, peripheral vascular disease (e.g., peripheral artery disease (PAD)), cardiac stress resistance, and stroke.
  • CAD coronary artery disease
  • MI myocardial infarction
  • chemotherapeutic agent(s) refers to a drug used to treat cancer.
  • chemotherapeutic agents include but is not limited to Tamoxifen, Femara, Herceptin, Letrozole, Taxol, Soltamox, Epirubicin, Trastuzumab, Leuprolide, Paclitaxel, Ellence, Pharmorubicin PFS, Neratinib, Nerlynx, Ogivri, Opdivo, Intron A, Sylatron, Yervoy, Interferon alfa-2b, Nivolumab, Proleukin, Pembrolizumab, dacarbazine, Keytruda, Aldesleukin, Lmlygic, Ipilimumab, DTIC-Dome, Temozolomide, Peginterferon alfa-2b, Talimogene laherparepvec, Mekinist, Tafmlar, Zelboraf, Tra
  • CTCs cancer tumor cells
  • CTCs can be marked by EPCAM, CK, or any other epithelial or mesenchymal specific marker.
  • CTCs can be marked by MUC4 or epithelial-cancer specific marker.
  • CTCs can be isolated from blood using antibody based separation, magnetic separation, size based separation, magnetic levitation or other means.
  • CTCs can be either negative for blood cell specific markers such as CD45, or positive for blood specific markers, also known as circulating hybrid cells (CHCs).
  • Dormant cells refers to, but is not limited to, cancer cells that are non-dividing and arrested in any state of the cell cycle.
  • Dormant cells can be present in the primary tumor, in micrometastases, in lymph nodes, in distant tissues, or in residual disease after treatment.
  • Dormant cells can reside within the body for any length of time and can be re activated at any time.
  • Dormant cells can also include cells just after extravasation to a metastatic site such as lung, liver, brain, bone, lymph node and all other tissues.
  • “Inflammatory or autoimmune disease(s) or disorder(s),” as used herein, refers to, but is not limited to, inflammatory diseases or disorders, such as by way of non-limiting example, osteoarthritis, or autoimmune diseases or disorders, such as by way of non-limiting example, osteoporosis, psoriasis, oral mucositis, rheumatoid arthritis, inflammatory bowel disease, eczema, kyphosis, herniated intervertebral disc, and the pulmonary diseases, COPD and idiopathic pulmonary fibrosis.
  • inflammatory diseases or disorders such as by way of non-limiting example, osteoarthritis, or autoimmune diseases or disorders, such as by way of non-limiting example, osteoporosis, psoriasis, oral mucositis, rheumatoid arthritis, inflammatory bowel disease, eczema, kyphosis, herniated intervertebral disc, and the
  • Neoadjuvant therapy refers to, but is not limited to, the treatment given as a first step to shrink a tumor before the main treatment, which is usually surgery, is given.
  • neoadjuvant therapy include chemotherapy, radiation therapy, hormone therapy or other novel therapies that will be recognized by those skilled in the art.
  • Primary therapy “Primary Treatment” as used herein, refers to but is not limited to the first treatment given for a disease which includes standard set of treatments, such as surgery followed by chemotherapy and radiation.
  • primary therapy is the one accepted as the best treatment. If it doesn’t cure the disease or it causes severe side effects, other treatment may be added in following therapies. Examples of the primary therapy includes but is not limited to, surgical therapy, radiotherapy, chemotherapy, immunotherapy, targeted therapy, hormone therapy, biological therapy or other novel therapies that will be recognized by those skilled in the art.
  • Pulmonary disease(s) and disorder(s), refers to, but is not limited to, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, bronchiectasis, and emphysema.
  • IPF idiopathic pulmonary fibrosis
  • COPD chronic obstructive pulmonary disease
  • asthma a chronic obstructive pulmonary disease
  • cystic fibrosis bronchiectasis
  • emphysema emphysema
  • Other dermatological diseases and disorders that may be associated with senescence include rhytides (wrinkles due to aging); pruritis (linked to diabetes and aging); dysesthesia (chemotherapy side effect that is linked to diabetes and multiple sclerosis); psoriasis (as noted) and other papulosquamous disorders, for example, erythroderma, lichen planus, and lichenoid dermatosis; atopic dermatitis (a form of eczema and associated with inflammation); eczematous eruptions (often observed in aging subjects and linked to side effects of certain small molecule).
  • Other dermatological diseases and disorders associated with senescence include cutaneous lymphomas, eosinophilic dermatosis (linked to certain kinds of hematologic cancers); reactive neutrophilic dermatosis; pemphigus, cutaneous lupus, pemphigoid and other immunobullous dermatosis fibrohistiocytic proliferations of skin.
  • “Senescence related diseases or disorders,” as used herein, refers to, but is not limited to, any of the following diseases or disorders a senescence-related disease or disorder, an inflammatory disease or disorder, an autoimmune disease or disorder, a cardiovascular disease or disorder, a pulmonary disease or disorder, an ophthalmic disease or disorder, a metabolic disease or disorder, a neurological disease or disorder, a senescence-associated dermatological disease or disorder, a nephrological disease or disorder, renal dysfunction, kyphosis, herniated intervertebral disc, frailty, hair loss, hearing loss, muscle fatigue, a gradual loss of function, or degeneration that occurs at the molecular, cellular, tissue, and organismal levels.
  • “Senolytic Agent(s),” as used herein, refers to, but is not limited to: Dasatinib, Quercetin, Navitoclax, Piperlongumin, Fisetin, BCL-XL inhibitors A1331852 and A1155463, all Fox04-related peptides and Foxo4-CR3 domain inhibiting peptides and Senolytic Small Molecule(s).
  • Senolytic Small Molecule(s) as used herein refers to any one of the compounds in listed in Table 1.
  • “Senolytic Therapy” as used herein refers to administration of Therapeutic Regimes in one or more treatments cycles for prevention or treatment of senescence- associated diseases and disorders and for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis as wherein Senolytic Agents are used alone or in combination or as an adjuvant, or for prevention, inhibition or retarding the progression of a Senescent Related Diseases or Disorders specifically including Cancer and or metastasis of a Cancer.
  • Treatment Regime(s) as used herein refers to administration of Senolytic Agents in Therapeutically Effective Dose in composition and in delivery methods and treatment cycles and regimes disclosed in the subject matter of this patent.
  • Treatment Regimes definition includes but not limited to regimes described in Schedule 1 and Impulse, Shock and Sustained regimes variations thereof.
  • “Therapeutically Effective Dose”, as used herein, refers to a dose which cause a therapeutic effect to some extent, some effect with respect to one or more of the symptoms of the disease, and includes curing a disease.“Curing” means that the symptoms of active disease are eliminated. However, certain long-term or permanent effects of the disease may exist even after a cure is obtained (such as where extensive tissue damage is present).
  • the Therapeutically Effective Dose is typically one which is sufficient to achieve the desired effect and may vary according to the nature and severity of the disease condition, and the potency of the particular Senolytic Small Molecule administered, its purity, and its composition (e.g. 90%, or 95% or 98% etc.).
  • рамно different doses may be employed for embodiments where the Senolytic Small Molecule(s) is administered for preventive use rather than for treatment of an active disease.
  • Therapeutically Effective Dose definition shall include the dosage for the Senolytic Agents as disclosed in Schedule 1.
  • Subject refers to a target of a treatment or therapy, including but not limited to, a human or a non-human mammal, a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate or a bird, for example, a chicken, as well as any other vertebrate or invertebrate.
  • Treatment refers to administration of a composition for therapeutic purposes and includes the Treatment Regimes as disclosed in the subject matter of the patent.
  • the FoxO family of proteins express similar domain compositions such that they contain DNA binding domains entitled as the Forkhead (FH) domain and C-terminal domains entitled as CR1, CR2 and CR3 for transactivation.
  • the FH and CR3 domains interact with each other and the binding surface of these interactions have been resolved in an NMR based study using Fox03.
  • FH includes basic (positively charged) and hydrophobic amino acids (R153, R154, W157 and G158) that contribute to this intramolecular interaction, while a central portion of CR3 that includes acidic (negatively charged) and hydrophobic amino acids (the amino acids from D623 to M633) interacts with the FH.
  • the FoxO proteins are very similar proteins that share high sequence similarity and domain composition. This similarity in the protein sequence was further observed at the functional level by in vivo experiments in mice. The resulting functional redundancy will suggest the validity of the findings on the Fox03 for Fox04.
  • blocking of the CR3 domain of Fox04 has the potential to inhibit the interaction between Fox04 and p53 and thus liberate p53 in senescent cells.
  • the CR3 domain of Foxo4 interacts with the DBD of p53 leading to inhibition of the transcriptional activity of p53 required for apoptosis.
  • blocking of CR3 domain of Foxo4 is critical to the efforts to release p53 DBD to induce apoptosis.
  • small molecules and/or peptides targeting the CR3 domain of Foxo4 can be used as Senolytic Agents.
  • ZINC 15 one of the largest virtual ligand databases, ZINC 15 that holds more than 1 billion compounds. Initially, the ZINC 15 database were screened to collect all of the positively charged compounds at neural pH owing to the high net negative charge of the CR3 domain. A total of 141,971, 146 different compounds were collected. This library was screened to identify the potential ligands with high binding affinity towards the CR3 domain of Foxo4.
  • the initial screenings allocated 30 million selected compounds from the library composed of -150 million compounds.
  • the initial screening was performed by docking. For this step, 30 million compounds were docked to 20 different conformations of the CR3 domain, which were obtained from molecular dynamics simulations performed by using two different force fields.
  • the top scoring 100 thousand ligands were progressed to next round of screening according to their docking scores.
  • 1 st round of free energy calculations was conducted by using the MM-GBSA method and top scoring 2500 ligands were selected. For these 2500 ligands, Amber scoring was carried out to select the top scoring 100 ligands. These 100 small molecules were extensively analyzed by molecular dynamics simulations for 200 ns.
  • the small molecules listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • the small molecules that carry a similar chemical scaffold to the ones listed in the Table 1 ( Figure 1-12) can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • the small molecules found in the ZINC 15 database will be screened in the successive virtual screening rounds by following the same procedure described herein, to treat senescence-associated diseases and disorders if they possess that strong binding affinities and high ligand efficiency values to effectively inhibit Fox04.
  • inhibitors of HIV Reverse transcriptase can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • small molecules that carry a similar chemical scaffold to one of the inhibitors of HIV Reverse transcriptase listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • inhibitor of reductases can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • small molecules that carry a similar chemical scaffold to one of the inhibitors of inhibitor of reductases listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence- associated diseases and disorders.
  • other anti-cancer agents can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • the small molecules that carry a similar chemical scaffold to one of other anti-cancer agents listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • the small molecules that carry a similar function and/or chemical scaffold to one of small molecules numbered 5, 6, 7, 8, 10, 11 of the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
  • Table 1 lists the small molecules that possess strong binding affinity toward the CR3 domain of Foxo4 and thus can be re-purposed as a senolytic.
  • methods for selectively inducing apoptosis in (e.g., killing) senescent cells in a subject who has a senescence-associated disease or disorder may generally comprise administering one or more of the Senolytic Small Molecule(s) to the subject in need thereof, for example, according to one or more of the administration methods described herein.
  • the method may comprise causing an artificial compound comprising one of the chemical scaffolds of the small molecules listed in the Table 1 to interfere with the CR3 domain of Forkhead box protein 04 (Fox04) of the senescent cell.
  • the proportion of senescent cells in a tissue of a mammal varies with the biological age and could vary substantially depending on the cohort that mammal subject belongs to. Moreover, the proportion of senescent cells present may further vary with the type of the tissue in a given subject. These variations may create a challenge in specifying a dose for the Senolytic Agent for a rejuvenation therapy. Additional complexities in specifying a dose arise when said senescent cell proportion is above a certain threshold and therefore accelerated apoptosis could result in frailty. Fortunately, unlike for cancer interventions, complete elimination of senescent cells may not be necessary for achieving beneficial effects.
  • the Senolytic Small Molecules may be administered via any suitable methodology.
  • any suitable methodology By way of non-limiting example, disclosed herein three different treatment regimes, namely an Impulse Regime, a Sustained Regime, and Gentle Regime via which the Senolytic Small Molecule(s) may be administered.
  • a method for treating a senescence-associated disease or disorder comprising administering to a subject in need thereof a Therapeutically Effective Dose of the Senolytic Small Molecule(s), for example, the Senolytic Small Molecule(s) may be administered intermittently in one or multiple treatment cycles. Each treatment cycle may extend over 1 or 2 or 3 days. Each administration may include equivalent doses adjusted so as to cumulatively reach the Therapeutically Effective Dose at the end of each treatment cycle. In some embodiments, the Therapeutically Effective Dose is administered equivalently in a single day or intermittently in two consecutive days or in three consecutive days or 2 or 3 administration in alternate days.
  • the Impulse Regime This regime of administration of the Senolytic Small Molecules is referred to herein as the Impulse Regime.
  • the Therapeutically Effective Dose is achieved through one or two subsequent administration(s).
  • after the first cycle there may be a two week-four-week senescence clearance interval, for example, allowing a period of time effective for a decrease in senescent cells.
  • the subject is evaluated, both before the Impulse Regime and after Senescence Clearance Interval period, by one skilled in the art to determine levels of various SASP markers for determination of the Therapeutically Effective Dose and Follow-up Treatment, respectively.
  • the Therapeutically Effective Dose is achieved through a single or multiple administrations in a period of 1-3 weeks.
  • the quantity of the Senolytic Small Molecule for each administration is equivalent and adjusted to cumulatively reach the Therapeutically Effective Dose at the end of the treatment.
  • This regime of administration of the Senolytic Small Molecules is referred to as the Sustained Regime.
  • the Therapeutically Effective Dose administered in the Sustained Regime may be higher than the Therapeutically Effective Dose as would be administered in the Impulse Regime.
  • the Therapeutically Effective Dose is administered intermittently in one or multiple treatment cycles wherein each treatment cycle comprised of 1 or 2 or 3 or 4 or 5 or 6 administration days equally distributed in 1-3 weeks where each administration is in equivalent doses adjusted to cumulatively reach the Therapeutically Effective Dose at the end of each treatment cycle.
  • the subject may be evaluated, both before the Sustained Regime and after Senescence Clearance Interval period, by one skilled in the art based on the levels of various SASP markers for determination of the Therapeutically Effective Dose and follow-up Treatment, respectively.
  • the Therapeutically Effective Dose is achieved through single or multiple administration cycles in the period of 3-4 weeks.
  • This regime of administration of the Senolytic Small Molecules is referred to herein as the Gentle Regime.
  • Both the Therapeutically Effective Dose and the quantity of the Senolytic Small Molecule for a single administration in the Gentle Regime may be lower than those in the Impulse Regime and Sustained Regime.
  • the Therapeutically Effective Dose may be administered intermittently in one or multiple treatment cycles. Each treatment cycle may be comprised of 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 administration days equally distributed in 3-4 weeks. Each administration may be in an equivalent dose adjusted so as to cumulatively reach the Therapeutically Effective Dose at the end of each treatment cycle.
  • the subject may be evaluated both before the Gentle Regime and after Senescence Clearance Interval period, by those skilled in the art based on the levels of various SASP markers for determination of the Therapeutically Effective Dose and follow-up Treatment, respectively.
  • the Therapeutically Effective Dose amount is delivered to a subject in need thereof by any one of several routes known to a person skilled in the art.
  • the Therapeutically Effective Dose is delivered orally, intravenously, intraperitoneally, by infusion (e.g., a bolus infusion), subcutaneously, enteral, rectal, intranasal, by injection, inhalation, buccal, sublingual, intramuscular, transdermal, intradermal, topically, intraocularly, vaginally, rectally, intrathecally or by intracranial injection, or by directly into the target tissue including a tumor or organ, or subcutaneous route or by some combination of the above thereof.
  • infusion e.g., a bolus infusion
  • the Therapeutically Effective Dose may be delivered in combinations of other Senolytic Agents and/or with chemotherapy. In some embodiments the Therapeutically Effective Dose may be delivered in combinations of other Senolytic Agents and/or with chemotherapy concomitantly or concurrently or not concurrently but in appropriate intervals.
  • a delivery method includes controlled or sustained release drug(s), drug- coated or permeated stents for which the drug is the Senolytic Small Molecule. In some embodiments Therapeutically Effective Dose amount is further adjusted depending on the delivery route and selected Treatment Regime.
  • examples 1-5 indicate that the Senolytic Small Molecule(s) can safely be administered at doses of 5 mg/Kg, 10 mg/Kg, and 15 mg/Kg. More particularly, Example 4 indicates that the Senolytic Small Molecule(s) is non-toxic at levels up to 100 mg/kg. Due to the molecular similarity of the various Senolytic Small Molecules in the Table 1 and compound possessing the same chemical scaffold thereto, atomistic structures are similar and therefore they exert their senolytic effects in a similar way. In some embodiments, the Therapeutically Effective Dose amount can further depend upon the patient's height, weight, sex, age and medical history.
  • treatment methods may include monitoring the population of the senescent cells on convenient biological samples taken from the subject at the beginning and 2 or 4 weeks after the treatment to determine the effectiveness of the therapy and or whether a second or a third treatment course is required.
  • an individualized treatment course may be implemented and may include monitoring of senescent cell population at the beginning of a therapy and a certain period after each treatment course and adjusting the treatment course or the dose or the treatment regime.
  • the administration of a Therapeutically Effective Dose may depend upon the subject’s state of health and subject’s response to the treatment throughout the period of the therapy.
  • the biological sample shall be the skin biopsy specimens obtained from skin tissue of the subject is collected with minimally invasive methods.
  • the detection of the senescent cells may be achieved using senescence associated markers
  • senescent cells diverge from other quiescent and terminally differentiated cells, they do not display a unique phenotype but a variety of phenotypes which define the senescent state. Hallmarks of the senescent cells in this state include permanent and irreversible growth arrest; increase in cell size; expression of senescence-associated beta-galactosidase (SA- b-Gal) enzyme resulting in increased lysosomal content; expression of a tumor suppressor, plg iNK4a j w0 hiomarkers have been extensively used for identification of senescent cells:
  • SA-P-Gal Enzyme activity assayed at pH 6 using X-Gal as the substrate.
  • p i 6 INK4a Monitoring the expression levels of pl6 INK4a protein which is a CDK4/6 inhibitor and is involved in maintenance of growth arrest.
  • the Senolytic Small Molecule(s) do not have to be continuously present to exert an intended effect. For example, brief disruption of pro-survival pathways is adequate to kill senescent cells and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells. Thus, the Senolytic Small Molecule(s) can be effective as Senolytic Small Molecules when administered intermittently.
  • Senolytic Agents such as the tyrosine kinase inhibitor (D) and the flavonoid, quercetin (Q), were shown to induce apoptosis in senescent cells.
  • D + Q Intermittent administration of D + Q alleviated frailty, neurological dysfunction, osteoporosis, and vertebral disk degeneration related to loss of glycosaminoglycans on an accelerated aging-like state. Furthermore, in mice with impaired mobility due to radiation of one of their legs 3 months previously, treadmill endurance improved within 4 days after completing a single course of D + Q. Said improvement persisted for at least 7 months. D + Q has an elimination half-life of a few hours. These outcomes following intermittent or single courses of agents with short elimination half-lives are consistent with the long-lasting type of effect expected from reducing senescent cell abundance, as opposed to what would be expected if D + Q had to be continuously present to suppress or activate cellular processes by occupying a receptor or acting on an enzyme.
  • At least one Senolytic Small Molecule may be administered in a Therapeutically Effective Dose with at least one or more other available Senolytic Agent which together act additively or synergistically to selectively kill senescent cells.
  • a Senolytic Small Molecule dose administration is optimization for use with the Senolytic Agents including but not limited to Dasatinib, Quercetin, Navitoclax, Piperlongumin, Fisetin, BCL-X L inhibitors A1331852 and A1155463, FOX04-related peptide, Foxo4-CR3 domain inhibiting peptides or other small molecules or combinations of these.
  • the effectiveness of the Senolytic Small Molecule treatment can be determined by a person skilled in the medical and clinical arts by employing combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein, may be used for monitoring the health status of the subject, in addition to the senescent cell monitoring described herein for the Treatment Regimes.
  • the effectiveness of the Senolytic Small Molecules when treating a subject for pulmonary diseases and disorders can be conducted. Pulmonary function tests, or PFTs, measure how well subject’s lungs work.
  • spirometry and lung volume tests include tests that measure lung size and air flow, such as spirometry and lung volume tests. Other tests measure how well gases such as oxygen get in and out of subject’s blood. These tests include pulse oximetry and arterial blood gas tests.
  • Another pulmonary function test called fractional exhaled nitric oxide (FeNO), measures nitric oxide, which is a marker for inflammation in the lungs. The subject may have one or more of these tests to diagnose lung and airway diseases, compare subject’s lung function to expected levels of function, monitor if your disease is stable or worsening, and see if said senolytic treatment is beneficial.
  • FeNO fractional exhaled nitric oxide
  • senescent cell population in the Biological skin sample of the subject undergoing treatment will be monitored before, and after a treatment course. In some embodiments, monitoring is performed during a treatment course and /or between the treatment courses or cycles.
  • the Senolytic Agent comprises any one of the Senolytic Small Molecules or combinations Senolytic Agents.
  • the Senolytic Agent is administered in a treatment window comprising 11 to 28 days.
  • the Senolytic Agent is administered daily or alternating days for 14 days followed by minimum 14 days off.
  • the Senolytic Agent is administered daily for 13 days followed by minimum 14 days off.
  • the Senolytic Agent is administered daily or alternating days for 12 days followed by minimum 14 days off. [000101] In some embodiments, the Senolytic Agent is administered daily for 11 days followed by minimum 14 days off.
  • the Senolytic Agent is administered daily or alternating days for 10 days followed by minimum 14 days off.
  • the Senolytic Agent is administered daily for 9 days followed by minimum 14 days off.
  • the Senolytic Agent is administered daily or alternating days for 8 days followed by minimum 12 days off.
  • the Senolytic Agent is administered daily for 7 days followed by minimum 12 days off.
  • the Senolytic Agent is administered daily or alternating days for 6 days followed by minimum 12 days off.
  • the Senolytic Agent is administered daily for 5 days followed by minimum 12 days off.
  • the Senolytic Agent is administered daily or alternating days for 4 days followed by minimum 12 days off.
  • the Senolytic Agent is administered daily for 3 days followed by minimum 10 days off.
  • the Senolytic Agent is administered daily for 2 days followed by minimum 10 days off.
  • the Senolytic Agent is administered for 1 day followed by minimum 10 days off.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 0. lmg/kg to 20 mg/kg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 0. lmg/kg to 15 mg/kg. [000114] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0.1 mg/kg to 10 mg/kg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 0. lmg/kg to 5 mg/kg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 0.1 mg/kg to 3 mg/kg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 0.1 mg/kg to 0.5 mg/kg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 1400 mg per day.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 1000 mg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 700 mg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 200 mg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 35 mg.
  • the Senolytic Agent is administered daily for 14 days in a dose of about 3.5 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 1400 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 1000 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 700 mg. [000127] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 200 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 35 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 1400 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 1000 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 700 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 200 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 35 mg.
  • the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg.
  • the Senolytic Agent is administered in one of the above doses daily for 1, 2, 3, 4, 5, or 6 days or 8, 9, 10, 11, 12, 13 days. Additionally, or alternatively, in some embodiments, the Senolytic Agent is administered in one of the above doses are administered alternative daily for 2, 4, or 6 days or 8, 10, 12, 14 days.
  • the doses described in this Schedule 1 shall be a Therapeutically sufficient dose for the administration of any one of the Senolytic Agents.
  • Therapeutically Effective Dose is administered by any one of several compositions and delivery routes known to a person skilled in the art and as disclosed in the subject matter.
  • the composition may be delivered orally, intravenously, intraperitoneally, by infusion (e.g., a bolus infusion), subcutaneously, enteral, rectal, intranasal, by injection, inhalation, buccal, sublingual, intramuscular, transdermal, intradermal, topically, intraocularly, vaginally, rectally, intrathecally, or intracranially, or by some combination thereof.
  • the regiment of administration in this Schedule 1 shall be the method of administration of any one of the Senolytic Agents.
  • the Therapeutically Effective Dose is delivered in methods of the disclosed subject matter including but limited to as in controlled or sustained release drug(s), drug-coated or permeated stents for which the drug is the Senolytic Agent.
  • Senolytic Small Molecule(s) is administered as described in this Schedule 1 together with another Senolytic Agent including but not limited to Navitoclax (ABT-263), Fisetin, A133185240, A115546340, Quercetin, Dasatinib, Piperlongumine, 17- AAG (tanespimycin), Geldanamycin 17-DMAG (alvespimycin), Famotidine, Deferoxamine, Mitoxantrane, Lapatinib, Neratinib with therapeutically sufficient doses for each of these Senolytic Agents but strictly in the regiments as described herein this Schedule 1.
  • Senolytic Therapy can be determined by a person skilled in the medical and clinical arts by employing combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein, may be used for monitoring the health status of the subject, in addition to the monitoring described herein for the treatment regimes.
  • the effectiveness of the Senolytic Small Molecule(s) with respect to treating a senescence-associated disease or disorder described herein can readily be determined by a person skilled in the medical and clinical arts.
  • One or any combination of diagnostic methods appropriate for the particular disease or disorder which methods are well known to a person skilled in the art, including physical examination, subject self-assessment, assessment and monitoring of clinical symptoms, performance of analytical tests and methods, including clinical laboratory tests, physical tests, and exploratory surgery, for example, may be used for monitoring the health status of the subject and the effectiveness of the Senolytic Small Molecule.
  • the effects of the methods of treatment described herein can be analyzed using techniques known in the art, such as comparing symptoms of subjects suffering from or at risk of a particular disease or disorder that have received the composition comprising the Senolytic Small Molecule with those of subjects who were not treated with the Senolytic Small Molecule or who received a placebo treatment.
  • a subject in need of treatment with the Senolytic Small Molecule(s) as described herein may be a human or may be a non-human primate or other animal (i.e., veterinary use) who has developed symptoms of a senescence cell-associated disease or disorder or who is at risk for developing a senescence cell-associated disease or disorder.
  • Non-human animals that may be treated include mammals, for example, non- human primates (e.g., monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, rabbits), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, bovine, elephants, bears and other domestic, farm, and zoo animals.
  • rodents e.g., rats, mice, gerbils, hamsters, ferrets, rabbits
  • lagomorphs e.g., swine (e.g., pig, miniature pig)
  • swine e.g., pig, miniature pig
  • equine canine
  • feline bovine
  • elephants bears and other domestic, farm, and zoo animals.
  • administering can prolong survival when compared to expected survival if a subject were not receiving treatment.
  • Subjects in need of treatment include those who already have the disease or disorder as well as subjects prone to have or at risk of developing the disease or disorder, and those in which the disease, condition, or disorder is to be treated prophylactically.
  • a subject may have a genetic predisposition for developing a disease or disorder that would benefit from clearance of senescent cells or may be of a certain age wherein receiving the Senolytic Small Molecule would provide clinical benefit to delay development or reduce severity of a disease, including an age-related disease or disorder.
  • use of the Senolytic Small Molecules may be restricted during wound healing (including pre- or post-operations).
  • senescent cells may be induced around the wound.
  • Senescent cells make growth factors that are required for wound healing.
  • this innate mechanism is not disturbed unless the Senolytic Small Molecule(s) is administered at the time of the wound healing.
  • Cellular senescence is a cell fate that involves essentially irreversible replicative arrest, apoptosis resistance, and frequently increased protein synthesis, metabolic shifts with increased glycolysis, decreased fatty acid oxidation, increased reactive oxygen species generation, and acquisition of a senescence-associated secretory phenotype (SASP).
  • SASP senescence-associated secretory phenotype
  • the methods of use of Senolytic Agents for treatment of senescence-associated diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment of senescence-associated diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • Senolytic Therapy for treatment of senescence-associated diseases and disorders
  • Certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • the Senolytic Small Molecule(s) inhibits senescence of adult stem cells or inhibits accumulation, kills, or otherwise facilitates removal of adult stem cells that have become senescent. Therefore, the Senolytic Small Molecules may also be useful for treating or preventing of an age-related disease or disorder that occurs as part of the natural aging process or that occurs when the subject is exposed to a senescence inducing agent or factor (e.g., irradiation, chemotherapy, smoking tobacco, high-fat/high sugar diet, other environmental factors).
  • a senescence inducing agent or factor e.g., irradiation, chemotherapy, smoking tobacco, high-fat/high sugar diet, other environmental factors.
  • frailty as an aging-associated decline may be treated or prevented (i.e., the likelihood of occurrence of is reduced) by administering the Senolytic Small Molecule.
  • the effectiveness of the senolytic therapy can be measured by monitoring the frailty index of the patient.
  • the age related disease or disorder is scoliosis. Effectiveness of the senolytic therapy are measured by, inter alia, physical examination of spine, ribs, hips and shoulders and/or X-RAY, CT and/or MRI to determine bone curvature.
  • the methods of use of Senolytic Agents for treatment of general age-related diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of said Treatment Regimes for age-related diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells on a biopsy sample taken from the subject at the beginning and throughout the period of the therapy and thus monitoring the target cell population decline.
  • CTCs circulating tumor cells
  • the effectiveness of a method of treatment described herein may be manifested by reducing the number of symptoms of an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus, decreasing the severity of one or more symptoms, or delaying the progression of an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus.
  • preventing an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus refers to preventing (i.e., reducing the likelihood of occurrence) or delaying onset of an age-related disorder or age- sensitive trait associated with a senescence-inducing stimulus, or reoccurrence of one or more age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus.
  • a Senolytic Agent could influence the health span of mice in which senescence and the concomitant loss of tissue homeostasis were allowed to develop spontaneously as a consequence of aging.
  • the results of in vivo mouse experiments indicate that a Senolytic Agent can reduce cellular senescence and counteract hair loss (objectively measured as the fur density) and general frailty (objectively measured by increased running wheel activity or muscle size increase) in mice.
  • Tissues contain high levels of senescent cells, which due to chronic SASP secretion, would inflict permanent reprogramming on their neighboring cells. Senescence was recently shown to trigger tissue reprogramming in vivo, leading to Nanog-positive cells in the vicinity of areas of senescence. Senescent cells might thus trigger reprogramming of neighboring cells into more pluripotent cells. However, since the release of SASP factors (such as IL6) is continuous, they would effectively make this change permanent and keep their neighboring recipient cells locked in this stem like state. If the number of senescent cells are reduced to that of relatively young tissues (with fewer senescent cells) then a transient SASP response, causing temporary cell reprogramming and subsequent proliferation/differentiation responses would be able to replenish damaged and lost cells.
  • SASP factors such as IL6
  • the Senolytic Agents is applied in Treatment Regimes for tissue rejuvenation therapy of a scalp tissue or an osteoarthritic joint or a pulmonary tissue or a renal tissue.
  • senolytic Agents is administered in Treatment Regimes for scalp treatment and hair regeneration by applying the senolytic tissue externally.
  • the methods of use of Senolytic Agents for tissue rejuvenation comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for tissue rejuvenation include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • Certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • Senescence-associated diseases or disorders treatable by administering the Senolytic Agents include dermatological diseases or disorders.
  • dermatological diseases or disorder are associated with the accumulation of senescent cells.
  • said dermatological diseases and disorders are psoriasis, vitiligo and eczema.
  • vitiligo is an acquired disorder characterized by depigmentation.
  • oxidative stress and underlying premature melanocyte senescence are considered to be key factors in vitiligo progression. Melanocytes from non-lesion vitiligo lesions were shown to exhibit a pre-senescent phenotype in vitro.
  • the methods of use of Senolytic Agents for treatment of dermatological diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Agents for treatment of dermatological diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • Senolytic Small Molecules could be external on the surface of the body such that skin cell penetration of said molecule is achieved as described in detail in the disclosed subject matter.
  • Chronic inflammation is a major factor in wide range of disease associated with old age. Recently a global clinical trial of 10,000 subjects who had previous heart attacks of an anti inflammatory drug which targeted a portion of the inflammatory pathway — focusing specifically on interleukin- lbeta (IL-Ib), a cell-signaling protein - showed that it reduced their risk of further heart attacks or strokes. The drug prevents these cells from going into overdrive, but presumably leaves the remaining immune system intact.
  • the chronic inflammatory diseases include rheumatoid arthritis when inflammation occurs in joints. Individuals with Rheumatoid arthritis exhibit accelerated immunosenescence possibly as a result of inflammatory mechanisms. Therefore, the Senolytic Agents are suitable for curing or managing chronic inflammation by substantially stopping SASP by killing the senescent cells.
  • the senescence-associated secretory phenotype comprises a range of different proteins, including several proteins known to play a role in aging and age-related diseases, including chemokines such as CCL2 and CLL11 and prominent interleukins such as IL-1, IL-6 and IL-12. When above a certain threshold, such factors can significantly impair tissue function and impair functioning of neighboring cells. As such, senescent cells are thought to be major contributors of inflammation; a theory stating that low, but chronic, levels of inflammation are drivers of age-related decline.
  • the definition of chronic inflammatory diseases includes osteoarthritis which is characterized by progressive tissue remodeling and loss of joint function and paralleled by increased age. It is the most prevalent disease of the synovial joints. During osteoarthritis, levels of various senescence markers increase in chondrocytes with SASP profiles similar to classical senescent cells which in turn supports the hypothesis that senescence of cells within joint tissues may play a pathological role in the causation of osteoarthritis. Therefore, the Senolytic Agents is suitable to cure or manage chronic inflammation by substantially stopping SASP by killing the senescent cells.
  • administering is effective in the treatment of osteoarthritis.
  • the osteoarthritis disease parameters which are measured include, inter alia, joint pain, redness, stiffness and/or swelling and joint motion range, X-RAY and/or MRI for bone spurs, blood tests and joint fluid analyses to rule out other causes.
  • administering is effective in the treatment of kyphosis.
  • the kyphosis disease parameters which are measured include, inter alia, measurement of spine curvature by X-RAY, CT and/or MRI.
  • the methods of use of Senolytic Agents for treatment of inflammatory or autoimmune diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment of inflammatory or autoimmune diseases and disorder include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • Senolytic Therapy for treatment of inflammatory or autoimmune diseases and disorders
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • the senescence-associated disease or disorder treated by the methods described herein is a cardiovascular disease.
  • the cardiovascular disease is often caused by atherosclerosis and is the primary cause of mortality in the developed countries.
  • Atherosclerosis is a disease of major arteries in which high levels of low-density lipoprotein bearing oxidative modifications accumulate in vessel walls, attracting phagocytic immune cells to form plaques. Telomere shortening and oxidative stress caused by smooth-muscle proliferation and declining levels of endothelial nitric oxide synthase during plaque formation and expansion cause senescence induction. Human and mouse atheromas have been reported to exhibit senescent vascular smooth muscle and endothelial cells.
  • the Senolytic Agents administered as Treatment Regimes are suitable to slow down the progression of cardiovascular disease by reducing the chronic inflammation in the body substantially by stopping SASP.
  • (cardio)vascular disease parameters which are measured include, inter alia, cardiac ejection fraction, blood vessel stiffness and blood pressure.
  • Senolytic Therapy is effective in treatment of atherosclerosis.
  • atherosclerosis disease parameters which are measured include blood tests including measurements of cholesterol, glucose, electrocardiogram, angiography, computerized tomography scan and/or ophthalmoscopy.
  • the methods of use of Senolytic Agents for treatment of cardiovascular diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment of cardiovascular diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a cardiovascular disease e.g., atherosclerosis
  • a cardiovascular disease e.g., atherosclerosis
  • Health status of the subject may be monitored by one or any combination of diagnostic methods, including but not limited to physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein and practiced in the art (e.g., angiography, electrocardiography, stress test, non-stress test).
  • the effects of the treatment of the Senolytic Small Molecule can be analyzed using techniques known in the art, such as comparing symptoms of subjects suffering from or at risk of cardiovascular disease that have received the treatment with those of subjects without such a treatment or with placebo treatment.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • the senescence-associated disease or disorder treated by the Senolytic Therapy methods described herein is a Pulmonary Diseases and Disorders.
  • Pulmonary Disease and Disorders treated by administration of Senolytic Therapy described herein include Chronic obstructive pulmonary disease (COPD)/emphysema characterized by lung inflammation induced by accelerated lung aging involving inflammatory mediators such as tumor necrosis factor alpha, interleukin- 1, interleukin-6, reactive oxygen species and proteases.
  • COPD chronic obstructive pulmonary disease
  • Mechanisms involved in COPD include telomere shortening, cellular senescence, activation of PI3 kinase-mTOR signaling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence, and a low-grade chronic inflammation. Therefore, the Senolytic Small Molecule(s) is suitable for treatment of premature senescence involved in Pulmonary Diseases or Disorders by removing the senescent cells in controlled and safe manner.
  • Pulmonary Disease and Disorders treated by Senolytic Therapy described herein include Idiopathic Pulmonary Fibrosis (IPF).
  • IPF is the most common and severe idiopathic interstitial pneumonia.
  • familial interstitial pneumonia the telomerase complex is affected by the genetic lesions present eventually leading to telomere shortening in both leukocytes and pulmonary tissue which is also observed in sporadic IPF.
  • Pathology of IPF points out to a mechanism with the involvement of cellular senescence in disease progression.
  • Senolytic Therapy provided herein is suitable for treating or preventing (i.e., reducing the likelihood of occurrence of pulmonary disease or disorder by killing senescent cells associated with the disease or disorder) particularly the senescence of pulmonary artery-smooth muscle cells in a subject who has the disease or disorder.
  • the Senolytic Therapy is effective in the treatment of lung emphysema.
  • the effectiveness of the therapy can be established by taking certain measurements which include but not limited to, inter alia, breathlessness, chest size, lung volume, decreased breath sounds through the stethoscope, fingertip shape, style of breathing, hypoxemia, hypercaria, cyanosis, malnutrition lung volume, lung ejection capacity, dead volume in the lungs, airflow changes after bronchodilator medication, chest X-RAY and CT scan of the chest and red blood cell counts.
  • the Senolytic Therapy is effective in the treatment of COPD.
  • the senolytic therapy of the COPD disease parameters include, inter alia, spirometry and lung functional tests as described for lung emphysema, including breathlessness, chest size, decreased breath sounds through the stethoscope, fingertip shape, style of breathing, hypoxemia, hypercaria, cyanosis, malnutrition, lung volume, lung ejection capacity, dead volume in the lungs, airflow changes after bronchodilator medication, chest X- RAY and CT scan of the chest and red blood cell counts.
  • the methods of use of Senolytic Agents for treatment of Pulmonary Disease and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment of Pulmonary Disease and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subj ecf s progress can be monitored appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subject at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • Senolytic Small Molecule(s) is suitable to slow down the progression of neurodegeneration by reducing the chronic inflammation in the body substantially by stopping SASP.
  • the method of use of Senolytic Agents for senescence- associated neurological diseases or disorders comprises Treatment Regimes as disclosed in the subject matter of this patent.
  • the methods of use of Senolytic Agents for treatment of senescence-associated neurological diseases or disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Senolytic Therapy as disclosed in the subject matter of this patent.
  • Treatment Regimes of Senolytic Therapy for treatment of senescence-associated neurological diseases or disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • Senolytic Therapy for treatment of senescence-associated neurological diseases or disorders
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • the effectiveness of Senolytic Therapy on senescence- associated neurological diseases or disorders, as administered in Treatment Regimes described herein, can be established by monitoring of a subject by a person skilled in the medical and clinical arts.
  • one or any combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein are used for monitoring the effectiveness of said treatment.
  • the effects of administering one or more Senolytic Small Molecules can be further analyzed using techniques known in the art, such as comparing symptoms of subjects suffering from or at risk of Alzheimer's disease that have received the treatment with those of subjects without such a treatment or with placebo treatment.
  • the Senolytic Agent(s) based Senolytic Therapy is effective in the effective in the treatment of Alzheimer's disease.
  • the Alzheimer’s disease parameters which are measured include, inter alia, changes in ability to carry out daily activities, and changes in behavior and personality, tests of memory, problem solving, attention, counting, and language, blood and urine tests, brain scans, such as computed tomography (CT), magnetic resonance imaging (MRI), or positron emission tomography (PET) and/or biomarker analysis.
  • CT computed tomography
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • the Senolytic Therapy is effective in the treatment of Parkinson's disease.
  • Parkinson’s disease parameters which are measured include, inter alia, analysis for tremors, limb or neck stiffness, general fitness and balance and/or locomotor function.
  • the Senolytic Agent(s) based Senolytic Therapy is effective in the treatment of depression.
  • Depression parameters that are to be measured are, inter alia, physical examination, sadness or depressed mood most of the day, major changes in weight, insomnia or excessive sleep, fatigue or loss of energy most of the day, feelings of hopelessness or worthlessness or excessive guilt, problems with concentration or decision making, recurring thoughts of death or suicide.
  • Senescence-associated diseases or disorders treatable by administering the Senolytic Small Molecule as described in Treatment Regimes herein include ophthalmic diseases or disorders.
  • ophthalmic diseases and disorders include but not limited to age-related macular degeneration, cataracts, glaucoma, vision loss, presbyopia.
  • Ophthalmic Diseases or Disorders involve age related macular degeneration (AMD) resulting in irreversible blindness which is associated with the degradation of retinal pigment epithelium (RPE) cells, photoreceptors, and choriocapillaris.
  • AMD age related macular degeneration
  • Oxidative stress, inflammation (IL-17 involvement) and some genetic factors are known to be involved in AMD pathogenesis. Oxidative stress can induce DNA damage response (DDR), autophagy, and cell senescence.
  • DDR DNA damage response
  • autophagy autophagy
  • cell senescence include DNA damage response (DDR), autophagy, and cell senescence.
  • the Senolytic Small Molecule(s) is suitable for treatment of premature senescence involved Ophthalmic Diseases and Disorders by removing the senescent cells in controlled and safe manner.
  • Treatment Regimes provided herein for treating or preventing i.e., reducing the likelihood of occurrence of; delaying the onset or development of, or inhibiting, retarding, slowing, or impeding progression or severity of
  • an ophthalmic disease, disorder, or condition e.g., presbyopia, cataracts, macular degeneration
  • the methods of use of Senolytic Agents for treatment of ophthalmic diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of said Treatment Regimes of Senolytic Agents for treatment of ophthalmic diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • Senescence-associated diseases or disorders treatable or preventable by administering the Senolytic Therapy include metabolic diseases or disorders. Said senescence- associated diseases and disorders include diabetes, metabolic syndrome, diabetic ulcers, and obesity.
  • Diagnosis of type 2 diabetes is based on symptoms (e.g., increased thirst and frequent urination, increased hunger, weight loss, fatigue, blurred vision, slow-healing sores or frequent infections, and/or areas of darkened skin), medical history, and/or by taking measurements which include, inter alia, basal blood glucose levels, average blood glucose levels over a period of time (2-3 months; AIC test), fasting plasma glucose, oral glucose tolerance test, plasma glucose test N.B. physical examination of a subject.
  • symptoms e.g., increased thirst and frequent urination, increased hunger, weight loss, fatigue, blurred vision, slow-healing sores or frequent infections, and/or areas of darkened skin
  • medical history e.g., increased thirst and frequent urination, increased hunger, weight loss, fatigue, blurred vision, slow-healing sores or frequent infections, and/or areas of darkened skin
  • measurements which include, inter alia, basal blood glucose levels, average blood glucose levels over a period of time (2-3
  • Senolytic Therapy can readily be determined by a person skilled in the medical and clinical arts.
  • diagnostic methods including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods, such as those described herein, may be used for monitoring the health status of the subject.
  • a subject who is receiving Senolytic Therapy for treatment or prophylaxis of diabetes can be monitored, for example, by assaying glucose and insulin tolerance, energy expenditure, body composition, fat tissue, skeletal muscle, and liver inflammation, and/or lipotoxicity (muscle and liver lipid by imaging in vivo and muscle, liver, bone marrow, and pancreatic b-cell lipid accumulation and inflammation by histology).
  • Other characteristic features or phenotypes of type 2 diabetes are known and can be assayed as described herein and by using other methods and techniques known and routinely practiced in the art.
  • Obesity and obesity-related disorders are used to refer to conditions of subjects who have a body mass that is measurably greater than ideal for their height and frame.
  • Body Mass Index (BMI) is a measurement tool used to determine excess body weight, and is calculated from the height and weight of a subject.
  • a human is considered overweight when the person has a BMI of 25-29; a person is considered obese when the person has a BMI of 30-39, and a person is considered severely obese when the person has a BMI of 40.
  • the terms obesity and obesity-related refer to human subjects with body mass index values of greater than 30, greater than 35, or greater than 40.
  • abdominal obesity A category of obesity not captured by BMI is called “abdominal obesity” in the art, which relates to the extra fat found around a subject's middle, which is an important factor in health, even independent of BMI.
  • the simplest and most often used measure of abdominal obesity is waist size.
  • abdominal obesity in women is defined as a waist size 35 inches or higher, and in men as a waist size of 40 inches or higher.
  • More complex methods for determining obesity require specialized equipment, such as magnetic resonance imaging or dual energy X-ray absorptiometry machines.
  • a condition or disorder associated with diabetes and senescence is a diabetic ulcer (i.e., diabetic wound).
  • An ulcer is a breakdown in the skin, which may extend to involve the subcutaneous tissue or even muscle or bone. These lesions occur, particularly, on the lower extremities.
  • Patients with diabetic venous ulcer exhibit elevated presence of cellular senescence at sites of chronic wounds. Chronic inflammation is also observed at sites of chronic wounds, such as diabetic ulcers suggesting that the proinflammatory cytokine phenotype of senescent cells has a role in the pathology.
  • Metabolic syndrome in humans is typically associated with obesity and characterized by one or more of cardiovascular disease, liver steatosis, hyperlipidemia, diabetes, and insulin resistance.
  • a subject with metabolic syndrome may present with a cluster of metabolic disorders or abnormalities which may include, for example, one or more of hypertension, type-2 diabetes, hyperlipidemia, dyslipidemia (e.g., hypertriglyceridemia, hypercholesterolemia), insulin resistance, liver steatosis (steatohepatitis), hypertension, atherosclerosis, and other metabolic disorders.
  • the diabetes type II disease is effectively treatable or preventable by administering the Senolytic Therapy.
  • the efficacy of the Treatment Regimes is established by taking measurements which-include, inter alia, basal blood glucose levels, average blood glucose levels over a period of time (2-3 months; AIC test), fasting plasma glucose, oral glucose tolerance test, plasma glucose test N.B.
  • the obesity disorder is effectively treatable or preventable by administering the Senolytic Therapy.
  • the obesity parameters which are measured include alia body weight, Body-Mass-Index (BMI), waist circumference, waist-to-hip ratio, skinfold thicknesses, and bioelectrical impedance, magnetic resonance imaging and/or dual energy X-ray absorptiometry.
  • the metabolic syndrome is treatable or preventable effectively by administering the Senolytic Therapy.
  • the metabolic syndrome disease parameters which are measured include, inter alia, measurements for obesity (see above, e.g. waist circumference), blood levels of triglycerides, HDL cholesterol, blood pressure, fasting glucose.
  • the hepatic insufficiency is treatable or preventable effectively by administering the Senolytic Therapy.
  • the hepatic insufficiency disease parameters which are measured include, inter alia, blood AST and ALT values.
  • cirrhosis disease is treatable or preventable effectively by administering the Senolytic Therapy.
  • the cirrhosis disease parameters which are measured include, inter alia, measurements of blood-clotting factors and international normalized ratio for blood clotting, liver stiffness by magnetic resonance elastography, liver imaging by CT and/or MRI, physical examination, blood testing for bilirubin and creatinine, and/or liver biopsy analysis for liver damage.
  • the methods of use of Senolytic Agents for treatment or prevention of metabolic diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment or prevention of metabolic diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • Nephrological pathologies arise in the elderly. Glomerulonephritis is characterized by inflammation of the kidney and by the expression of two proteins, ILla and I L I b . ILla and IL l b are considered master regulators of SASP. Glomerular disease is associated with elevated presence of senescent cells, especially in fibrotic kidneys. Therefore, the Senolytic Small Molecule(s) is suitable to renal dysfunction by substantially stopping SASP by killing the senescent cells.
  • renal insufficiency is effectively treatable or preventable by administering the Senolytic Therapy.
  • the renal insufficiency disease parameters which are measured include, inter alia, blood pressure, heart/lung sound analysis, nervous system exam, urinalysis for protein content, analysis for creatinine clearance and level of Blood Urea Nitrogen, CT, MRI and/or ultrasound of abdomen and kidneys, kidney biopsy for damage analysis.
  • Glomerulosclerosis is another pathology associated with renal aging supported by the accumulation of senescent cells as indicated by an increase in the levels of senescence markers such as pl6 and SA-P-Gal by aging.
  • renal dysfunction caused by nephrological pathologies such as glomerulosclerosis is effectively treatable or preventable by administering the Senolytic Therapy.
  • the glomerulosclerosis disease parameters which are measured include, inter alia, swellings in limbs, weight gains, changes in urine due to proteinuria, distortion or compression of the small capillaries in the glomerulus that filter blood in a biopsy and plasma Urea or protein concentration, blood pressure, glomerular filtration rate, and/or kidney ultrasound.
  • the methods of use of Senolytic Agents for treatment of renal dysfunction and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment of renal dysfunction and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • SIPS Stress-induced premature senescence
  • chemotherapeutic small molecule and ionizing radiation Both stresses cause substantial collateral macromolecular damage to non-neoplastic cells and responsible for the early aging phenotypes frequently observed in cancer survivors. Contrary to chronic senescence resulting from normal aging mechanisms and declining macromolecular repair mechanisms, therapy-induced senescence results from abrupt exogenous stresses placed on tissues during cancer therapy.
  • the Senolytic Therapy may be administered to the subjects who may also have cancer, as an adjuvant for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis using the methods as described herein.
  • Senolytic Therapy is selected the primary therapy for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis.
  • Senolytic Therapy as an adjuvant therapy includes, but is not limited to, chemotherapy, radiation therapy, hormone therapy, targeted therapy, or biological therapy or other novel therapies that will be recognized by those skilled in the art.
  • Senolytic Therapy as an adjuvant for cancer therapy is administered in combination with chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s) used in primary therapy or following a primary therapy for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis
  • the methods of use of Senolytic Agents for inhibition or retarding the progression of metastasis in a subject who has a cancer comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Senolytic Therapy as disclosed in the subject matter of this patent.
  • Treatment Regimes of Senolytic Therapy for inhibition or retarding the progression of metastasis include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • Such the Senolytic Small Molecule when administered in a Therapeutically Effective Dose to a subject who has a cancer according to the methods described herein may inhibit tumor proliferation.
  • the methods of use of Senolytic Agents for targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells in a subject who has a cancer comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for targeting circulating tumor cells (CTCs) include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • Some examples of Treatment Regimes of Senolytic Therapy for targeting cancer stem cells include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • Treatment Regimes of Senolytic Therapy for targeting dormant cells include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • the Senolytic Therapy for targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells of cancer certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • Such the Senolytic Small Molecule when administered in a Therapeutically Effective Dose to a subject who has a cancer according to the methods described herein may inhibit tumor proliferation.
  • the methods described herein are also applicable for inhibiting, retarding or slowing progression of metastatic cancer, or targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells of any one of the types of tumors described in the medical art.
  • CTCs circulating tumor cells
  • multiple Senolytic Therapies are further customized and applied contemporaneously to the same patient for inhibiting, retarding or slowing progression of cancer tumor and/or metastasis and/or for targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells.
  • a Senolytic Agent is injected directly into a cancer tumor for inhibiting, retarding or slowing progression of the tumor and/or targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells and contemporaneously same or a separate Senolytic Agent is administered as a Senolytic Therapy via intravenous or intraperitoneal delivery method for inhibiting, retarding or slowing progression of metastasis and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells.
  • CTCs circulating tumor cells
  • CTCs circulating tumor cells
  • CTCs circulating tumor cells
  • dormant cells and/or cancer stem cells contemporaneously same or a separate Senolytic Agent is administered as a Senolytic Therapy via intravenous or intraperitoneal delivery method for inhibiting, retarding or slowing progression of metastasis and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells.
  • the cancer type is but not limited to metastatic melanoma, resistant breast cancer, radiotherapy resistant glioblastoma, colorectal cancer, or thyroid cancer.
  • administering are effective in the treatment of metastatic melanoma or as an adjuvant drug.
  • the metastatic melanoma disease parameters which are measured include, inter alia, a reduction in tumor size and/or metastasization by applying Treatment Regimes as disclosed in the subject matter of this patent i.e. by administering the Senolytic Agents’ Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • the methods of use of Senolytic Agents for the treatment of resistant breast cancer as adjuvants comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for treatment of resistant breast cancer as adjuvants include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • administering is effective in the treatment of resistant breast cancer or as an adjuvant drug.
  • the resistant breast cancer disease parameters which are measured include a reduction in tumor size and/or metastasization.
  • Senolytic Therapy is the primary therapy for treatment of the resistant breast cancer which involves one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s).
  • the primary therapy agent used for the treatment of the resistant breast cancer is a chemotherapeutic agent(s).
  • Said chemotherapeutic agent(s) include but are not limited to, Tamoxifen, Femara, Herceptin, Letrozole, Taxol, Soltamox, Epirubicin, Trastuzumab, Leuprolide, Paclitaxel, Ellence, Pharmorubicin PFS, Neratinib, Nerlynx, Ogivri and their derivatives.
  • the Senolytic Therapy is directed as an adjuvant therapy for the treatment of the resistant breast cancer, during or following Primary Therapy wherein Senolytic Agent is used in combination with at least one Chemotherapeutic Agent.
  • Senolytic Therapy is effective in treatment of a resistant glioblastoma or as an adjuvant drug.
  • the resistant glioblastoma disease parameters which are measured include, inter alia, a reduction in tumor size and/or metastasization.
  • the methods of use of Senolytic Agents for the treatment of resistant glioblastoma as adjuvants comprises Senolytic Therapy as disclosed in the subject matter of this patent.
  • Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for the treatment of resistant glioblastoma as adjuvants include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • Senolytic Therapy is used as the primary therapy used for treatment of the resistant glioblastoma and involves one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s).
  • Said chemotherapeutic agent(s) include but are not limited to, Temodar, Avastin, Temozolomide, Matulane, Bevacizumab, BiCNU, Gliadel, Carmustine, Hydroxyurea, Procarbazine, Mvasi and their derivatives.
  • the Senolytic Agents are used in adjuvant therapy for the treatment of the resistant glioblastoma, in combination with or following primary therapy with at least one chemotherapeutic agent. Said chemotherapeutic agent(s) could be used alone or in combination with the Senolytic Agents.
  • administration of Senolytic Therapy is effective in the treatment of colorectal cancer or as an adjuvant drug.
  • the colorectal cancer disease parameters which are measured include a reduction in tumor size and/or metastasization.
  • the methods of use of Senolytic Agents for the treatment of colorectal cancer as adjuvants comprises Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for the treatment of colorectal cancer, wherein Senolytic Agents are used as adjuvants include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • the Senolytic Therapy for the treatment of colorectal cancer, wherein Senolytic Agents are used as adjuvants
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • the Senolytic Therapy is used as the primary therapy used for treatment of colorectal cancer which involves one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s).
  • Said chemotherapeutic agent(s) include but are not limited to, Xeloda, Oxaliplatin, Avastin, Fluorouracil, Leucovorin, Capecitabine, Irinotecan, Stivarga, Bevacizumab, Erbitux, Camptosar, Cetuximab, Eloxatin, Vectibix, Zaltrap, Betaseron, Fusilev, Lonsurf, Methotrexate, Panitumumab, Wellcovorin, Regorafenib, Mvasi, Keytruda, Opdivo, Cyramza, Interferon beta- lb, Levoleucovorin, Nivolumab, Ramucirumab, Tipiracil/Trifluridine, Ziv-aflibercept, Pembrolizumab, Ipilimumab, Khapzory, Yervoy and their derivatives.
  • the Senolytic Agents are used in adjuvant therapy for the treatment of colorectal cancer, in combination with or following primary therapy with at least one chemotherapeutic agent.
  • Said chemotherapeutic agent(s) could be used alone or in combination with the Senolytic Agents.
  • administration of Senolytic Therapy is effective in the treatment of a thyroid cancer or as an adjuvant drug.
  • the thyroid cancer disease parameters which are measured include, inter alia, a reduction in tumor size and/or metastasization.
  • the methods of use of Senolytic Agents for the treatment of thyroid cancer as adjuvants comprises Senolytic Therapy as disclosed in the subject matter of this patent.
  • Senolytic Therapy as disclosed in the subject matter of this patent.
  • Treatment Regimes of Senolytic Therapy for the treatment of thyroid cancer as adjuvants include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • the primary therapy used for treatment of thyroid cancer could involve one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s).
  • Said chemotherapeutic agents include but are not limited to, Armour Thyroid, Nexavar, Nature-Throid, Thyrogen, Caprelsa, Adriamycin, Cometriq, Thyroid desiccated, Sodium iodide-i-131, Sorafenib, Lenvima, Doxorubicin, Lenvatinib, Lodotope, Cabozantinib, Westhroid, Thyrotropin alpha, Vandetanib, Hicon, NP Thyroid, Dabrafenib, Tafmlar, WP Thyroid, i3odine Max, Mekinist, Trametinib and their derivatives.
  • the Senolytic Agents are used in adjuvant therapy for the treatment of thyroid cancer, in combination with or following primary therapy with at least one chemotherapeutic agent.
  • Said chemotherapeutic agent(s) could be used alone or in combination with the Senolytic Agents.
  • the method of use of Senolytic Agents as treatment or adjuvants for the inhibition of metastasis of a cancer comprises Treatment Regimes as disclosed in the subject matter of this patent.
  • Some examples of said Treatment Regimes include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • the Treatment Regimes for Therapeutically Effective Dose and/or the duration of cancer treatment stated regimes could be altered depending on the subject’s state of health and/or the subject’s response to the treatment including but not limited to alteration of Therapeutically Effective Dose and/or the duration of treatment, or combinations of Senolytic Agents and delivery methods or combinations thereof.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subject at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline
  • a subject’s progress could additionally be followed by detecting and/or monitoring metastatic cells and/or cancer cells present in the subject before, during or after the therapy using the methods as recognized by those skilled in the art.
  • Progeroid syndromes are a group of fatal, severe and rare genetic disorders which mimic premature aging while exhibiting various clinical features and phenotypes. PSs mimic many of the characteristics of human ageing such as hair loss, short stature, skin tightness, cardiovascular diseases and osteoporosis. Therefore, the Senolytic Small Molecules may also be useful for treating or alleviation of the effects of Progeroid Syndromes that occur as a result of premature aging process induced by congenital genetic mutations in individuals. Although all progeroid syndromes are characterized by similar clinical features, their underlying mechanisms can vary depending on the mutated gene and the pathway that is consequently altered. As a result of genomic instabilities due to the mutated genes, premature senescence emerges as a key factor underlying these conditions.
  • these syndromes include clinically and genetically heterogeneous diseases such as ataxia-telangiectasia, Bloom syndrome, Cockayne syndrome, Fanconi anaemia, Hutchinson-Gilford Progeria syndrome, Rothmund-Thomson syndrome, trichothiodystrophy, xeroderma pigmentosum, and Werner syndrome (aka adult progeria).
  • diseases such as ataxia-telangiectasia, Bloom syndrome, Cockayne syndrome, Fanconi anaemia, Hutchinson-Gilford Progeria syndrome, Rothmund-Thomson syndrome, trichothiodystrophy, xeroderma pigmentosum, and Werner syndrome (aka adult progeria).
  • Progeroid Syndromes include Hutchinson-Gilford Progeria Syndrome (HGPS) which is a rare , fatal and genetic condition of childhood, characterized by growth reduction, failure to thrive, a typical facial appearance (prominent forehead, protuberant eyes, thin nose with a beaked tip, thin lips, micrognathia and protruding ears) and distinct dermatologic features (generalized alopecia, aged-looking skin, sclerotic and dimpled skin over the abdomen and extremities, prominent cutaneous vasculature, dyspigmentation, nail hypoplasia and loss of subcutaneous fat). Individuals with HGPS exhibit atherosclerosis, lipodystrophy, heart infarction and death during puberty.
  • HGPS Hutchinson-Gilford Progeria Syndrome
  • HGPS disease parameters are measured by, inter alia, features of accelerated aging, hair loss (alopecia), aged-looking skin, joint abnormalities, and a loss of fat under the skin.
  • Progeroid Syndromes include Trichothiodystrophy characterized by brittle hair causing hair loss, neurological defects, bone abnormalities and fitness decline.
  • PSs include Werner Syndrome characterized by the dramatic, rapid appearance of features associated with normal aging in affected individuals. Affected individuals usually develop accompanying disorders of aging early in life, such as cataracts, skin ulcers, type 2 diabetes, diminished fertility, atherosclerosis, osteoporosis, and some types of cancer.
  • premature aging-associated decline and symptoms may be treated or prevented (i.e., the likelihood of occurrence of is reduced) by administering the Senolytic Therapy
  • the methods of use of Senolytic Agents for treatment of progeroid syndromes comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for the treatment of progeroid syndromes include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • the effectiveness of a method of treatment described herein may be manifested by reducing the number of symptoms of a premature aging disease or progeroid trait associated with a senescence-inducing stimulus, decreasing the severity of one or more symptoms, or delaying the progression of a premature aging disease or progeroid trait associated with a senescence-inducing stimulus.
  • preventing a premature aging disease or progeroid trait associated with a senescence-inducing stimulus refers to preventing (i.e., reducing the likelihood of occurrence) or delaying onset of a premature aging disease or progeroid trait associated with a senescence-inducing stimulus, or reoccurrence of one or more premature aging disease or progeroid trait associated with a senescence-inducing stimulus.
  • Tumor proliferation may be determined by tumor size, which can be measured in various ways familiar to a person skilled in the art, such as by PET scanning, MRI, CAT scan, biopsy, for example.
  • the effect of the therapeutic agent on tumor proliferation may also be evaluated by examining differentiation of the tumor cells. It has been demonstrated that a Senolytic Agent lowers the threshold for senescent cells to enter apoptosis after DNA damage. In some embodiments the Senolytic Agent is used against chemotoxicity or radiation damage by removing the senescent cells in controlled and safe manner.
  • the Senolytic Small Molecule(s) described herein may be administered after the chemotherapy or radiotherapy to kill (or facilitate killing) of these senescent cells.
  • the following treatment courses for administration of the Senolytic Small Molecule may be used in methods described herein for treating or preventing (i.e., reducing the likelihood of occurrence, or reducing the severity) a chemotherapy or radiotherapy side effect. Removal or destruction of senescent cells may ameliorate acute toxicity, including acute toxicity comprising energy imbalance, of a chemotherapy or radiotherapy.
  • Acute toxic side effects include but are not limited to gastrointestinal toxicity (e.g., nausea, vomiting, constipation, anorexia, diarrhea), peripheral neuropathy, fatigue, malaise, low physical activity, hematological toxicity (e.g., anemia), hepatotoxicity, alopecia (hair loss), pain, infection, mucositis, fluid retention, dermatological toxicity (e.g., rashes, dermatitis, hyperpigmentation, urticaria, photosensitivity, nail changes), mouth (e.g., oral mucositis), gum or throat problems, or any toxic side effect caused by a chemotherapy or radiotherapy.
  • gastrointestinal toxicity e.g., nausea, vomiting, constipation, anorexia, diarrhea
  • peripheral neuropathy e.g., fatigue, malaise, low physical activity
  • hematological toxicity e.g., anemia
  • hepatotoxicity e.g., hepatotoxicity
  • alopecia hair loss
  • pain infection
  • methods for ameliorating (reducing, inhibiting, or preventing occurrence (i.e., reducing the likelihood of occurrence)) acute toxicity or reducing severity of a toxic side effect (i.e., deleterious side effect) of a chemotherapy or radiotherapy or both in a subj ect who receives the therapy, wherein the method comprises administering to the subject an agent that selectively kills, removes, or destroys or facilitates selective destruction of senescent cells.
  • Administration of the Senolytic Small Molecule for treating or reducing the likelihood of occurrence, or reducing the severity of a chemotherapy or radiotherapy side effect may be accomplished by the same treatment courses described above for treatment/prevention of metastasis.
  • the Senolytic Small Molecule is administered in a Therapeutically Effective Dose during the off-chemotherapy or off- radiotherapy time interval or after the chemotherapy or radiotherapy treatment regimen has been completed.
  • the methods of use of Senolytic Agents aim for reduction of the side effects of chemotherapy and radiotherapy comprise Senolytic Therapy as disclosed in the subject matter of this patent.
  • Some examples of Treatment Regimes of Senolytic Therapy for reduction of the side effects of chemotherapy and radiotherapy include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
  • certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment.
  • Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
  • a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
  • the number of cycles of a chemotherapy or radiotherapy or the total length of time of a chemotherapy or radiotherapy dose regime can vary depending on the subject's response to the cancer therapy.
  • the senolytic therapy plan timeframe will be adjusted and aligned with said chemotherapy or radiotherapy treatment by a person skilled in the oncology art.
  • compositions comprising a Senolytic Small Molecule can be formulated in a manner appropriate for the delivery method by using techniques routinely practiced in the art.
  • the composition may be in the form of a solid (e.g., tablet, capsule), semi solid (e.g., gel), liquid, or gas (aerosol).
  • the Senolytic Small Molecule (or a composition comprising same) is administered in a Therapeutically Effective Dose as a bolus infusion.
  • the Senolytic Small Molecule when the Senolytic Small Molecule is delivered in a Therapeutically Effective Dose by infusion wherein the Senolytic Small Molecule is delivered to an organ or tissue comprising senescent cells or cancer tumors as an adjuvant drug to a chemotherapy to be killed in accordance with techniques routinely performed by a person skilled in the medical art.
  • compositions are well-known in the pharmaceutical arts.
  • examples of pharmaceutically-acceptable excipients include sterile saline and phosphate buffered saline at physiological pH. Preservatives, stabilizers, dyes, buffers, and the like may be provided in the composition.
  • the type of excipient is selected based on the mode of administration, as well as the chemical composition of the active ingredient(s).
  • the Senolytic Small Molecule is formulated as a lyophilizate.
  • a composition based on the disclosed subject matter may be lyophilized or otherwise formulated as a lyophilized product using one or more appropriate excipient solutions for solubilizing and/or diluting the agent(s) of the composition upon administration.
  • the Senolytic Small Molecule is encapsulated within liposomes using technology known and practiced in the art.
  • compositions comprising the Senolytic Small Molecule is formulated and packaged for delivery for any appropriate manner of administration described in the disclosed subject matter and in the art.
  • a composition comprising the Senolytic Small Molecule(s) may be delivered to a subject in need thereof by any one of several routes known to a person skilled in the art.
  • the composition may be delivered orally, intravenously, intraperitoneally, by infusion (e.g., a bolus infusion), subcutaneously, enteral, rectal, intranasal, by inhalation, buccal, sublingual, intramuscular, transdermal, intradermal, topically, intraocularly, vaginally, rectally, intrathecally, intracranially, or by some combination thereof.
  • a delivery method includes drug-coated or permeated stents for which the drug is the Senolytic Small Molecule. Formulations suitable for such delivery methods are described in greater detail herein. [000281]
  • the Senolytic Small Molecule (which may be combined with at least one therapeutically-acceptable excipient to form a composition comprising the Senolytic Small Molecule(s)) is administered in a Therapeutically Effective Dose directly to the target tissue or tumor or organ comprising senescent cells that contribute to manifestation of the disease or disorder.
  • At least one Senolytic Small Molecule is administered in a Therapeutically Effective Dose directly to an osteoarthritic joint (i.e., intra-articularly) of a subject in need thereof.
  • the Senolytic Small Molecule(s) may be administered in a Therapeutically Effective Dose to the joint via topical, transdermal, intradermal, or subcutaneous route.
  • methods are provided herein for treating a cardiovascular disease or disorder associated with arteriosclerosis, such as atherosclerosis by administering directly into an artery.
  • the Senolytic Small Molecule (which may be combined with at least one pharmaceutically- acceptable excipient to form a composition comprising the Senolytic Small Molecule(s)) for treating a senescence-associated pulmonary disease or disorder may be administered in a Therapeutically Effective Dose by inhalation, intranasally, by intubation, or intracheally, for example, to provide the Senolytic Small Molecule more directly to the affected pulmonary tissue.
  • the Senolytic Small Molecule (or composition comprising the Senolytic Small Molecule) may be delivered directly to the eye either by injection (e.g., intraocular or intravitreal) or by conjunctival application underneath an eyelid of a cream, ointment, gel, or eye drops.
  • the Senolytic Small Molecule or a composition comprising the Senolytic Small Molecule may be formulated as a timed release (also called sustained release, controlled release) composition or may be administered in a Therapeutically Effective Dose as a bolus infusion.
  • a composition comprising the Senolytic Small Molecule(s) may be in the form of a liquid.
  • a liquid composition comprising the Senolytic Small Molecule(s) may include, for example, one or more of the following: a sterile diluent such as water, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils that may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents; antioxidants; chelating agents; buffers and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • a parenteral composition can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • an injectable composition comprising the Senolytic Small Molecule(s) is preferably sterile.
  • a liquid composition comprising the Senolytic Small Molecule(s) may be applied to the eye in the form of eye drops.
  • a liquid composition comprising the Senolytic Small Molecule(s) may be delivered orally.
  • At least one of the Senolytic Small Molecules described herein can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, and if desired, with diluents, buffering agents, moistening agents, preservatives, coloring agents, and flavoring agents.
  • the compounds may be formulated with a buffering agent to provide for protection of the compound from low pH of the gastric environment and/or an enteric coating.
  • the Senolytic Small Molecule included in a composition comprising the Senolytic Small Molecule(s) may be formulated for oral delivery with a flavoring agent, e.g., in a liquid, solid or semi-solid formulation and/or with an enteric coating.
  • a composition comprising any one of the Senolytic Small Molecules described herein may be formulated for sustained or slow release (also called timed release or controlled release).
  • Such compositions comprising the Senolytic Small Molecule may generally be prepared using well known technology and administered in a Therapeutically Effective Dose by, for example, oral, rectal, intradermal, or subcutaneous implantation, or by implantation at the desired target site.
  • Sustained-release formulations may contain the compound dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Excipients for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of active component release. The amount of active agent contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition, disease or disorder to be treated or prevented.
  • compositions comprising the Senolytic Small Molecule are formulated for transdermal, intradermal, or topical administration.
  • Said compositions can be administered in a Therapeutically Effective Dose using a syringe, bandage, transdermal patch, insert, or syringe-like applicator, as a powder/talc or other solid, liquid, spray, aerosol, ointment, foam, cream, gel, paste.
  • This preferably is in the form of a controlled release formulation or sustained release formulation administered in a Therapeutically Effective Dose topically or injected directly into the skin adjacent to or within the area to be treated (intradermally or subcutaneously).
  • the active compositions comprising the Senolytic Small Molecule can also be delivered via iontophoresis.
  • Preservatives can be used to prevent the growth of fungi and other microorganisms. Suitable preservatives include, but are not limited to, benzoic acid, butylparaben, ethyl paraben, methyl paraben, propylparaben, sodium benzoate, sodium propionate, benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetypyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, thimerosal, and combinations thereof.
  • compositions comprising the Senolytic Small Molecule can be formulated as emulsions for topical application.
  • An emulsion contains one liquid distributed within the body of a second liquid.
  • the emulsion may be an oil-in-water emulsion or a water- in-oil emulsion.
  • Either or both of the oil phase and the aqueous phase may contain one or more surfactants, emulsifiers, emulsion stabilizers, buffers, and other excipients.
  • the oil phase may contain other oily pharmaceutically-approved excipients.
  • Suitable surfactants include, but are not limited to, anionic surfactants, non-ionic surfactants, cationic surfactants, and amphoteric surfactants.
  • Compositions comprising the Senolytic Small Molecule for topical application may also include at least one suitable suspending agent, antioxidant, chelating agent, emollient, or humectant.
  • ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • Liquid sprays may be delivered from pressurized packs, for example, via a specially shaped closure.
  • Oil-in-water emulsions can also be used in the compositions comprising the Senolytic Small Molecule, patches, bandages and articles. These systems are semi solid emulsions, micro-emulsions, or foam emulsion systems.
  • the Senolytic Small Molecule(s) can be formulated with oleaginous bases or ointments to form a semisolid composition with a desired shape.
  • these semisolid compositions comprising the Senolytic Small Molecule can contain dissolved and/or suspended bactericidal agents, preservatives and/or a buffer system.
  • a petrolatum component that may be included may be any paraffin ranging in viscosity from mineral oil that incorporates isobutylene, colloidal silica, or stearate salts to paraffin waxes.
  • Absorption bases can be used with an oleaginous system.
  • Additives may include cholesterol, lanolin (lanolin derivatives, beeswax, fatty alcohols, wool wax alcohols, low HLB (hydrophobellipophobe balance) emulsifiers, and assorted ionic and nonionic surfactants, singularly or in combination.
  • lanolin lanolin derivatives, beeswax, fatty alcohols, wool wax alcohols, low HLB (hydrophobellipophobe balance) emulsifiers, and assorted ionic and nonionic surfactants, singularly or in combination.
  • a composition comprising any one of the Senolytic Small Molecules described herein may be formulated for sustained or slow release (which may also be called timed release or controlled release).
  • Controlled or sustained release transdermal or topical formulations can be achieved by the addition of time-release additives, such as polymeric structures, matrices, that are available in the art.
  • the compositions comprising the Senolytic Small Molecule may be administered in a Therapeutically Effective Dose through use of hot-melt extrusion articles, such as bioadhesive hot-melt extruded film.
  • the formulation can comprise a cross-linked polycarboxylic acid polymer formulation.
  • a cross- linking agent may be present in an amount that provides adequate adhesion to allow the system to remain attached to target epithelial or endothelial cell surfaces for a sufficient time to allow the desired release of the compound.
  • an insert, transdermal patch, bandage or article comprise a mixture or coating of polymers that provide release of the active agents at a constant rate over a prolonged period of time.
  • the article, transdermal patch or insert comprises water-soluble pore forming agents, such as polyethylene glycol (PEG) that can be mixed with water insoluble polymers to increase the durability of the insert and to prolong the release of the active ingredients.
  • PEG polyethylene glycol
  • Transdermal devices may also comprise a water insoluble polymer.
  • rate controlling polymers are useful for administration to sites where pH change can be used to effect release. Said rate controlling polymers are applied using a continuous coating film during the process of spraying and drying with the active compound.
  • the coating formulation is used to coat pellets comprising the active ingredients that are compressed to form a solid, biodegradable insert.
  • a polymer formulation is utilized to provide controlled or sustained release including bioadhesive polymers.
  • a sustained-release gel and the compound may be incorporated in a polymeric matrix, such as a hydrophobic polymer matrix.
  • a polymeric matrix include a microparticle. The microparticles can be microspheres, and the core may be of a different material than the polymeric shell.
  • the polymer may be cast as a thin slab or film, a powder produced by grinding or other standard techniques, or a gel such as a hydrogel.
  • said polymer is in the form of a coating or part of a bandage, stent, catheter, vascular graft, or other device to facilitate delivery of the Senolytic Small Molecule.
  • the matrices can be formed by solvent evaporation, spray drying, solvent extraction and other methods known to those skilled in the art.
  • one or more Senolytic Small Molecule(s) may be delivered directly into a blood vessel (e.g., an artery) via a stent.
  • a stent is used for delivering the Senolytic Small Molecule to an atherosclerotic blood vessel (an artery).
  • the Senolytic Small Molecule may also be incorporated into the stent (for example as a coating or pores in the metal stent itself).
  • the Senolytic Small Molecule may be formulated within liposomes and applied to a stent. Placement of stents in an atherosclerotic artery is performed by a person skilled in the medical art.
  • the Senolytic Small Molecule is administered in a Therapeutically Effective Dose to a subject who has an ophthalmic senescence-associated or disease or disorder may be delivered intraocularly or intravitreally.
  • the Senolytic Small Molecule(s) may be administered in a Therapeutically Effective Dose to the eye by a conjunctival route, applying the Senolytic Small Molecule to the mucous membrane and tissues of the eyelid, either upper, lower, or both. Any of these administrations may be bolus infusions.
  • the Senolytic Small Molecule(s) is administered in a Therapeutically Effective Dose in the forms that will promote intracellular delivery. In some embodiments, the Senolytic Small Molecule(s) is administered in a Therapeutically Effective Dose in the forms that will be recognized by the person in the skilled art.
  • the Senolytic Small Molecule(s) could be co-administered with enzyme inhibitors to alleviate proteolytic degradation thereby increasing bioavailability. In some embodiments of the Senolytic Small Molecule(s) is co-administered with enzyme inhibitor. [000297] As will be recognized by those skilled in the art in some embodiments, the Senolytic Small Molecule(s) could be co-administered with absorption enhancers such as chitin and its derivatives such as chitosan to enhance absorption of hydrophilic drug molecules. In some embodiments of the Senolytic Small Molecule(s) is co-administered with absorption enhancers such as chitin and its derivatives (i.e. chitosan).
  • the first embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule exhibits maximized interference with the CR3 domain of Fox04.
  • the second embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule exhibits reduced direct interaction with the p53DBD compared to that of the endogenous Fox04.
  • the third embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule preferably exhibits a reduced interference with the CR3 domains of FoxOl or Fox03 compared to that of said Fox04.
  • the forth embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule exhibits reduced interference with DNA compared to that of said Fox04.
  • the fifth embodiment is the method according to any one of the first to forth embodiments, wherein the senescent cell is characterized as expressing the senescence- associated secretory phenotype (SASP).
  • SASP senescence- associated secretory phenotype
  • the sixth embodiment is the method according to any one of the first to forth embodiments, wherein the method comprises treatment of a senescence-associated disease or disorder.
  • the seventh embodiment is the method according to any one of the first to forth embodiments, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the small molecule is administered before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent.
  • the eight embodiment is the method according to any one of the first to forth embodiments, wherein the said cancer is characterized as resistant to therapy.
  • the ninth embodiment is the method according to any one of the first to forth embodiments, wherein said therapy-resistant cancer comprises is metastatic melanoma, breast cancer, or glioblastoma, and wherein the therapy to which the cancer is resistant is radiation therapy or chemotherapy.
  • the tenth embodiment is the method according to any one of the first to forth embodiments, wherein the subject comprises a human characterized as suffering from, or expected to suffer from chronic inflammatory diseases or a senescence related disease or disorder.
  • the eleventh embodiment is the method according to any one of the first to forth embodiments, wherein the method is effective to remove cells from the subject that express pl6INK4a, wherein the subject is characterized as suffering from, or expected to suffer from a senescence-associated disease or disorder.
  • the twelfth embodiment is the method according to any one of the first to forth embodiments, wherein the method is effective to alter levels of the Serine-46 phosphorylated p53 foci in the subject, wherein the subject is characterized as suffering from, or expected to suffer from, a senescence-associated disease or disorder.
  • the thirteenth embodiment is the method according to any one of the first to forth embodiments, wherein the method comprises administering the small molecules according to the Therapeutically Regime or an Impulse Regime, Sustained Regime, a Gentle Shock Regime, or combinations thereof.
  • the fourteenth embodiment is the method of inducing the apoptosis of a senescent cell in a subject, wherein the method uses one or more of the Senolytic Small Molecules listed in the Table 1.
  • the fifteenth embodiment is the method of inducing the apoptosis of a senescent cell in a subject, wherein the method uses one or more compounds that carry a similar scaffold to the Senolytic Small Molecules listed in the Table 1.
  • the sixteenth embodiment is the method of use of the Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the seventeenth embodiment is the method of use of the compounds that carry a similar chemical scaffold to any of the Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the eighteenth embodiment is the method of use of the HIV reverse transcriptase inhibitors listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the nineteenth embodiment is the method of use of the compounds that carry a similar chemical scaffold to the HIV reverse transcriptase inhibitors listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the twentieth embodiment is the method of use of the reductase inhibitor listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the twenty first embodiment is the method of use of the compounds that carry a similar chemical scaffold to the reductase inhibitor listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the twenty second embodiment is the method of use of the anti-cancer Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the twenty third embodiment is the method of use of the compounds that carry a similar chemical scaffold to the anti-cancer Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
  • the twenty forth embodiment is the method according to any one of claims 1-10, wherein causing the Senolytic Small Molecule to interfere with the CR3 domain of Fox04 of the senescent cell comprises administering a pharmaceutical composition comprising the Senolytic Small Molecule according to any one of the embodiments.
  • the twenty fifth embodiment is the method according to any one of the embodiments, wherein a small molecule exhibits maximized interference with the CR3 domain of F ox04.
  • the twenty sixth embodiment is the method according to any one of the embodiments, wherein a small molecule exhibits reduced direct interaction with the p53DBD compared to that of the endogenous Foxo4.
  • the twenty seventh embodiment is the method according to any one of the embodiments, wherein a small molecule preferably exhibits a reduced interference with the CR3 domains of FoxOl or Fox03 compared to that of said Fox04.
  • the twenty eighth embodiment is the method according to any one of the embodiments, wherein a small molecule exhibits reduced interference with DNA compared to that of said Fox04.
  • the twenty ninth embodiment is the method according to any one of the embodiments, wherein the senescent cell is characterized as expressing the senescence- associated secretory phenotype (SASP).
  • SASP senescence- associated secretory phenotype
  • the thirtieth embodiment is the method according to any one of the embodiments, wherein the method comprises administering Therapeutic Regimes for treatment of a senescence-associated disease or disorder.
  • Another embodiment is the method of use of the Senolytic Small Molecules, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the Senolytic Agent is administered before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent.
  • the thirty first embodiment is the method according to any one of the embodiments, wherein the said cancer is characterized as resistant to therapy.
  • the thirty second embodiment is the method according to any one of the embodiments, wherein said therapy-resistant cancer comprises is metastatic melanoma, breast cancer, or glioblastoma, and wherein the therapy to which the cancer is resistant to radiation therapy or chemotherapy.
  • the thirty third embodiment is the method according to any one of the embodiments, wherein the subject comprises a human characterized as suffering from, or expected to suffer from chronic inflammatory diseases or a senescence related disease or disorder.
  • the thirty forth embodiment is the method according to any one of the embodiments, wherein the method is effective to remove cells from the subject that express pl6INK4a, wherein the subject is characterized as suffering from, or expected to suffer from a senescence- associated disease or disorder.
  • the thirty fifth embodiment is the method according to any one of the embodiments, wherein the method is effective to alter levels of the Serine-46 phosphorylated p53 foci in the subject, wherein the subject is characterized as suffering from, or expected to suffer from, a senescence-associated disease or disorder.
  • the thirty sixth embodiment is the method according to any one of the embodiments, wherein the method comprises Senolytic Therapy using one or more Senolytic Small Molecules.
  • Another embodiment is the method use of the Senolytic Small Molecules, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the Senolytic Agent is administered as Therapeutic Regimes before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent or during an immunotherapy.
  • the Senolytic Small Molecule(s) and the disclosed method described in the present application can be modified and varied over a tremendous range of applications to produce a wide range of Senolytic Small Molecules which may be directed to specific therapies which target those subsets of senescent cells and accordingly the scope of patented subject matter is not limited by any of the specific exemplary teachings given. It is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.

Abstract

Provided herein are methods of use of 12 small molecules extracted from ZINC15 library for treating senescence-associated diseases and disorders by specifically targeting the FoxO4 protein. The diseases and disorders treatable with said repurposed Senolytic Agents include diseases with inflammatory origin including but not restricted to diabetes, cardiovascular diseases, or pulmonary diseases, including COPD; asthma, emphysema, or breathlessness; renal or hepatic insufficiency, cirrhosis, or osteoarthritis; senescence-associated ophthalmic diseases and disorders; and senescence-associated dermatological diseases and disorders; diabetic ulcers; kyphosis; scoliosis; weight loss; hair loss; muscle loss; loss of bone density; frailty and/or reduced fitness; hearing loss such as deafness; or combinations thereof.

Description

REPURPOSING SMALL MOLECULES FOR SENESCENCE-RELATED DISEASES
AND DISORDERS
BACKGROUND
[0001] Senescence and apoptosis are among mechanisms that, when activated, restrict tumor growth. Through apoptosis, damaged cells are cleared from the organism while senescent cells remain alive in the organism though permanently restricted from entering the cell cycle. Senescence may be associated with an increase in metabolic activity. In the majority of cases, senescent cells develop a defined, but heterogeneous, secretory profile termed as senescence- associated secretory phenotype (SASP). The SASP entails release of pro-inflammatory cytokines and chemokines, tissue-damaging proteases, factors that can affect stem and progenitor cell function, haemostatic factors, and growth factors, among others. Senescent cells that express the SASP can have substantial local and systemic pathogenic effects.
[0002] The SASP secretion comprises a range of different proteins, including several proteins known to play a role in aging and age-related diseases, including matrix metalloproteases such as MMP3, growth factors, chemokines such as CCL2 and CLL11, and prominent interleukins (ILs) such as ILl, IL6, and IL8. Above a certain threshold, such factors can significantly impair tissue function. The chronic SASP secretion by senescent cells may impair the functioning of neighboring cells. As such, senescent cells are thought to be major contributors of inflammation. Theoretically, low, but chronic, levels of inflammation are drivers of age-related decline in function. Consistent with this theory, senescence and SASP are elevated in a number of fast-aging mouse models and, where tested, senescence clearance delays their decline in health.
[0003] Additionally, mutations occurring in the senescent cells may lead to changes that allow those senescent cells to escape from cell cycle arrest and eventually be convert to tumorigenic cells. Thus senescent cells that display resistance to apoptosis and accumulate with age are targets of anti-aging research. The main aim of such research has been to discover the molecular pathways that direct cells to senescence but not apoptosis and, eventfully, to develop agents that interfere with these pathways so that administration of such agents will induce apoptosis in a senescent cell in a safe and predictable manner. [0004] The forkhead box (FOX) protein family comprises the FOX class O subfamily (FoxO) that has 4 mammalian members: Forkhead box protein 01 (FoxOl) (FKHR, FoxOl A), Forkhead box protein 03 (Fox03) (FKHRL1, Fox03A), Forkhead box protein 04 (Fox04) (AFX, AFXl, MLLT7), and Forkhead box protein 06 (Fox06). FoxOs are transcription factors that play important roles in suppression of tumors. Despite the fact that the exact roles of FoxOs in senescence remain to be unraveled, it has been well-established for the Fox04 that its mRNA and protein levels have been specifically increased in response to genotoxic activation of senescence. In line with this observation, Fox04 function was essential for senescence activation by genotoxic damage, while the loss of Fox04 function was associated with the apoptosis of senescent cells. Hence, Fox04 has a pivotal role in the cells’ direction to either senescence or apoptosis. Interfering with this key molecule Fox04 represents efficient way of blocking senescence and steering the cells’ fate towards apoptosis.
[0005] Senescence has been characterized to be a complex phenomenon that can be activated by distinct signals. In particular, genotoxic activation of senescence has been characterized by the formation of DNA-SCARS (DNA Segments with Chromatin Alterations Reinforcing Senescence). When genotoxic damage is present, Fox04 is recruited to DNA- SCARS that also includes tumor protein 53 (p53) as a major component. Oncogenic BRAF mutation at V600E are encountered in ~7% of all human tumors with particularly enhanced occurrence in melanoma (~70%). Additionally, melanoma cells were found to generally have an elevated number of DNA-SCARS containing FOX04. It has been shown that the BRAF mutation promotes the Fox04 leading to senescence, while at the same time it also causes phosphorylation of Serine 46 (S46) of p53, the condition which was known to strongly favor apoptosis over senescence. It has been shown for the DNA-damaged melanoma cells that interference with their Fox04 expression results in a marked increase of apoptosis. Overall these findings pointed out the Fox04 presence inhibited the apoptosis by the S46 phosphorylated p53. Furthermore, it has been shown that inhibition of the kinase that phosphorylates the S46 of p53 led to impaired apoptosis of the senescent cells even in the absence of Fox04. These particular observations proposed a pivotal role for Fox04 in restraining the apoptosis mediated by the S46 phosphorylated p53. Therefore, eradicating the senescence and inducing the apoptosis via blocking the action of Fox04 on p53 is of interest, as discussed herein.
[0006] It is well established that chronic inflammation is the cause of many diseases. As stated above, SASP may contribute to chronic inflammation in old age. However, there is no generic Senolytic Agent, but a number of small molecule with separate Senescent Cell Anti- apoptotic Pathways (SCAPs) and cell types some of which are Dasatinib (which acts on Dependence receptor/Src kinase/tyrosine kinase and target Primary human and mouse preadipocytes (adipose-derived stem cells), Quercetin (which acts on Bcl-2 family, p53/p21/serpine, & PI3K/AKT and target HUVECs, mouse bone marrow-derived mesenchymal stem cells), Navitoclax (which ascts on ABT263 and target MR-90 Cells, HUVECs), Piperlongumin (which acts on A1331852/ A1155463 and target IMR-90 Cells, HUVECs), and Fisetin (which acts on PI3K/AKT and targets HUVECs). The molecular pathway of the Senolytic Small Molecules disclosed herein are different than those mentioned in this paragraph therefore the methods of use of these Senolytic Small Molecule(s) for said diseases and disorders are different.
SUMMARY OF THE INVENTION
[0007] Selectively killing a senescent cell in a mammal is a rejuvenation therapy particularly for post-production adult human cohort. It enables treatment of senescence-related diseases and disorders. Review of a structural model of the Fox04-p53 complex helps in an understanding of the inhibitory action of the CR3 domain of Fox04 on the p53. Thus, by blocking the CR3 domain of Fox04, the inhibitory action of Fox04 on p53 would be eliminated and p53 will be released from the Fox04 complex to function in the apoptotic pathways.
[0008] One aspect of the present invention relates to a novel method of use for the 12 small molecules which are listed in the Table 1 and shown in Figures 1-12 as Senolytic Agents for treatment of senescence-associated diseases. These listed small molecules are normally used for other purposes other than to treat senescence-associated diseases and disorders. After extensive in-silico analysis, these small molecules were discovered to target the CR3 domain of Fox04, thus said small molecules are selected to be suitable to target senescent cells by blocking Fox04-p53 complex, and induce them to apoptosis.
[0009] Another aspect of the present invention relates to novel chemical structures which are derived from chemical scaffolds shown in the Figure 2. Not intending to be bound by theory, an extensive in silico analysis suggested that these selected 12 small molecules are able to interfere with SCAP by targeting the CR3 domain of Fox04.The novel derivatives of said small molecules disclosed herein with a similar scaffold will still be able to integrfere with CR3 domain of Fox04 thereby interfering with SCAP. From here on the novel Senolytic Agents definition hereafter shall include the small molecules listed in Table 1 and their novel derivatives.
[00010] Preferably, the novel Senolytic Agents of the present invention do not have to be continuously present to exert their effects. Brief disruption of Fox04 expressed pro-survival pathways is adequate to kill senescent cells. The inventive treatment specifies that novel Senolytic Agents may be administered intermittently. The novel Senolytic Agents trigger apoptosis of the senescent cells which in turn stimulate overall rejuvenation in a safe manner by reactivating the stem cell.
[00011] A method is disclosed herein for selectively killing senescent cells and for treating senescence-associated diseases and disorders by administering the novel Senolytic Agents of the present invention to minimize the interaction between Fox04 and p53. The method of use of the novel Senolytic Agents may comprise three different dose regimes and their variations, wherein each dose regime is expressed by distinct therapeutically effective amount and distinct administration frequencies. Each said dose regime is developed to treat different types of senescence-associated diseases and disorders. Killing senescent cells reduces the inflammatory SASP, and therefore reduces the chronic inflammation in the metabolism. Further, said treatment stimulates overall rejuvenation in a safe manner. The diseases and disorders treatable with said novel Senolytic Agents include all diseases with inflammatory origin including diabetes, cardiovascular diseases, pulmonary diseases, osteoarthritis; senescence-associated ophthalmic diseases and disorders; and senescence-associated dermatological diseases and disorders. The applicable regimes and individualization of the treatment for said diseases are described under corresponding headings within this application.
[00012] Disclosed herein are repurposing small molecules, referred to as the“Senolytic Small Molecules,” including those listed in Table 1 which are effective to induce apoptosis of senescent cells in a subject, such as a mammal, by inhibiting the action of Fox04 on p53. In some embodiments, the Senolytic Small Molecules may be designed to effectively block the CR3 domain of Fox04 from interfering with the DNA binding function of p53 that is phosphorylated at Serine 46, particularly, from interfering with the bulky FH domain of Fox04. Additionally, or alternatively, in some embodiments the Senolytic Small Molecules may be rationally designed to minimize their interaction with the DBD of p53, other FoxOs, and the DNA duplex containing a FoxO consensus binding.
[00013] In some embodiments, a method for selectively inducing apoptosis of senescent cells and/or for treating a senescence-associated disease or disorder comprises administering one or more of the Senolytic Small Molecules which minimize the interaction between Fox04 and p53. In various embodiments, the method may comprise administering one or more of the Senolytic Small Molecule(s) via a treatment regime as disclosed herein or some variation thereof. In various embodiments, treatment regimes may vary as to administration frequency and/or dosing (for example, as to a dosage that will be therapeutically effective). Particular treatment regimes may be specifically developed to treat different types of senescence-associated diseases or disorders. Induction of apoptosis in senescent cells (that is, killing senescent cells) reduces the inflammatory senescence-associated secretory phenotype and therefore significantly reduce the chronic inflammation in the metabolism. Further, said treatment may stimulate overall rejuvenation in a safe manner. Thus, the diseases and disorders treatable via the Senolytic Small Molecules may include, but not limited to, all diseases with inflammatory origin including diabetes, cardiovascular diseases, pulmonary diseases, osteoarthritis; senescence-associated ophthalmic diseases and disorders; and senescence- associated dermatological diseases and disorders. The applicable regimes and individualization of the treatment for said diseases are presented under corresponding headings within this application.
[00014] In some embodiments, the Senolytic Small Molecules may not have to be continuously present to exert an effect. For examples, brief disruption of pro-survival pathways, such as by administration of a Senolytic Small Molecule, is adequate to kill senescent cells. Thus, in some embodiments, the Senolytic Small Molecules are suitable to be administered intermittently.
[00015] It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
BRIEF DESCRIPTION OF THE DRAWINGS
[00016] Figure 1 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 22863770. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00017] Figure 2 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 377845189. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00018] Figure 3 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 5390317. The snapshots were obtained from the last frame of the 200 ns MD simulations. [00019] Figure 4 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 95566015. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00020] Figure 5 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 8402312. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00021] Figure 6 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 70664738. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00022] Figure 7 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 25725416. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00023] Figure 8 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 150421585. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00024] Figure 9 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 13513540. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00025] Figure 10 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 22074898. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00026] Figure 11 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 150421592. The snapshots were obtained from the last frame of the 200 ns MD simulations.
[00027] Figure 12 shows the 3D complexes formed by the CR3 domain of Foxo4 and ZINC ID 95608759. The snapshots were obtained from the last frame of the 200 ns MD simulations
[00028] Figure 13 A illustrates the dose in which the Senolytic Peptides shall be administered following guidance provided in the Impulse Regime. The Therapeutically Effective Dose and frequency of administration may vary within this regime. The Impulse Regime presents an optional follow up treatment course in which the Therapeutically Effective Dose may be increased or decreased. Figure 13A depicts at least six different considerations: (1) Treatment Cycle (comprised of subject’s initial assessment, administration of Therapeutically Effective Dose and final assessment); (2) Therapeutically Effective Dose delivered in 1, 2, or 3 days; (3) Optional further treatment cycle(s) may be required if senescent cell reduction is unsatisfactory; (4) Senescence Clearance Interval 2-4 weeks; (5) Initial senescence assessment via biopsy samples; and (6) Final senescence assessment via biopsy samples.
[00029] Figure 13B illustrates the dose in which the Senolytic Peptides shall be administered following guidance provided in the Sustained Regime. The Therapeutically Effective Dose and frequency of administration may vary within this regime. The Sustained Regime presents an optional follow up treatment course in which the Therapeutically Effective Dose may be increased or decreased. Figure 13B depicts: (1) Treatment Cycle (comprised of subject’s initial assessment, administration of Therapeutically Effective Dose and final assessment); (2) Therapeutically Effective Dose delivered in 1, 2, or 3 weeks; (3) Optional further treatment cycle(s) may be required if senescent cell reduction is unsatisfactory. Therapeutically Effective Dose may be readjusted; (4) Senescence Clearance Interval 2-4 weeks; (5) Initial senescence assessment via biopsy samples; and (6) Final senescence assessment via biopsy samples.
[00030] Figure 13C illustrates the dose in which the Senolytic Peptides shall be administered following guidance provided in the Gentle Regimes. The Therapeutically Effective Dose and frequency of administration may vary within this regime. The Gentle Regime presents an optional follow up treatment course in which the Therapeutically Effective Dose may be increased or decreased. Figure 13C depicts: (1) Treatment Cycle (comprised of subject’s initial assessment, administration of Therapeutically Effective Dose and final assessment); (2) Therapeutically Effective Dose delivered in 3 or 3 weeks; (3) Optional further treatment cycle(s) may be required if senescent cell reduction is unsatisfactory. Therapeutically Effective Dose may be readjusted; (4) Senescence Clearance Interval 2-4 weeks; (5) Initial senescence assessment via biopsy samples; and (6) Final senescence assessment via biopsy samples.
[00031] Table 1 shows the binding affinities and the ligand affinities of the small molecules disclosed as Senolytic Agents in the application which are able to inhibit the CR3 domain of Foxo4, thereby inducing p53 dependent apoptosis.
Figure imgf000009_0001
[00032] DETAILED DESCRIPTION OF THE INVENTION
[00033] With respect to the following disclosure, the following definitions apply:
[00034] “Adjuvant” as used herein, refers to, but is not limited to, chemicals, small molecule, peptides such as the Senolytic Agent(s) including the Senolytic Small Molecule(s) or methods that are used prior to, in combination with or following the primary therapy which includes but not limited to surgery in order to enhance or modify the effect of the primary therapy, lower the risk of cancer recurrence and increase patient survival.
[00035] “Adjuvant therapy” as used herein, refers to, but not limited to, the therapy or therapies that are used prior to, in combination with; concomitant or concurrent therapy or following the primary therapy in order to enhance or modify the effect of the primary therapy which includes but not limited to surgery, lower the risk of cancer recurrence and increase patient survival. The adjuvant therapy includes, but is not limited to, chemotherapy, radiation therapy, hormone therapy, targeted therapy, biological therapy or other novel therapies that will be recognized by those skilled in the art.
[00036] “Administration” or“administering”“administer(ed)” as used herein, refers to a method of giving a dosage of a compound or composition to subject, such as a vertebrate or invertebrate, including a mammal, a bird, a fish, or an amphibian, via a suitable mode of administration, for example, intra-respiratory, topical, oral, intravenous, intraperitoneal, intramuscular, buccal, rectal, sublingual and/or intrathecal. In various embodiments as will be disclosed herein, the preferred mode of administration can vary depending on various factors, such as the components being administered, the tissue site being targeted (e.g., a tissue in which the disease or disorder resides, is present, or is manifested which may be a tumor), the particular disease or disorder involved, and the severity of the disease or disorder.
[00037] “Autoimmune disease(s) or disorder(s),” as used herein, refers to autoimmune diseases or disorders such, but not limited to, osteoporosis, psoriasis, oral mucositis, rheumatoid arthritis, inflammatory bowel disease, eczema, kyphosis, herniated intervertebral disc, and the pulmonary diseases, COPD, and idiopathic pulmonary fibrosis.
[00038] “Biological sample” or“biopsy sample,” as used herein, refers to a biological sample which is obtained from a subject by invasive, non-invasive or minimally invasive methods, for example, a blood sample, a serum sample, a plasma sample, a biopsy specimen, body fluids (for example, lung lavage, ascites, mucosal washings, synovial fluid, vitreous fluid, or spinal fluid), bone marrow, lymph nodes, tissue explant, skin tissue sample, vaginal tissue, organ culture, or any other tissue or cell preparation from a subject.
[00039] “Cancer,” as used herein, refers to, but is not limited to, cancers which are solid tumors or liquid tumors. Solid tumors may include, for example, prostate cancer, testicular cancer, breast cancer, brain cancer (including glioblastoma) , pancreatic cancer, colon cancer, colorectal cancer, thyroid cancer, stomach cancer, lung cancer, ovarian cancer, Kaposi's sarcoma, skin cancer (including squamous cell skin cancer), renal cancer, head and neck cancers, throat cancer, squamous carcinomas (e.g., that form on the moist mucosal linings of the nose, mouth, throat, etc. such as laryngeal and hypopharyngeal cancers), bladder cancer, osteosarcoma (bone cancer), cervical cancer, endometrial cancer, esophageal cancer, liver cancer, hepatocellular carcinoma, and kidney cancer and further including the metastasis of melanoma cells, prostate cancer cells, testicular cancer cells, breast cancer cells, brain cancer cells, pancreatic cancer cells, colon cancer cells, thyroid cancer cells, stomach cancer cells, lung cancer cells, ovarian cancer cells, Kaposi's sarcoma cells, skin cancer cells, renal cancer cells, head or neck cancer cells, throat cancer cells, squamous carcinoma cells, bladder cancer cells, osteosarcoma cells, cervical cancer cells, endometrial cancer cells, esophageal cancer cells, liver cancer cells, or kidney cancer cells. Liquid tumors may include, for example, cancers occurring in blood, bone marrow, and lymph nodes and include generally, leukemias (myeloid and lymphocytic) including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), and hairy cell leukemia lymphomas (e.g., Hodgkin lymphoma), and melanoma, including multiple myeloma).
[00040] “Cancer stem cells” (CSC) as used herein, refers to, but is not limited to, cancer cells that are tumorigenic or tumor initiating. CSCs possess properties of normal stem cells such as self-renewal to generate more CSCs and differentiation into different cell types to generate non- CSCs and increase tumor burden. CSCs may be a distinct population of cells within a cancer that result in cancer progression, relapse, and/or metastasis. CSCs may exhibit stem cell plasticity, where the cell-state transitions between a non-CSC and a CSC.
[00041] “Cardiovascular disease(s)” as used herein, refers to, but is not limited to, angina, arrhythmia, atherosclerosis, cardiomyopathy, congestive heart failure, coronary artery disease (CAD), carotid artery disease, endocarditis, heart attack (coronary thrombosis, myocardial infarction [MI]), high blood pressure/hypertension, aortic aneurysm, brain aneurysm, cardiac fibrosis, cardiac diastolic dysfunction, hypercholesterolemia/hyperlipidemia, mitral valve prolapse, peripheral vascular disease (e.g., peripheral artery disease (PAD)), cardiac stress resistance, and stroke.
[00042] “Chemotherapeutic agent(s)” as used herein, refers to a drug used to treat cancer. Examples of chemotherapeutic agents include but is not limited to Tamoxifen, Femara, Herceptin, Letrozole, Taxol, Soltamox, Epirubicin, Trastuzumab, Leuprolide, Paclitaxel, Ellence, Pharmorubicin PFS, Neratinib, Nerlynx, Ogivri, Opdivo, Intron A, Sylatron, Yervoy, Interferon alfa-2b, Nivolumab, Proleukin, Pembrolizumab, Dacarbazine, Keytruda, Aldesleukin, Lmlygic, Ipilimumab, DTIC-Dome, Temozolomide, Peginterferon alfa-2b, Talimogene laherparepvec, Mekinist, Tafmlar, Zelboraf, Trametinib, Dabrafenib, Vemurafenib, Cotellic, Isotretinoin, Peglntron, Braftovi, Mektovi, Cobimetinib, Binimetinib, Encorafenib, Xeloda, Oxaliplatin, Avastin, Fluorouracil, Leucovorin, Capecitabine, Irinotecan, Stivarga, Bevacizumab, Erbitux, Camptosar, Cetuximab, Eloxatin, Vectibix, Zaltrap, Betaseron, Fusilev, Lonsurf, Methotrexate, Panitumumab, Wellcovorin, Regorafenib, Mvasi, Cyramza, Interferon beta- lb, Levoleucovorin, Ramucirumab, Tipiracil/Trifluridine, Ziv- aflibercept, Khapzory, Temodar, Temozolomide, Matulane, BiCNU, Gliadel, Carmustine, Hydroxyurea, Procarbazine, Gleevec, Mercaptopurine, Sprycel, Adriamycin, Dasatinib, Trexall, Iclusig, Blincyto, Pegaspargase, Doxorubicin, Imatinib, Oncaspar, Purinethol, Blinatumomab, Erwinaze, Ponatinib, Vincristine liposome, Asparaginase erwinia chrysanthemi, Marqibo, Vumon, Xatmep, Arranon, Besponsa, Clolar, Kymriah, Clofarabine, Nelarabine, Purixan, Teniposide, Inotuzumab ozogamicin, Tisagenlecleucel, Prednisone, Dexamethasone, Sprycel, Cytarabine, Dexamethasone Intensol, Triamcinolone, Idarubicin, Clinacort, Idamycin, Fludarabine, Kenalog-40, Dexpak Taperpak, De-Sone LA, Idamycin PFS, Bicalutamide, Casodex, Zytiga, Lupron Depot, Xtandi, Zoladex, Eligard, Firmagon, Abiraterone, Provenge, Taxotere, Enzalutamide, Goserelin, Trelstar, Degarelix, Estrace, Estradiol, Delestrogen, Docetaxel, Estradiol Patch, Flutamide, Triptorelin, Jevtana, Nilandron, Zofigo, Zoladex 3 -Month, Cabazitaxel, Premarin, Conjugated estrogens, Cyclophosphamide, Menest, Nilutamide, Novantrone, Sipuleucel-T, Trelstar Depot, Trelstar LA, Vantas, Apalutamide, Emcyt, Erleada, Eulexin, Histrelin, Mitoxantrone, Radium 223 dichloride, Viadur, Yonsa, Esterified estrogens, Estramustine, Premarin intravenous, Supprelin LA, Tarceva, Alimta, Iressa, Erlotinib, Cisplatin, Gemzar, Abraxane, Tagrisso, Xalkori, Navelbine, Pemetrexed, Gefitinib, Crizotinib, Gilotrif, Venorelbine, Platinol, Tecentriq, Afatinib, Gemcitabine, Ceritinib, Platinol-AQ, Photofrin, Alecensa, Alectinib, Paclitaxel protein-bound, Porfimer, Zykadia, Atezolizumab, Durvalumab, Imfinzi, Necitumumab, Osimertinib, Alunbrig, Brigatinib, Dacomitinib, Infiigem, Lorbrena, Lorlatinib, Portrazza, Vizimpro, Etoposide, Topotecan, Hycamtin, VePesid, Toposar, Etopophos, Armour Thyroid, Nexavar, Nature- Throid, Thyrogen, Caprelsa, Cometriq, Thyroid desiccated, Sodium iodide-i-131, Sorafenib, Lenvima, Lenvatinib, Lodotope, Cabozantinib, Westhroid, Thyrotropin alpha, Vandetanib, Hicon, NP Thyroid, WP Thyroid, i3odine Max, Carboplatin, Doxil, Doxorubicin liposomal, Alkeran, Paraplatin, Lynparza, Olaparib, Zejula, Cosmegen, Melphalan, Rubraca, Dactinomycin, Niraparib, Tepadina, Rucaparib, Thiotepa, Afmitor, Sutent, Everolimus, Pancreatin, Zanosar, Mutamycin, Onivyde, Sunitinib, Streptozocin, Mitomycin, Irinotecan liposomal, Bleomycin, Velban, Ifex, Ifosfamide, Vinblastine, Bleo 15k, Iomustine, CeeNU, Gleostine, Panretin, Alitretinoin, Votrient, Inlyta, Pazopanib, Torisel, Axitinib, Medroxyprogesterone, Cabometyx, Temsirolimus, 5-fluorouracil, Cemiplimab, Libtayo, Caboplatin, Megestrol, Provera, Anastrozole, Depo-Provera, Depo-subQ provera 104, Depo- Provera Contraceptive, Profimer and their derivatives.
[00043] “Chemotherapy” as used herein, refers to administering Chemotherapeutic agent. [00044] “Circulating tumor cells (CTCs)” as used herein, refers to, but is not limited to, epithelial cells found in circulation with either intact viable nuclei and with fragmented, apoptotic nuclei. CTCs have extravasated from the primary tumor and will ultimately intravasate into a distant tissue forming a metastasis. CTCs can be marked by EPCAM, CK, or any other epithelial or mesenchymal specific marker. CTCs can be marked by MUC4 or epithelial-cancer specific marker. CTCs can be isolated from blood using antibody based separation, magnetic separation, size based separation, magnetic levitation or other means. CTCs can be either negative for blood cell specific markers such as CD45, or positive for blood specific markers, also known as circulating hybrid cells (CHCs).
[00045] “Dormant cells”, as used herein, refers to, but is not limited to, cancer cells that are non-dividing and arrested in any state of the cell cycle. Dormant cells can be present in the primary tumor, in micrometastases, in lymph nodes, in distant tissues, or in residual disease after treatment. Dormant cells can reside within the body for any length of time and can be re activated at any time. Dormant cells can also include cells just after extravasation to a metastatic site such as lung, liver, brain, bone, lymph node and all other tissues.
[00046] “Inflammatory or autoimmune disease(s) or disorder(s),” as used herein, refers to, but is not limited to, inflammatory diseases or disorders, such as by way of non-limiting example, osteoarthritis, or autoimmune diseases or disorders, such as by way of non-limiting example, osteoporosis, psoriasis, oral mucositis, rheumatoid arthritis, inflammatory bowel disease, eczema, kyphosis, herniated intervertebral disc, and the pulmonary diseases, COPD and idiopathic pulmonary fibrosis.
[00047] “Neoadjuvant therapy” as used herein, refers to, but is not limited to, the treatment given as a first step to shrink a tumor before the main treatment, which is usually surgery, is given. Examples of neoadjuvant therapy include chemotherapy, radiation therapy, hormone therapy or other novel therapies that will be recognized by those skilled in the art.
[00048] “Primary therapy”“Primary Treatment” as used herein, refers to but is not limited to the first treatment given for a disease which includes standard set of treatments, such as surgery followed by chemotherapy and radiation. When used by itself, primary therapy is the one accepted as the best treatment. If it doesn’t cure the disease or it causes severe side effects, other treatment may be added in following therapies. Examples of the primary therapy includes but is not limited to, surgical therapy, radiotherapy, chemotherapy, immunotherapy, targeted therapy, hormone therapy, biological therapy or other novel therapies that will be recognized by those skilled in the art. [00049] “Pulmonary disease(s) and disorder(s),” as used herein, refers to, but is not limited to, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, bronchiectasis, and emphysema.
[00050] “Senescence associated dermatological disease(s) and disorder(s)” as used herein, refers to, but is not limited to, psoriasis, vitiligo, and eczema, which are also inflammatory diseases and are discussed in greater detail. Other dermatological diseases and disorders that may be associated with senescence include rhytides (wrinkles due to aging); pruritis (linked to diabetes and aging); dysesthesia (chemotherapy side effect that is linked to diabetes and multiple sclerosis); psoriasis (as noted) and other papulosquamous disorders, for example, erythroderma, lichen planus, and lichenoid dermatosis; atopic dermatitis (a form of eczema and associated with inflammation); eczematous eruptions (often observed in aging subjects and linked to side effects of certain small molecule). Other dermatological diseases and disorders associated with senescence include cutaneous lymphomas, eosinophilic dermatosis (linked to certain kinds of hematologic cancers); reactive neutrophilic dermatosis; pemphigus, cutaneous lupus, pemphigoid and other immunobullous dermatosis fibrohistiocytic proliferations of skin.
[00051] “Senescence related diseases or disorders,” as used herein, refers to, but is not limited to, any of the following diseases or disorders a senescence-related disease or disorder, an inflammatory disease or disorder, an autoimmune disease or disorder, a cardiovascular disease or disorder, a pulmonary disease or disorder, an ophthalmic disease or disorder, a metabolic disease or disorder, a neurological disease or disorder, a senescence-associated dermatological disease or disorder, a nephrological disease or disorder, renal dysfunction, kyphosis, herniated intervertebral disc, frailty, hair loss, hearing loss, muscle fatigue, a gradual loss of function, or degeneration that occurs at the molecular, cellular, tissue, and organismal levels.
[00052] “Senolytic Agent(s),” as used herein, refers to, but is not limited to: Dasatinib, Quercetin, Navitoclax, Piperlongumin, Fisetin, BCL-XL inhibitors A1331852 and A1155463, all Fox04-related peptides and Foxo4-CR3 domain inhibiting peptides and Senolytic Small Molecule(s).
[00053] Senolytic Small Molecule(s) as used herein, refers to any one of the compounds in listed in Table 1.
[00054] “Senolytic Therapy” as used herein refers to administration of Therapeutic Regimes in one or more treatments cycles for prevention or treatment of senescence- associated diseases and disorders and for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis as wherein Senolytic Agents are used alone or in combination or as an adjuvant, or for prevention, inhibition or retarding the progression of a Senescent Related Diseases or Disorders specifically including Cancer and or metastasis of a Cancer.
[00055] Treatment Regime(s) as used herein refers to administration of Senolytic Agents in Therapeutically Effective Dose in composition and in delivery methods and treatment cycles and regimes disclosed in the subject matter of this patent. Treatment Regimes definition includes but not limited to regimes described in Schedule 1 and Impulse, Shock and Sustained regimes variations thereof.
[00056] “Therapeutically Effective Dose”, as used herein, refers to a dose which cause a therapeutic effect to some extent, some effect with respect to one or more of the symptoms of the disease, and includes curing a disease.“Curing” means that the symptoms of active disease are eliminated. However, certain long-term or permanent effects of the disease may exist even after a cure is obtained (such as where extensive tissue damage is present). The Therapeutically Effective Dose is typically one which is sufficient to achieve the desired effect and may vary according to the nature and severity of the disease condition, and the potency of the particular Senolytic Small Molecule administered, its purity, and its composition (e.g. 90%, or 95% or 98% etc.). In some embodiments, different doses may be employed for embodiments where the Senolytic Small Molecule(s) is administered for preventive use rather than for treatment of an active disease. Therapeutically Effective Dose definition shall include the dosage for the Senolytic Agents as disclosed in Schedule 1.
[00057] “Subject,” as used herein, refers to a target of a treatment or therapy, including but not limited to, a human or a non-human mammal, a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate or a bird, for example, a chicken, as well as any other vertebrate or invertebrate.
[00058] “Treat,”“treatment,” or“treating,” as used herein, refers to administration of a composition for therapeutic purposes and includes the Treatment Regimes as disclosed in the subject matter of the patent.
[00059] Disclosed herein is a successful model for the interaction network between the domains of Fox04 and p53 at atomic resolution, by which the selective inhibition of the action of Fox04 on p53 was achieved. [00060] Both Fox04 and p53 have multiple domains with distinct functions. Prior art investigations as to possible interactions of Fox04 with p53 implied that Fox04 can interact with p53 through multiple domains. Not intending to be bound by theory, these potential interactions between Fox04 and p53 may explain the cause of the restrain elicited by Fox04 on the apoptotic function of p53. Hence in the rational design, the possible complexes of Fox04 and p53 was elucidated at the atomic resolution.
[00061] The FoxO family of proteins express similar domain compositions such that they contain DNA binding domains entitled as the Forkhead (FH) domain and C-terminal domains entitled as CR1, CR2 and CR3 for transactivation. The FH and CR3 domains interact with each other and the binding surface of these interactions have been resolved in an NMR based study using Fox03. FH includes basic (positively charged) and hydrophobic amino acids (R153, R154, W157 and G158) that contribute to this intramolecular interaction, while a central portion of CR3 that includes acidic (negatively charged) and hydrophobic amino acids (the amino acids from D623 to M633) interacts with the FH.
[00062] This NMR study also shed light on the action of Fox03 on p53. Essentially, in vitro pull down assays were used to identify the critical domain(s) of both p53 and Fox03 for their intermolecular interactions. According to these results, the DNA binding domain of p53 (p53DBD) was found to be necessary for binding to the Fox03, while the C-terminal portion of the Fox03 that includes the CR3 domain was revealed to be the most critical domain for p53 interaction. Further experiments showed that addition of the p53DBD disrupted the intramolecular interaction between the FH and the CR3 domains of Fox03, in that the binding affinities of both of the FH-CR3 and p53DBD-CR3 complexes were comparable with each other. As resolved by NMR shift experiments, the binding interface of the CR3 domain of Fox03 has been overlapped in both of the complexes formed by the FH domain of Fox03 and the p53DBD. This particular finding implied a competition of the FH domain of Fox03 with the p53DBD to bind to the same surface on the CR3 domain of Fox03. Sequestration of the CR3 domain, particularly from the surface that it binds to p53DBD will circumvent its interaction with the p53DBD, liberating the p53DBD and its transcriptional activity to initiate apoptosis.
[00063] The FoxO proteins are very similar proteins that share high sequence similarity and domain composition. This similarity in the protein sequence was further observed at the functional level by in vivo experiments in mice. The resulting functional redundancy will suggest the validity of the findings on the Fox03 for Fox04. Hence, in connection to the insights obtained from the NMR findings [1], blocking of the CR3 domain of Fox04 has the potential to inhibit the interaction between Fox04 and p53 and thus liberate p53 in senescent cells. [1] Wang, F., et al, Biochemical and structural characterization of an intramolecular interaction in F0X03a and its binding with p53. J Mol Biol, 2008. 384(3): p. 590-603
[00064] Interference in the p53 structure or pathway may bear harmful consequences to cells and thus should be avoided by any means. Nevertheless, the structural findings suggested that the CR3 domain of Fox04 binds to and possibly inhibits the function of the p53DBD. Blocking of the CR3 domain will inhibit“the inhibition of the p53DBD” and will promote the function of p53DBD. Experiments conversely showed that promoting the inhibition of p53 by an oncoprotein, named gankryin, enhanced tumor growth. Gankyrin acts as a promoter of MDM2 which is the well-known inhibitor of p53. Thus interference with the CR3 domain of Fox04, without any direct interference with the p53 protein, will have the potential to remove the said restrain on the p53 mediated apoptosis by Fox04, eradicating the senescence.
[00065] As described in the PCT Application Serial No. PCT/GB2018/052812 the CR3 domain of Foxo4 interacts with the DBD of p53 leading to inhibition of the transcriptional activity of p53 required for apoptosis. Thus, blocking of CR3 domain of Foxo4 is critical to the efforts to release p53 DBD to induce apoptosis. In the cell backgrounds wherein Foxo4-p53 interaction is dominant such as in senescence and melanoma, and/or wherein p53 DBD is similarly blocked by CR3 similar domains from other proteins, small molecules and/or peptides targeting the CR3 domain of Foxo4 can be used as Senolytic Agents. Relying on this paradigm, inventors screened one of the largest virtual ligand databases, ZINC 15 that holds more than 1 billion compounds. Initially, the ZINC 15 database were screened to collect all of the positively charged compounds at neural pH owing to the high net negative charge of the CR3 domain. A total of 141,971, 146 different compounds were collected. This library was screened to identify the potential ligands with high binding affinity towards the CR3 domain of Foxo4.
[00066] The initial screenings allocated 30 million selected compounds from the library composed of -150 million compounds. The initial screening was performed by docking. For this step, 30 million compounds were docked to 20 different conformations of the CR3 domain, which were obtained from molecular dynamics simulations performed by using two different force fields. The top scoring 100 thousand ligands were progressed to next round of screening according to their docking scores. Next, 1st round of free energy calculations was conducted by using the MM-GBSA method and top scoring 2500 ligands were selected. For these 2500 ligands, Amber scoring was carried out to select the top scoring 100 ligands. These 100 small molecules were extensively analyzed by molecular dynamics simulations for 200 ns. The last stable period of the MD simulations was used to calculate the binding free energy of these molecules. Top scoring 12 ligands were selected to have a strong binding affinity toward the CR3 domain. Ligand efficiency (LE) scores of these ligands were also assessed, the score which gives the binding free energy per nonhydrogen atom of the ligand. Overall a total of 12 ligands were found to strongly bind to the CR3 domain after a throughout screening work flow (Figure 1). The binding affinities and the ligand affinities of the small molecules listed in Table 1 shows that they are able to inhibit the CR3 domain of Foxo4, thereby inducing p53 dependent apoptosis. Figure 1-12 shows the complexes obtained at the end of 200 ns of MD simulation.
[00067] In one embodiment, the small molecules listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders. In another embodiment the small molecules that carry a similar chemical scaffold to the ones listed in the Table 1 (Figure 1-12) can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
[00068] In one embodiment, the small molecules found in the ZINC 15 database and will be screened in the successive virtual screening rounds by following the same procedure described herein, to treat senescence-associated diseases and disorders if they possess that strong binding affinities and high ligand efficiency values to effectively inhibit Fox04.
[00069] In another embodiment, other inhibitors of HIV Reverse transcriptase can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders. In another embodiment the small molecules that carry a similar chemical scaffold to one of the inhibitors of HIV Reverse transcriptase listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
[00070] In another embodiment, other inhibitor of reductases can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders. In another embodiment the small molecules that carry a similar chemical scaffold to one of the inhibitors of inhibitor of reductases listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence- associated diseases and disorders.
[00071] In another embodiment, other anti-cancer agents can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders. In another embodiment the small molecules that carry a similar chemical scaffold to one of other anti-cancer agents listed in the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
[00072] In another embodiment, the small molecules that carry a similar function and/or chemical scaffold to one of small molecules numbered 5, 6, 7, 8, 10, 11 of the Table 1 can be used to inhibit Fox04 thereby to treat senescence-associated diseases and disorders.
[00073] Table 1 lists the small molecules that possess strong binding affinity toward the CR3 domain of Foxo4 and thus can be re-purposed as a senolytic.
[00074] In some embodiments, methods for selectively inducing apoptosis in (e.g., killing) senescent cells in a subject who has a senescence-associated disease or disorder may generally comprise administering one or more of the Senolytic Small Molecule(s) to the subject in need thereof, for example, according to one or more of the administration methods described herein. For example, in some embodiments, the method may comprise causing an artificial compound comprising one of the chemical scaffolds of the small molecules listed in the Table 1 to interfere with the CR3 domain of Forkhead box protein 04 (Fox04) of the senescent cell.
[00075] Method of Use of the Senolytic Agents
[00076] The proportion of senescent cells in a tissue of a mammal varies with the biological age and could vary substantially depending on the cohort that mammal subject belongs to. Moreover, the proportion of senescent cells present may further vary with the type of the tissue in a given subject. These variations may create a challenge in specifying a dose for the Senolytic Agent for a rejuvenation therapy. Additional complexities in specifying a dose arise when said senescent cell proportion is above a certain threshold and therefore accelerated apoptosis could result in frailty. Fortunately, unlike for cancer interventions, complete elimination of senescent cells may not be necessary for achieving beneficial effects.
[00077] In various embodiments, the Senolytic Small Molecules may be administered via any suitable methodology. By way of non-limiting example, disclosed herein three different treatment regimes, namely an Impulse Regime, a Sustained Regime, and Gentle Regime via which the Senolytic Small Molecule(s) may be administered.
[00078] In some embodiments, a method is provided for treating a senescence-associated disease or disorder comprising administering to a subject in need thereof a Therapeutically Effective Dose of the Senolytic Small Molecule(s), for example, the Senolytic Small Molecule(s) may be administered intermittently in one or multiple treatment cycles. Each treatment cycle may extend over 1 or 2 or 3 days. Each administration may include equivalent doses adjusted so as to cumulatively reach the Therapeutically Effective Dose at the end of each treatment cycle. In some embodiments, the Therapeutically Effective Dose is administered equivalently in a single day or intermittently in two consecutive days or in three consecutive days or 2 or 3 administration in alternate days. This regime of administration of the Senolytic Small Molecules is referred to herein as the Impulse Regime. In the Impulse Regime, the Therapeutically Effective Dose is achieved through one or two subsequent administration(s). In some embodiments, after the first cycle, there may be a two week-four-week senescence clearance interval, for example, allowing a period of time effective for a decrease in senescent cells. In some embodiments, the subject is evaluated, both before the Impulse Regime and after Senescence Clearance Interval period, by one skilled in the art to determine levels of various SASP markers for determination of the Therapeutically Effective Dose and Follow-up Treatment, respectively.
[00079] Additionally, or alternatively, in some embodiments, the Therapeutically Effective Dose is achieved through a single or multiple administrations in a period of 1-3 weeks. The quantity of the Senolytic Small Molecule for each administration is equivalent and adjusted to cumulatively reach the Therapeutically Effective Dose at the end of the treatment. This regime of administration of the Senolytic Small Molecules is referred to as the Sustained Regime. The Therapeutically Effective Dose administered in the Sustained Regime may be higher than the Therapeutically Effective Dose as would be administered in the Impulse Regime.
[00080] Additionally, or alternatively, in some embodiments the Therapeutically Effective Dose is administered intermittently in one or multiple treatment cycles wherein each treatment cycle comprised of 1 or 2 or 3 or 4 or 5 or 6 administration days equally distributed in 1-3 weeks where each administration is in equivalent doses adjusted to cumulatively reach the Therapeutically Effective Dose at the end of each treatment cycle.
[00081] In some embodiments, after the first cycle, there is a two or three weeks’ senescence clearance interval allowing a period of time effective for a decrease in senescent cells. In some embodiments, the subject may be evaluated, both before the Sustained Regime and after Senescence Clearance Interval period, by one skilled in the art based on the levels of various SASP markers for determination of the Therapeutically Effective Dose and Follow-up Treatment, respectively.
[00082] In some embodiments, the Therapeutically Effective Dose is achieved through single or multiple administration cycles in the period of 3-4 weeks. This regime of administration of the Senolytic Small Molecules is referred to herein as the Gentle Regime. Both the Therapeutically Effective Dose and the quantity of the Senolytic Small Molecule for a single administration in the Gentle Regime may be lower than those in the Impulse Regime and Sustained Regime. The Therapeutically Effective Dose may be administered intermittently in one or multiple treatment cycles. Each treatment cycle may be comprised of 1 or 2 or 3 or 4 or 5 or 6 or 7 or 8 or 9 administration days equally distributed in 3-4 weeks. Each administration may be in an equivalent dose adjusted so as to cumulatively reach the Therapeutically Effective Dose at the end of each treatment cycle.
[00083] In some embodiments, after the first treatment cycle, there is a two or three weeks Senescence Clearance Interval allowing a period of time for the treatment becoming effective with gradual decrease in senescent cells. In some embodiments, the subject may be evaluated both before the Gentle Regime and after Senescence Clearance Interval period, by those skilled in the art based on the levels of various SASP markers for determination of the Therapeutically Effective Dose and Follow-up Treatment, respectively.
[00084] In various embodiments, the Therapeutically Effective Dose amount is delivered to a subject in need thereof by any one of several routes known to a person skilled in the art. In various embodiments, by way of non-limiting example, the Therapeutically Effective Dose is delivered orally, intravenously, intraperitoneally, by infusion (e.g., a bolus infusion), subcutaneously, enteral, rectal, intranasal, by injection, inhalation, buccal, sublingual, intramuscular, transdermal, intradermal, topically, intraocularly, vaginally, rectally, intrathecally or by intracranial injection, or by directly into the target tissue including a tumor or organ, or subcutaneous route or by some combination of the above thereof. In some embodiments the Therapeutically Effective Dose may be delivered in combinations of other Senolytic Agents and/or with chemotherapy. In some embodiments the Therapeutically Effective Dose may be delivered in combinations of other Senolytic Agents and/or with chemotherapy concomitantly or concurrently or not concurrently but in appropriate intervals. In some embodiments, a delivery method includes controlled or sustained release drug(s), drug- coated or permeated stents for which the drug is the Senolytic Small Molecule. In some embodiments Therapeutically Effective Dose amount is further adjusted depending on the delivery route and selected Treatment Regime.
[00085] As discussed herein below, examples 1-5 indicate that the Senolytic Small Molecule(s) can safely be administered at doses of 5 mg/Kg, 10 mg/Kg, and 15 mg/Kg. More particularly, Example 4 indicates that the Senolytic Small Molecule(s) is non-toxic at levels up to 100 mg/kg. Due to the molecular similarity of the various Senolytic Small Molecules in the Table 1 and compound possessing the same chemical scaffold thereto, atomistic structures are similar and therefore they exert their senolytic effects in a similar way. In some embodiments, the Therapeutically Effective Dose amount can further depend upon the patient's height, weight, sex, age and medical history.
[00086] In some embodiments, treatment methods, such as according to the Impulse Regime, Sustained Regime, or Gentle Regime, may include monitoring the population of the senescent cells on convenient biological samples taken from the subject at the beginning and 2 or 4 weeks after the treatment to determine the effectiveness of the therapy and or whether a second or a third treatment course is required. In some embodiments, an individualized treatment course may be implemented and may include monitoring of senescent cell population at the beginning of a therapy and a certain period after each treatment course and adjusting the treatment course or the dose or the treatment regime. Additionally, or alternatively, in some embodiments the administration of a Therapeutically Effective Dose may depend upon the subject’s state of health and subject’s response to the treatment throughout the period of the therapy. In some embodiments, the biological sample shall be the skin biopsy specimens obtained from skin tissue of the subject is collected with minimally invasive methods. In some embodiments, the detection of the senescent cells may be achieved using senescence associated markers
[00087] Although senescent cells diverge from other quiescent and terminally differentiated cells, they do not display a unique phenotype but a variety of phenotypes which define the senescent state. Hallmarks of the senescent cells in this state include permanent and irreversible growth arrest; increase in cell size; expression of senescence-associated beta-galactosidase (SA- b-Gal) enzyme resulting in increased lysosomal content; expression of a tumor suppressor, plgiNK4a jw0 hiomarkers have been extensively used for identification of senescent cells:
SA-P-Gal: Enzyme activity assayed at pH 6 using X-Gal as the substrate. p i 6INK4a : Monitoring the expression levels of pl6INK4a protein which is a CDK4/6 inhibitor and is involved in maintenance of growth arrest.
[00088] In some embodiments, the Senolytic Small Molecule(s) do not have to be continuously present to exert an intended effect. For example, brief disruption of pro-survival pathways is adequate to kill senescent cells and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells. Thus, the Senolytic Small Molecule(s) can be effective as Senolytic Small Molecules when administered intermittently. [00089] It has already been reported that other known Senolytic Agents such as the tyrosine kinase inhibitor (D) and the flavonoid, quercetin (Q), were shown to induce apoptosis in senescent cells. Intermittent administration of D + Q alleviated frailty, neurological dysfunction, osteoporosis, and vertebral disk degeneration related to loss of glycosaminoglycans on an accelerated aging-like state. Furthermore, in mice with impaired mobility due to radiation of one of their legs 3 months previously, treadmill endurance improved within 4 days after completing a single course of D + Q. Said improvement persisted for at least 7 months. D + Q has an elimination half-life of a few hours. These outcomes following intermittent or single courses of agents with short elimination half-lives are consistent with the long-lasting type of effect expected from reducing senescent cell abundance, as opposed to what would be expected if D + Q had to be continuously present to suppress or activate cellular processes by occupying a receptor or acting on an enzyme.
[00090] Thus, intermittent rather than continuous treatment with Senolytic Agents may be effective in alleviating senescence-related diseases or disorders, allowing these agents to be administered during periods of good health and potentially decreasing risk of side-effects. In all of the shock dose regimes, there will be an optional follow-up treatment wherein the Therapeutically Effective Dose can be increased or decreased. In between the initial treatment and follow-up treatment there will be a non-treatment interval, wherein the non-treatment course duration could vary depending on the subject’s state of health and/or the subject’s response to the treatment which can be monitored by the said detection of senescent cells in the subject throughout the period of the therapy.
[00091] In some embodiments, at least one Senolytic Small Molecule may be administered in a Therapeutically Effective Dose with at least one or more other available Senolytic Agent which together act additively or synergistically to selectively kill senescent cells. By way of non-limiting example, in some embodiments, a Senolytic Small Molecule dose administration is optimization for use with the Senolytic Agents including but not limited to Dasatinib, Quercetin, Navitoclax, Piperlongumin, Fisetin, BCL-XL inhibitors A1331852 and A1155463, FOX04-related peptide, Foxo4-CR3 domain inhibiting peptides or other small molecules or combinations of these.
[00092] The effectiveness of the Senolytic Small Molecule treatment can be determined by a person skilled in the medical and clinical arts by employing combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein, may be used for monitoring the health status of the subject, in addition to the senescent cell monitoring described herein for the Treatment Regimes. For example in one embodiment the effectiveness of the Senolytic Small Molecules when treating a subject for pulmonary diseases and disorders Pulmonary Function Tests (https://www.nhlbi.nih.gov/health/health-topics/topics/lft) can be conducted. Pulmonary function tests, or PFTs, measure how well subject’s lungs work. They include tests that measure lung size and air flow, such as spirometry and lung volume tests. Other tests measure how well gases such as oxygen get in and out of subject’s blood. These tests include pulse oximetry and arterial blood gas tests. Another pulmonary function test, called fractional exhaled nitric oxide (FeNO), measures nitric oxide, which is a marker for inflammation in the lungs. The subject may have one or more of these tests to diagnose lung and airway diseases, compare subject’s lung function to expected levels of function, monitor if your disease is stable or worsening, and see if said senolytic treatment is beneficial.
[00093] It should be noted that Fox04 is expressed in a tissues during fast growth or regeneration. As such, in some embodiments a senolytic therapy using the Senolytic Small Molecule(s) may exclude such cohorts.
[00094] In some embodiments using the Senolytic Small Molecule(s) on a subject, senescent cell population in the Biological skin sample of the subject undergoing treatment will be monitored before, and after a treatment course. In some embodiments, monitoring is performed during a treatment course and /or between the treatment courses or cycles.
[00095] Method of Use of the Senolytic Agents -Schedule 1
[00096] In some embodiments, the Senolytic Agent comprises any one of the Senolytic Small Molecules or combinations Senolytic Agents.
[00097] In some embodiments, the Senolytic Agent is administered in a treatment window comprising 11 to 28 days.
[00098] By way of non-limiting example, in some embodiments, the Senolytic Agent is administered daily or alternating days for 14 days followed by minimum 14 days off.
[00099] In some embodiments, the Senolytic Agent is administered daily for 13 days followed by minimum 14 days off.
[000100] In some embodiments, the Senolytic Agent is administered daily or alternating days for 12 days followed by minimum 14 days off. [000101] In some embodiments, the Senolytic Agent is administered daily for 11 days followed by minimum 14 days off.
[000102] In some embodiments, the Senolytic Agent is administered daily or alternating days for 10 days followed by minimum 14 days off.
[000103] In some embodiments, the Senolytic Agent is administered daily for 9 days followed by minimum 14 days off.
[000104] In some embodiments, the Senolytic Agent is administered daily or alternating days for 8 days followed by minimum 12 days off.
[000105] In some embodiments, the Senolytic Agent is administered daily for 7 days followed by minimum 12 days off.
[000106] In some embodiments, the Senolytic Agent is administered daily or alternating days for 6 days followed by minimum 12 days off.
[000107] In some embodiments, the Senolytic Agent is administered daily for 5 days followed by minimum 12 days off.
[000108] In some embodiments, the Senolytic Agent is administered daily or alternating days for 4 days followed by minimum 12 days off.
[000109] In some embodiments, the Senolytic Agent is administered daily for 3 days followed by minimum 10 days off.
[000110] In some embodiments, the Senolytic Agent is administered daily for 2 days followed by minimum 10 days off.
[000111] In some embodiments, the Senolytic Agent is administered for 1 day followed by minimum 10 days off.
[000112] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0. lmg/kg to 20 mg/kg.
[000113] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0. lmg/kg to 15 mg/kg. [000114] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0.1 mg/kg to 10 mg/kg.
[000115] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0. lmg/kg to 5 mg/kg.
[000116] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0.1 mg/kg to 3 mg/kg.
[000117] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 0.1 mg/kg to 0.5 mg/kg.
[000118] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 1400 mg per day.
[000119] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 1000 mg.
[000120] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 700 mg.
[000121] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 200 mg.
[000122] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 35 mg.
[000123] In some embodiments, the Senolytic Agent is administered daily for 14 days in a dose of about 3.5 mg.
[000124] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 1400 mg.
[000125] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 1000 mg.
[000126] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 700 mg. [000127] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 200 mg.
[000128] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg to 35 mg.
[000129] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 1400 mg.
[000130] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 1000 mg.
[000131] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 700 mg.
[000132] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 200 mg.
[000133] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 35 mg.
[000134] In some embodiments, the Senolytic Agent is administered daily for 7 days in a dose of about 3.5 mg.
[000135] In some embodiments, the Senolytic Agent is administered in one of the above doses daily for 1, 2, 3, 4, 5, or 6 days or 8, 9, 10, 11, 12, 13 days. Additionally, or alternatively, in some embodiments, the Senolytic Agent is administered in one of the above doses are administered alternative daily for 2, 4, or 6 days or 8, 10, 12, 14 days.
[000136] In some embodiments the doses described in this Schedule 1 shall be a Therapeutically sufficient dose for the administration of any one of the Senolytic Agents.
[000137] In some embodiments Therapeutically Effective Dose is administered by any one of several compositions and delivery routes known to a person skilled in the art and as disclosed in the subject matter. By way of non-limiting example, the composition may be delivered orally, intravenously, intraperitoneally, by infusion (e.g., a bolus infusion), subcutaneously, enteral, rectal, intranasal, by injection, inhalation, buccal, sublingual, intramuscular, transdermal, intradermal, topically, intraocularly, vaginally, rectally, intrathecally, or intracranially, or by some combination thereof.
[000138] In some embodiments the regiment of administration in this Schedule 1 shall be the method of administration of any one of the Senolytic Agents.
[000139] In some embodiments, the Therapeutically Effective Dose is delivered in methods of the disclosed subject matter including but limited to as in controlled or sustained release drug(s), drug-coated or permeated stents for which the drug is the Senolytic Agent.
[000140] In some embodiments Senolytic Small Molecule(s) is administered as described in this Schedule 1 together with another Senolytic Agent including but not limited to Navitoclax (ABT-263), Fisetin, A133185240, A115546340, Quercetin, Dasatinib, Piperlongumine, 17- AAG (tanespimycin), Geldanamycin 17-DMAG (alvespimycin), Famotidine, Deferoxamine, Mitoxantrane, Lapatinib, Neratinib with therapeutically sufficient doses for each of these Senolytic Agents but strictly in the regiments as described herein this Schedule 1.
[000141] The effectiveness of the Senolytic Therapy can be determined by a person skilled in the medical and clinical arts by employing combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein, may be used for monitoring the health status of the subject, in addition to the monitoring described herein for the treatment regimes.
[000142] Medical Therapies using the Senolytic Small Molecules
[000143] The effectiveness of the Senolytic Small Molecule(s) with respect to treating a senescence-associated disease or disorder described herein can readily be determined by a person skilled in the medical and clinical arts. One or any combination of diagnostic methods appropriate for the particular disease or disorder, which methods are well known to a person skilled in the art, including physical examination, subject self-assessment, assessment and monitoring of clinical symptoms, performance of analytical tests and methods, including clinical laboratory tests, physical tests, and exploratory surgery, for example, may be used for monitoring the health status of the subject and the effectiveness of the Senolytic Small Molecule. The effects of the methods of treatment described herein can be analyzed using techniques known in the art, such as comparing symptoms of subjects suffering from or at risk of a particular disease or disorder that have received the composition comprising the Senolytic Small Molecule with those of subjects who were not treated with the Senolytic Small Molecule or who received a placebo treatment.
[000144] A subject in need of treatment with the Senolytic Small Molecule(s) as described herein may be a human or may be a non-human primate or other animal (i.e., veterinary use) who has developed symptoms of a senescence cell-associated disease or disorder or who is at risk for developing a senescence cell-associated disease or disorder. Non-human animals that may be treated include mammals, for example, non- human primates (e.g., monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, rabbits), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, bovine, elephants, bears and other domestic, farm, and zoo animals.
[000145] In some embodiments, administration of the Senolytic Small Molecule(s) described herein can prolong survival when compared to expected survival if a subject were not receiving treatment. Subjects in need of treatment include those who already have the disease or disorder as well as subjects prone to have or at risk of developing the disease or disorder, and those in which the disease, condition, or disorder is to be treated prophylactically. A subject may have a genetic predisposition for developing a disease or disorder that would benefit from clearance of senescent cells or may be of a certain age wherein receiving the Senolytic Small Molecule would provide clinical benefit to delay development or reduce severity of a disease, including an age-related disease or disorder.
[000146] In some embodiments, use of the Senolytic Small Molecules may be restricted during wound healing (including pre- or post-operations). When a wound is present, senescent cells may be induced around the wound. Senescent cells make growth factors that are required for wound healing. However, this innate mechanism is not disturbed unless the Senolytic Small Molecule(s) is administered at the time of the wound healing.
[000147] Method of Use of the Senolytic Small Molecules in Senescence- Associated Diseases and Disorders
[000148] Cellular senescence is a cell fate that involves essentially irreversible replicative arrest, apoptosis resistance, and frequently increased protein synthesis, metabolic shifts with increased glycolysis, decreased fatty acid oxidation, increased reactive oxygen species generation, and acquisition of a senescence-associated secretory phenotype (SASP). [000149] Methods are provided herein for treating conditions, diseases or disorders related to, associated with, or caused by cellular senescence in a subject in need thereof.
[000150] In some embodiments the methods of use of Senolytic Agents for treatment of senescence-associated diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of senescence-associated diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of senescence-associated diseases and disorders certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000151] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000152] Method of Use of the Senolytic Small Molecule(s) for treatment of general Age- Related Diseases and Disorders
[000153] In some embodiments, the Senolytic Small Molecule(s) inhibits senescence of adult stem cells or inhibits accumulation, kills, or otherwise facilitates removal of adult stem cells that have become senescent. Therefore, the Senolytic Small Molecules may also be useful for treating or preventing of an age-related disease or disorder that occurs as part of the natural aging process or that occurs when the subject is exposed to a senescence inducing agent or factor (e.g., irradiation, chemotherapy, smoking tobacco, high-fat/high sugar diet, other environmental factors).
[000154] In some embodiments, frailty as an aging-associated decline may be treated or prevented (i.e., the likelihood of occurrence of is reduced) by administering the Senolytic Small Molecule. The effectiveness of the senolytic therapy can be measured by monitoring the frailty index of the patient.
[000155] In some embodiments, the age related disease or disorder is scoliosis. Effectiveness of the senolytic therapy are measured by, inter alia, physical examination of spine, ribs, hips and shoulders and/or X-RAY, CT and/or MRI to determine bone curvature.
[000156] In some embodiments the methods of use of Senolytic Agents for treatment of general age-related diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of said Treatment Regimes for age-related diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of general age-related diseases and disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000157] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells on a biopsy sample taken from the subject at the beginning and throughout the period of the therapy and thus monitoring the target cell population decline.
[000158] The effectiveness of a method of treatment described herein may be manifested by reducing the number of symptoms of an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus, decreasing the severity of one or more symptoms, or delaying the progression of an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus. In some embodiments, preventing an age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus refers to preventing (i.e., reducing the likelihood of occurrence) or delaying onset of an age-related disorder or age- sensitive trait associated with a senescence-inducing stimulus, or reoccurrence of one or more age-related disorder or age-sensitive trait associated with a senescence-inducing stimulus. [000159] Method Use of the Senolytic Small Molecule(s) in Tissue Rejuvenation
[000160] It has been reported in the reference that a Senolytic Agent could influence the health span of mice in which senescence and the concomitant loss of tissue homeostasis were allowed to develop spontaneously as a consequence of aging. The results of in vivo mouse experiments indicate that a Senolytic Agent can reduce cellular senescence and counteract hair loss (objectively measured as the fur density) and general frailty (objectively measured by increased running wheel activity or muscle size increase) in mice.
[000161] Tissues contain high levels of senescent cells, which due to chronic SASP secretion, would inflict permanent reprogramming on their neighboring cells. Senescence was recently shown to trigger tissue reprogramming in vivo, leading to Nanog-positive cells in the vicinity of areas of senescence. Senescent cells might thus trigger reprogramming of neighboring cells into more pluripotent cells. However, since the release of SASP factors (such as IL6) is continuous, they would effectively make this change permanent and keep their neighboring recipient cells locked in this stem like state. If the number of senescent cells are reduced to that of relatively young tissues (with fewer senescent cells) then a transient SASP response, causing temporary cell reprogramming and subsequent proliferation/differentiation responses would be able to replenish damaged and lost cells.
[000162] In some embodiments the Senolytic Agents is applied in Treatment Regimes for tissue rejuvenation therapy of a scalp tissue or an osteoarthritic joint or a pulmonary tissue or a renal tissue. In some embodiments senolytic Agents is administered in Treatment Regimes for scalp treatment and hair regeneration by applying the senolytic tissue externally.
[000163] In some embodiments the method of delivery of Senolytic Small Molecules in treatment of senescence-associated diseases and disorders are detailed herein this document.
[000164] In some embodiments the methods of use of Senolytic Agents for tissue rejuvenation comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for tissue rejuvenation include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment for tissue rejuvenation certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000165] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000166] Method Use of the Senolytic Small Molecule(s) for treatment of Senescence- Associated Dermatological Disease or Disorders
[000167] Senescence-associated diseases or disorders treatable by administering the Senolytic Agents include dermatological diseases or disorders. Several dermatological diseases or disorder are associated with the accumulation of senescent cells. In some embodiments, said dermatological diseases and disorders are psoriasis, vitiligo and eczema. Among them, vitiligo is an acquired disorder characterized by depigmentation. In addition to genetic susceptibility and autoimmunity, oxidative stress and underlying premature melanocyte senescence are considered to be key factors in vitiligo progression. Melanocytes from non-lesion vitiligo lesions were shown to exhibit a pre-senescent phenotype in vitro.
[000168] In some embodiments the methods of use of Senolytic Agents for treatment of dermatological diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Agents for treatment of dermatological diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of dermatological diseases and disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders. [000169] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000170] In certain appropriate senescence-associated dermatological diseases and disorders the method of application of the Senolytic Small Molecules could be external on the surface of the body such that skin cell penetration of said molecule is achieved as described in detail in the disclosed subject matter.
[000171] Method Use of the Senolytic Small Molecule(s) for treatment of Inflammatory and Autoimmune Diseases and Disorders
[000172] Chronic inflammation is a major factor in wide range of disease associated with old age. Recently a global clinical trial of 10,000 subjects who had previous heart attacks of an anti inflammatory drug which targeted a portion of the inflammatory pathway — focusing specifically on interleukin- lbeta (IL-Ib), a cell-signaling protein - showed that it reduced their risk of further heart attacks or strokes. The drug prevents these cells from going into overdrive, but presumably leaves the remaining immune system intact. The chronic inflammatory diseases include rheumatoid arthritis when inflammation occurs in joints. Individuals with Rheumatoid arthritis exhibit accelerated immunosenescence possibly as a result of inflammatory mechanisms. Therefore, the Senolytic Agents are suitable for curing or managing chronic inflammation by substantially stopping SASP by killing the senescent cells.
[000173] The senescence-associated secretory phenotype (SASP) comprises a range of different proteins, including several proteins known to play a role in aging and age-related diseases, including chemokines such as CCL2 and CLL11 and prominent interleukins such as IL-1, IL-6 and IL-12. When above a certain threshold, such factors can significantly impair tissue function and impair functioning of neighboring cells. As such, senescent cells are thought to be major contributors of inflammation; a theory stating that low, but chronic, levels of inflammation are drivers of age-related decline.
[000174] The definition of chronic inflammatory diseases includes osteoarthritis which is characterized by progressive tissue remodeling and loss of joint function and paralleled by increased age. It is the most prevalent disease of the synovial joints. During osteoarthritis, levels of various senescence markers increase in chondrocytes with SASP profiles similar to classical senescent cells which in turn supports the hypothesis that senescence of cells within joint tissues may play a pathological role in the causation of osteoarthritis. Therefore, the Senolytic Agents is suitable to cure or manage chronic inflammation by substantially stopping SASP by killing the senescent cells.
[000175] In some embodiments, administration of Senolytic Therapy is effective in the treatment of osteoarthritis. During the senolytic treatment, the osteoarthritis disease parameters which are measured include, inter alia, joint pain, redness, stiffness and/or swelling and joint motion range, X-RAY and/or MRI for bone spurs, blood tests and joint fluid analyses to rule out other causes.
[000176] In some embodiments, administration of Senolytic Therapy is effective in the treatment of kyphosis. During the senolytic treatment, the kyphosis disease parameters which are measured include, inter alia, measurement of spine curvature by X-RAY, CT and/or MRI.
[000177] In some embodiments the methods of use of Senolytic Agents for treatment of inflammatory or autoimmune diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of inflammatory or autoimmune diseases and disorder include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of inflammatory or autoimmune diseases and disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000178] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000179] Method Use of the Senolytic Small Molecule(s) for treatment of Cardiovascular Diseases and Disorders [000180] In one embodiment, the senescence-associated disease or disorder treated by the methods described herein is a cardiovascular disease. The cardiovascular disease is often caused by atherosclerosis and is the primary cause of mortality in the developed countries.
[000181] There is a growing body of evidence for inflammation as the key process in atherosclerosis into clinical and public health practice. Atherosclerosis is a disease of major arteries in which high levels of low-density lipoprotein bearing oxidative modifications accumulate in vessel walls, attracting phagocytic immune cells to form plaques. Telomere shortening and oxidative stress caused by smooth-muscle proliferation and declining levels of endothelial nitric oxide synthase during plaque formation and expansion cause senescence induction. Human and mouse atheromas have been reported to exhibit senescent vascular smooth muscle and endothelial cells. Basic science and epidemiological studies have developed an impressive case that atherogenesis is essentially an inflammatory response to a variety of risk factors and the consequences of this response lead to the development of acute coronary syndrome. These findings raise the possibility of multi-step involvement of senescent cells in atherogenesis. Therefore, in some embodiments, the Senolytic Agents, administered as Treatment Regimes are suitable to slow down the progression of cardiovascular disease by reducing the chronic inflammation in the body substantially by stopping SASP.
[000182] During the Senolytic Therapy of (cardio)vascular disease, (cardio)vascular disease parameters which are measured include, inter alia, cardiac ejection fraction, blood vessel stiffness and blood pressure.
[000183] Senolytic Therapy is effective in treatment of atherosclerosis. During the Senolytic Small Molecule treatment atherosclerosis disease parameters which are measured include blood tests including measurements of cholesterol, glucose, electrocardiogram, angiography, computerized tomography scan and/or ophthalmoscopy.
[000184] In some embodiments the methods of use of Senolytic Agents for treatment of cardiovascular diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of cardiovascular diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of cardiovascular diseases and disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000185] The effectiveness of one or more Senolytic Small Molecules for treating or preventing (i.e., reducing or decreasing the likelihood of developing or occurrence of) a cardiovascular disease (e.g., atherosclerosis) can readily be determined by a person skilled in the medical and clinical arts. Health status of the subject may be monitored by one or any combination of diagnostic methods, including but not limited to physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein and practiced in the art (e.g., angiography, electrocardiography, stress test, non-stress test). The effects of the treatment of the Senolytic Small Molecule can be analyzed using techniques known in the art, such as comparing symptoms of subjects suffering from or at risk of cardiovascular disease that have received the treatment with those of subjects without such a treatment or with placebo treatment.
[000186] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000187] Method Use of the Senolytic Small Molecule(s) for treatment of Pulmonary Diseases and Disorders
[000188] In one embodiment, the senescence-associated disease or disorder treated by the Senolytic Therapy methods described herein is a Pulmonary Diseases and Disorders.
[000189] In some embodiments, Pulmonary Disease and Disorders treated by administration of Senolytic Therapy described herein include Chronic obstructive pulmonary disease (COPD)/emphysema characterized by lung inflammation induced by accelerated lung aging involving inflammatory mediators such as tumor necrosis factor alpha, interleukin- 1, interleukin-6, reactive oxygen species and proteases. Mechanisms involved in COPD include telomere shortening, cellular senescence, activation of PI3 kinase-mTOR signaling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence, and a low-grade chronic inflammation. Therefore, the Senolytic Small Molecule(s) is suitable for treatment of premature senescence involved in Pulmonary Diseases or Disorders by removing the senescent cells in controlled and safe manner.
[000190] In some embodiments, Pulmonary Disease and Disorders treated by Senolytic Therapy described herein include Idiopathic Pulmonary Fibrosis (IPF). IPF is the most common and severe idiopathic interstitial pneumonia. In familial interstitial pneumonia, the telomerase complex is affected by the genetic lesions present eventually leading to telomere shortening in both leukocytes and pulmonary tissue which is also observed in sporadic IPF. Pathology of IPF points out to a mechanism with the involvement of cellular senescence in disease progression.
[000191] In both COPD and IPF, premature cellular senescence likely affects distinct progenitor’s cells (mesenchymal stem cells in COPD, alveolar epithelial precursors in IPF), leading to stem cell exhaustion.
[000192] In one embodiment, Senolytic Therapy provided herein is suitable for treating or preventing (i.e., reducing the likelihood of occurrence of pulmonary disease or disorder by killing senescent cells associated with the disease or disorder) particularly the senescence of pulmonary artery-smooth muscle cells in a subject who has the disease or disorder.
[000193] In some embodiments, the Senolytic Therapy is effective in the treatment of lung emphysema.
[000194] During such a Senolytic Therapy of Pulmonary Diseases and disorders including lung emphysema, the effectiveness of the therapy can be established by taking certain measurements which include but not limited to, inter alia, breathlessness, chest size, lung volume, decreased breath sounds through the stethoscope, fingertip shape, style of breathing, hypoxemia, hypercaria, cyanosis, malnutrition lung volume, lung ejection capacity, dead volume in the lungs, airflow changes after bronchodilator medication, chest X-RAY and CT scan of the chest and red blood cell counts.
[000195] In some embodiments, the Senolytic Therapy is effective in the treatment of COPD. During the senolytic therapy of the COPD disease parameters which are measured include, inter alia, spirometry and lung functional tests as described for lung emphysema, including breathlessness, chest size, decreased breath sounds through the stethoscope, fingertip shape, style of breathing, hypoxemia, hypercaria, cyanosis, malnutrition, lung volume, lung ejection capacity, dead volume in the lungs, airflow changes after bronchodilator medication, chest X- RAY and CT scan of the chest and red blood cell counts.
[000196] In some embodiments the methods of use of Senolytic Agents for treatment of Pulmonary Disease and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of Pulmonary Disease and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of Pulmonary Diseases and disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000197] During the therapy, a subj ecf s progress can be monitored appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subject at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000198] Method Use of the Senolytic Small Molecule(s) for treatment of Neurological Diseases and Disorders
[000199] Chronic inflammation is a major contributor to a range of neurodegeneration the progressive dysfunction and loss of neurons in the central nervous system whereas neurodegeneration is the major cause of cognitive and motor dysfunction including in Alzheimer's and Parkinson's diseases, neurotropic infections, traumatic brain and spinal cord injury, stroke, neoplastic disorders, prion diseases, multiple sclerosis and amyotrophic lateral sclerosis. Therefore, the Senolytic Small Molecule(s) is suitable to slow down the progression of neurodegeneration by reducing the chronic inflammation in the body substantially by stopping SASP. [000200] In some embodiments the method of use of Senolytic Agents for senescence- associated neurological diseases or disorders, including but not limited to Parkinson’s disease, Alzheimer's disease (and other dementias), motor neuron dysfunction (MND), mild cognitive impairment (MCI), Huntington's disease, and diseases and disorders of the eyes, such as age- related macular degeneration, comprises Treatment Regimes as disclosed in the subject matter of this patent.
[000201] In some embodiments the methods of use of Senolytic Agents for treatment of senescence-associated neurological diseases or disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of senescence-associated neurological diseases or disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of senescence-associated neurological diseases or disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000202] In some embodiments the effectiveness of Senolytic Therapy on senescence- associated neurological diseases or disorders, as administered in Treatment Regimes described herein, can be established by monitoring of a subject by a person skilled in the medical and clinical arts. In some embodiments one or any combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods described herein are used for monitoring the effectiveness of said treatment. The effects of administering one or more Senolytic Small Molecules can be further analyzed using techniques known in the art, such as comparing symptoms of subjects suffering from or at risk of Alzheimer's disease that have received the treatment with those of subjects without such a treatment or with placebo treatment.
[000203] In some embodiments, the Senolytic Agent(s) based Senolytic Therapy is effective in the effective in the treatment of Alzheimer's disease. During the senolytic therapy the Alzheimer’s disease parameters which are measured include, inter alia, changes in ability to carry out daily activities, and changes in behavior and personality, tests of memory, problem solving, attention, counting, and language, blood and urine tests, brain scans, such as computed tomography (CT), magnetic resonance imaging (MRI), or positron emission tomography (PET) and/or biomarker analysis.
[000204] In some embodiments, the Senolytic Therapy is effective in the treatment of Parkinson's disease. During the senolytic therapy the Parkinson’s disease parameters which are measured include, inter alia, analysis for tremors, limb or neck stiffness, general fitness and balance and/or locomotor function.
[000205] In some embodiments, the Senolytic Agent(s) based Senolytic Therapy is effective in the treatment of depression. Depression parameters that are to be measured are, inter alia, physical examination, sadness or depressed mood most of the day, major changes in weight, insomnia or excessive sleep, fatigue or loss of energy most of the day, feelings of hopelessness or worthlessness or excessive guilt, problems with concentration or decision making, recurring thoughts of death or suicide.
[000206] Method Use of the Senolytic Small Molecule(s) for treatment of Ophthalmic Diseases and Disorders
[000207] Senescence-associated diseases or disorders treatable by administering the Senolytic Small Molecule as described in Treatment Regimes herein include ophthalmic diseases or disorders. Such ophthalmic diseases and disorders include but not limited to age-related macular degeneration, cataracts, glaucoma, vision loss, presbyopia. In some embodiments, Ophthalmic Diseases or Disorders involve age related macular degeneration (AMD) resulting in irreversible blindness which is associated with the degradation of retinal pigment epithelium (RPE) cells, photoreceptors, and choriocapillaris. Oxidative stress, inflammation (IL-17 involvement) and some genetic factors are known to be involved in AMD pathogenesis. Oxidative stress can induce DNA damage response (DDR), autophagy, and cell senescence.
[000208] Therefore, in some embodiments, the Senolytic Small Molecule(s) is suitable for treatment of premature senescence involved Ophthalmic Diseases and Disorders by removing the senescent cells in controlled and safe manner. [000209] In some embodiments, Treatment Regimes provided herein for treating or preventing (i.e., reducing the likelihood of occurrence of; delaying the onset or development of, or inhibiting, retarding, slowing, or impeding progression or severity of) an ophthalmic disease, disorder, or condition (e.g., presbyopia, cataracts, macular degeneration); for selectively killing senescent cells in an eye of a subject, and/or inducing collagen production in the eye of a subject in need thereof by administering at least one Senolytic Small Molecule which may be combined with at least one therapeutically acceptable excipient to form a composition comprising the Senolytic Small Molecule(s)) directly to an eye.
[000210] In some embodiments the methods of use of Senolytic Agents for treatment of ophthalmic diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of said Treatment Regimes of Senolytic Agents for treatment of ophthalmic diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of ophthalmic diseases certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000211] Method Use of the Senolytic Small Molecule(s) for treatment of Metabolic Diseases or Disorders
[000212] Senescence-associated diseases or disorders treatable or preventable by administering the Senolytic Therapy include metabolic diseases or disorders. Said senescence- associated diseases and disorders include diabetes, metabolic syndrome, diabetic ulcers, and obesity.
[000213] Chronic inflammation affects the whole body and is linked to diseases like type 2 diabetes. Low-grade chronic inflammation appears to change the way glucose is absorbed by cells. Anakinra, a biologic anti-inflammatory drug, has been found to improve some diabetes symptoms by blocking the cytokine protein IL-1, as stated above the key instigator of the immune and inflammatory response. Therefore, the Senolytic Therapy is suitable to slow down the progression of the diabetes by reducing the chronic inflammation in the body substantially by stopping SASP.
[000214] Diagnosis of type 2 diabetes is based on symptoms (e.g., increased thirst and frequent urination, increased hunger, weight loss, fatigue, blurred vision, slow-healing sores or frequent infections, and/or areas of darkened skin), medical history, and/or by taking measurements which include, inter alia, basal blood glucose levels, average blood glucose levels over a period of time (2-3 months; AIC test), fasting plasma glucose, oral glucose tolerance test, plasma glucose test N.B. physical examination of a subject.
[000215] The effectiveness of the Senolytic Therapy can readily be determined by a person skilled in the medical and clinical arts. One or any combination of diagnostic methods, including physical examination, assessment and monitoring of clinical symptoms, and performance of analytical tests and methods, such as those described herein, may be used for monitoring the health status of the subject. In some embodiments a subject who is receiving Senolytic Therapy for treatment or prophylaxis of diabetes can be monitored, for example, by assaying glucose and insulin tolerance, energy expenditure, body composition, fat tissue, skeletal muscle, and liver inflammation, and/or lipotoxicity (muscle and liver lipid by imaging in vivo and muscle, liver, bone marrow, and pancreatic b-cell lipid accumulation and inflammation by histology). Other characteristic features or phenotypes of type 2 diabetes are known and can be assayed as described herein and by using other methods and techniques known and routinely practiced in the art.
[000216] Obesity and obesity-related disorders are used to refer to conditions of subjects who have a body mass that is measurably greater than ideal for their height and frame. Body Mass Index (BMI) is a measurement tool used to determine excess body weight, and is calculated from the height and weight of a subject. A human is considered overweight when the person has a BMI of 25-29; a person is considered obese when the person has a BMI of 30-39, and a person is considered severely obese when the person has a BMI of 40. Accordingly, the terms obesity and obesity-related refer to human subjects with body mass index values of greater than 30, greater than 35, or greater than 40. A category of obesity not captured by BMI is called “abdominal obesity” in the art, which relates to the extra fat found around a subject's middle, which is an important factor in health, even independent of BMI. The simplest and most often used measure of abdominal obesity is waist size. Generally abdominal obesity in women is defined as a waist size 35 inches or higher, and in men as a waist size of 40 inches or higher. More complex methods for determining obesity require specialized equipment, such as magnetic resonance imaging or dual energy X-ray absorptiometry machines.
[000217] A condition or disorder associated with diabetes and senescence is a diabetic ulcer (i.e., diabetic wound). An ulcer is a breakdown in the skin, which may extend to involve the subcutaneous tissue or even muscle or bone. These lesions occur, particularly, on the lower extremities. Patients with diabetic venous ulcer exhibit elevated presence of cellular senescence at sites of chronic wounds. Chronic inflammation is also observed at sites of chronic wounds, such as diabetic ulcers suggesting that the proinflammatory cytokine phenotype of senescent cells has a role in the pathology.
[000218] Subjects who have type 2 diabetes or who are at risk of developing type 2 diabetes may have metabolic syndrome. Metabolic syndrome in humans is typically associated with obesity and characterized by one or more of cardiovascular disease, liver steatosis, hyperlipidemia, diabetes, and insulin resistance. A subject with metabolic syndrome may present with a cluster of metabolic disorders or abnormalities which may include, for example, one or more of hypertension, type-2 diabetes, hyperlipidemia, dyslipidemia (e.g., hypertriglyceridemia, hypercholesterolemia), insulin resistance, liver steatosis (steatohepatitis), hypertension, atherosclerosis, and other metabolic disorders.
[000219] In some embodiments, the diabetes type II disease is effectively treatable or preventable by administering the Senolytic Therapy. During the Senolytic Therapy of diabetes type II disease, the efficacy of the Treatment Regimes is established by taking measurements which-include, inter alia, basal blood glucose levels, average blood glucose levels over a period of time (2-3 months; AIC test), fasting plasma glucose, oral glucose tolerance test, plasma glucose test N.B.
[000220] In some embodiments, the obesity disorder is effectively treatable or preventable by administering the Senolytic Therapy. During the Senolytic Therapy the obesity parameters which are measured include alia body weight, Body-Mass-Index (BMI), waist circumference, waist-to-hip ratio, skinfold thicknesses, and bioelectrical impedance, magnetic resonance imaging and/or dual energy X-ray absorptiometry.
[000221] In some embodiments, the metabolic syndrome is treatable or preventable effectively by administering the Senolytic Therapy. During the Senolytic Therapy the metabolic syndrome disease parameters which are measured include, inter alia, measurements for obesity (see above, e.g. waist circumference), blood levels of triglycerides, HDL cholesterol, blood pressure, fasting glucose.
[000222] The hepatic insufficiency is treatable or preventable effectively by administering the Senolytic Therapy. During the senolytic therapy, the hepatic insufficiency disease parameters which are measured include, inter alia, blood AST and ALT values.
[000223] In some embodiments, cirrhosis disease is treatable or preventable effectively by administering the Senolytic Therapy. During the senolytic therapy, the cirrhosis disease parameters which are measured include, inter alia, measurements of blood-clotting factors and international normalized ratio for blood clotting, liver stiffness by magnetic resonance elastography, liver imaging by CT and/or MRI, physical examination, blood testing for bilirubin and creatinine, and/or liver biopsy analysis for liver damage.
[000224] In some embodiments the methods of use of Senolytic Agents for treatment or prevention of metabolic diseases and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment or prevention of metabolic diseases and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy treatment or prevention of metabolic diseases and disorders certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000225] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000226] Method Use of the Senolytic Small Molecule(s) for treatment of Renal Dysfunction:
[000227] Nephrological pathologies, such as glomerular disease, arise in the elderly. Glomerulonephritis is characterized by inflammation of the kidney and by the expression of two proteins, ILla and I L I b . ILla and IL l b are considered master regulators of SASP. Glomerular disease is associated with elevated presence of senescent cells, especially in fibrotic kidneys. Therefore, the Senolytic Small Molecule(s) is suitable to renal dysfunction by substantially stopping SASP by killing the senescent cells.
[000228] In some embodiments, renal insufficiency is effectively treatable or preventable by administering the Senolytic Therapy. During the Senolytic Therapy, the renal insufficiency disease parameters which are measured include, inter alia, blood pressure, heart/lung sound analysis, nervous system exam, urinalysis for protein content, analysis for creatinine clearance and level of Blood Urea Nitrogen, CT, MRI and/or ultrasound of abdomen and kidneys, kidney biopsy for damage analysis.
[000229] Glomerulosclerosis is another pathology associated with renal aging supported by the accumulation of senescent cells as indicated by an increase in the levels of senescence markers such as pl6 and SA-P-Gal by aging.
[000230] In some embodiments, renal dysfunction caused by nephrological pathologies, such as glomerulosclerosis is effectively treatable or preventable by administering the Senolytic Therapy. During the Senolytic Therapy, the glomerulosclerosis disease parameters which are measured include, inter alia, swellings in limbs, weight gains, changes in urine due to proteinuria, distortion or compression of the small capillaries in the glomerulus that filter blood in a biopsy and plasma Urea or protein concentration, blood pressure, glomerular filtration rate, and/or kidney ultrasound.
[000231] In some embodiments the methods of use of Senolytic Agents for treatment of renal dysfunction and disorders comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of renal dysfunction and disorders include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of renal dysfunction and disorders certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000232] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline.
[000233] Method Use of the Senolytic Small Molecule(s) as an adjuvant agent in a cancer therapy and for preventing Metastasis
[000234] Stress-induced premature senescence (SIPS) occurs rapidly in response to various stresses such as chemotherapeutic small molecule and ionizing radiation. Both stresses cause substantial collateral macromolecular damage to non-neoplastic cells and responsible for the early aging phenotypes frequently observed in cancer survivors. Contrary to chronic senescence resulting from normal aging mechanisms and declining macromolecular repair mechanisms, therapy-induced senescence results from abrupt exogenous stresses placed on tissues during cancer therapy. The Senolytic Therapy may be administered to the subjects who may also have cancer, as an adjuvant for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis using the methods as described herein. Metastasis of a cancer occurs when the cancer cells (e.g., tumor cells) spread beyond the anatomical site of origin and initial colonization to other areas throughout the body of the subject. In some embodiments, Senolytic Therapy is selected the primary therapy for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis. In certain embodiments, Senolytic Therapy as an adjuvant therapy includes, but is not limited to, chemotherapy, radiation therapy, hormone therapy, targeted therapy, or biological therapy or other novel therapies that will be recognized by those skilled in the art.
[000235] In some embodiments, Senolytic Therapy as an adjuvant for cancer therapy is administered in combination with chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s) used in primary therapy or following a primary therapy for prevention, cure, inhibition or retarding the progression of cancer and/or metastasis
[000236] In some embodiments the methods of use of Senolytic Agents for inhibition or retarding the progression of metastasis in a subject who has a cancer comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for inhibition or retarding the progression of metastasis include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for inhibiting or retarding metastasis of cancer certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders. Such the Senolytic Small Molecule when administered in a Therapeutically Effective Dose to a subject who has a cancer according to the methods described herein may inhibit tumor proliferation.
[000237] In some embodiments the methods of use of Senolytic Agents for targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells in a subject who has a cancer comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for targeting circulating tumor cells (CTCs) include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. Some examples of Treatment Regimes of Senolytic Therapy for targeting cancer stem cells include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. Some examples of Treatment Regimes of Senolytic Therapy for targeting dormant cells include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells of cancer certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders. Such the Senolytic Small Molecule when administered in a Therapeutically Effective Dose to a subject who has a cancer according to the methods described herein may inhibit tumor proliferation.
[000238] The methods described herein are also applicable for inhibiting, retarding or slowing progression of metastatic cancer, or targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells of any one of the types of tumors described in the medical art. In some embodiments multiple Senolytic Therapies are further customized and applied contemporaneously to the same patient for inhibiting, retarding or slowing progression of cancer tumor and/or metastasis and/or for targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells. As a non-limiting example, a Senolytic Agent is injected directly into a cancer tumor for inhibiting, retarding or slowing progression of the tumor and/or targeting circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells and contemporaneously same or a separate Senolytic Agent is administered as a Senolytic Therapy via intravenous or intraperitoneal delivery method for inhibiting, retarding or slowing progression of metastasis and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells.
[000239] In some embodiments, the cancer type is but not limited to metastatic melanoma, resistant breast cancer, radiotherapy resistant glioblastoma, colorectal cancer, or thyroid cancer.
[000240] In some embodiments, administration of Senolytic Agents as described in Treatment Regimes, are effective in the treatment of metastatic melanoma or as an adjuvant drug. During the senolytic therapy, the metastatic melanoma disease parameters which are measured include, inter alia, a reduction in tumor size and/or metastasization by applying Treatment Regimes as disclosed in the subject matter of this patent i.e. by administering the Senolytic Agents’ Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1.
[000241] In some embodiments the methods of use of Senolytic Agents for the treatment of resistant breast cancer as adjuvants, comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for treatment of resistant breast cancer as adjuvants, include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of resistant breast cancer as adjuvants, certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000242] In some embodiments, administration of Senolytic Therapy is effective in the treatment of resistant breast cancer or as an adjuvant drug. During the senolytic therapy, the resistant breast cancer disease parameters which are measured include a reduction in tumor size and/or metastasization.
[000243] In some embodiments, Senolytic Therapy is the primary therapy for treatment of the resistant breast cancer which involves one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s).
[000244] In certain embodiments, the primary therapy agent used for the treatment of the resistant breast cancer is a chemotherapeutic agent(s). Said chemotherapeutic agent(s) include but are not limited to, Tamoxifen, Femara, Herceptin, Letrozole, Taxol, Soltamox, Epirubicin, Trastuzumab, Leuprolide, Paclitaxel, Ellence, Pharmorubicin PFS, Neratinib, Nerlynx, Ogivri and their derivatives.
[000245] In some embodiments, the Senolytic Therapy is directed as an adjuvant therapy for the treatment of the resistant breast cancer, during or following Primary Therapy wherein Senolytic Agent is used in combination with at least one Chemotherapeutic Agent.
[000246] In some embodiments, Senolytic Therapy, is effective in treatment of a resistant glioblastoma or as an adjuvant drug. During the senolytic therapy, the resistant glioblastoma disease parameters which are measured include, inter alia, a reduction in tumor size and/or metastasization.
[000247] In some embodiments the methods of use of Senolytic Agents for the treatment of resistant glioblastoma as adjuvants comprises Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for the treatment of resistant glioblastoma as adjuvants include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for the treatment of resistant glioblastoma as adjuvants, certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000248] In some embodiments, Senolytic Therapy is used as the primary therapy used for treatment of the resistant glioblastoma and involves one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s). Said chemotherapeutic agent(s) include but are not limited to, Temodar, Avastin, Temozolomide, Matulane, Bevacizumab, BiCNU, Gliadel, Carmustine, Hydroxyurea, Procarbazine, Mvasi and their derivatives.
[000249] In some embodiments, the Senolytic Agents are used in adjuvant therapy for the treatment of the resistant glioblastoma, in combination with or following primary therapy with at least one chemotherapeutic agent. Said chemotherapeutic agent(s) could be used alone or in combination with the Senolytic Agents. In some embodiments, administration of Senolytic Therapy is effective in the treatment of colorectal cancer or as an adjuvant drug. During the senolytic therapy, the colorectal cancer disease parameters which are measured include a reduction in tumor size and/or metastasization.
[000250] In some embodiments the methods of use of Senolytic Agents for the treatment of colorectal cancer as adjuvants comprises Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for the treatment of colorectal cancer, wherein Senolytic Agents are used as adjuvants, include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for the treatment of colorectal cancer, wherein Senolytic Agents are used as adjuvants, certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders. [000251] In some embodiments, the Senolytic Therapy is used as the primary therapy used for treatment of colorectal cancer which involves one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s). Said chemotherapeutic agent(s) include but are not limited to, Xeloda, Oxaliplatin, Avastin, Fluorouracil, Leucovorin, Capecitabine, Irinotecan, Stivarga, Bevacizumab, Erbitux, Camptosar, Cetuximab, Eloxatin, Vectibix, Zaltrap, Betaseron, Fusilev, Lonsurf, Methotrexate, Panitumumab, Wellcovorin, Regorafenib, Mvasi, Keytruda, Opdivo, Cyramza, Interferon beta- lb, Levoleucovorin, Nivolumab, Ramucirumab, Tipiracil/Trifluridine, Ziv-aflibercept, Pembrolizumab, Ipilimumab, Khapzory, Yervoy and their derivatives.
[000252] In some embodiments, the Senolytic Agents are used in adjuvant therapy for the treatment of colorectal cancer, in combination with or following primary therapy with at least one chemotherapeutic agent. Said chemotherapeutic agent(s) could be used alone or in combination with the Senolytic Agents. In some embodiments, administration of Senolytic Therapy is effective in the treatment of a thyroid cancer or as an adjuvant drug. During the senolytic therapy, the thyroid cancer disease parameters which are measured include, inter alia, a reduction in tumor size and/or metastasization.
[000253] In some embodiments, the methods of use of Senolytic Agents for the treatment of thyroid cancer as adjuvants, comprises Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for the treatment of thyroid cancer as adjuvants, include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for the treatment of thyroid cancer as adjuvants, certain parameters Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000254] In some embodiments, the primary therapy used for treatment of thyroid cancer could involve one or more chemotherapeutic, immunotherapeutic, radiotherapeutic or biological agent(s). Said chemotherapeutic agents include but are not limited to, Armour Thyroid, Nexavar, Nature-Throid, Thyrogen, Caprelsa, Adriamycin, Cometriq, Thyroid desiccated, Sodium iodide-i-131, Sorafenib, Lenvima, Doxorubicin, Lenvatinib, Lodotope, Cabozantinib, Westhroid, Thyrotropin alpha, Vandetanib, Hicon, NP Thyroid, Dabrafenib, Tafmlar, WP Thyroid, i3odine Max, Mekinist, Trametinib and their derivatives.
[000255] In some embodiments, the Senolytic Agents are used in adjuvant therapy for the treatment of thyroid cancer, in combination with or following primary therapy with at least one chemotherapeutic agent. Said chemotherapeutic agent(s) could be used alone or in combination with the Senolytic Agents.
[000256] In some embodiments, the method of use of Senolytic Agents as treatment or adjuvants for the inhibition of metastasis of a cancer comprises Treatment Regimes as disclosed in the subject matter of this patent. Some examples of said Treatment Regimes include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments the Treatment Regimes for Therapeutically Effective Dose and/or the duration of cancer treatment stated regimes could be altered depending on the subject’s state of health and/or the subject’s response to the treatment including but not limited to alteration of Therapeutically Effective Dose and/or the duration of treatment, or combinations of Senolytic Agents and delivery methods or combinations thereof.
[000257] In some embodiments, during the Senolytic Therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subject at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline
[000258] In other embodiments, during the Senolytic Therapy, a subject’s progress could additionally be followed by detecting and/or monitoring metastatic cells and/or cancer cells present in the subject before, during or after the therapy using the methods as recognized by those skilled in the art.
[000259] Method of Use of the Senolytic Small Molecule(s) for treatment of Progeroid Syndromes
[000260] Progeroid syndromes (PSs) are a group of fatal, severe and rare genetic disorders which mimic premature aging while exhibiting various clinical features and phenotypes. PSs mimic many of the characteristics of human ageing such as hair loss, short stature, skin tightness, cardiovascular diseases and osteoporosis. Therefore, the Senolytic Small Molecules may also be useful for treating or alleviation of the effects of Progeroid Syndromes that occur as a result of premature aging process induced by congenital genetic mutations in individuals. Although all progeroid syndromes are characterized by similar clinical features, their underlying mechanisms can vary depending on the mutated gene and the pathway that is consequently altered. As a result of genomic instabilities due to the mutated genes, premature senescence emerges as a key factor underlying these conditions.
[000261] In some embodiments, these syndromes include clinically and genetically heterogeneous diseases such as ataxia-telangiectasia, Bloom syndrome, Cockayne syndrome, Fanconi anaemia, Hutchinson-Gilford Progeria syndrome, Rothmund-Thomson syndrome, trichothiodystrophy, xeroderma pigmentosum, and Werner syndrome (aka adult progeria).
[000262] In some embodiments, Progeroid Syndromes include Hutchinson-Gilford Progeria Syndrome (HGPS) which is a rare , fatal and genetic condition of childhood, characterized by growth reduction, failure to thrive, a typical facial appearance (prominent forehead, protuberant eyes, thin nose with a beaked tip, thin lips, micrognathia and protruding ears) and distinct dermatologic features (generalized alopecia, aged-looking skin, sclerotic and dimpled skin over the abdomen and extremities, prominent cutaneous vasculature, dyspigmentation, nail hypoplasia and loss of subcutaneous fat). Individuals with HGPS exhibit atherosclerosis, lipodystrophy, heart infarction and death during puberty.
[000263] During the senolytic treatment HGPS disease parameters are measured by, inter alia, features of accelerated aging, hair loss (alopecia), aged-looking skin, joint abnormalities, and a loss of fat under the skin.
[000264] In some embodiments, Progeroid Syndromes include Trichothiodystrophy characterized by brittle hair causing hair loss, neurological defects, bone abnormalities and fitness decline.
[000265] In other embodiments, PSs include Werner Syndrome characterized by the dramatic, rapid appearance of features associated with normal aging in affected individuals. Affected individuals usually develop accompanying disorders of aging early in life, such as cataracts, skin ulcers, type 2 diabetes, diminished fertility, atherosclerosis, osteoporosis, and some types of cancer. [000266] In some embodiments, premature aging-associated decline and symptoms may be treated or prevented (i.e., the likelihood of occurrence of is reduced) by administering the Senolytic Therapy
[000267] In some embodiments, the methods of use of Senolytic Agents for treatment of progeroid syndromes comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for the treatment of progeroid syndromes include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for treatment of progeroid syndromes certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000268] The effectiveness of a method of treatment described herein may be manifested by reducing the number of symptoms of a premature aging disease or progeroid trait associated with a senescence-inducing stimulus, decreasing the severity of one or more symptoms, or delaying the progression of a premature aging disease or progeroid trait associated with a senescence-inducing stimulus. In some embodiments, preventing a premature aging disease or progeroid trait associated with a senescence-inducing stimulus refers to preventing (i.e., reducing the likelihood of occurrence) or delaying onset of a premature aging disease or progeroid trait associated with a senescence-inducing stimulus, or reoccurrence of one or more premature aging disease or progeroid trait associated with a senescence-inducing stimulus.
[000269] Method of Use of the Senolytic Small Molecule(s) to Reduce the Side Effects of Chemotherapy and Radiotherapy
[000270] Tumor proliferation may be determined by tumor size, which can be measured in various ways familiar to a person skilled in the art, such as by PET scanning, MRI, CAT scan, biopsy, for example. The effect of the therapeutic agent on tumor proliferation may also be evaluated by examining differentiation of the tumor cells. It has been demonstrated that a Senolytic Agent lowers the threshold for senescent cells to enter apoptosis after DNA damage. In some embodiments the Senolytic Agent is used against chemotoxicity or radiation damage by removing the senescent cells in controlled and safe manner.
[000271] Because cells may be induced to senesce by cancer therapies, such as radiation and certain chemotherapy small molecule (e.g., doxorubicin; paclitaxel; gemcitabine; pomalidomide; lenalidomide), the Senolytic Small Molecule(s) described herein may be administered after the chemotherapy or radiotherapy to kill (or facilitate killing) of these senescent cells.
[000272] As discussed herein and understood in the art, establishment of senescence, such as shown by the presence of a senescence-associated secretory phenotype (SASP), occurs over several days; therefore, administering the Senolytic Small Molecule to kill senescent cells and/or circulating tumor cells (CTCs) and/or dormant cells and/or cancer stem cells, and thereby reduce the likelihood of occurrence or reduce the extent of metastasis, is initiated when senescence and/or intravasation and/or extravasation has been established. As discussed herein, the following treatment courses for administration of the Senolytic Small Molecule may be used in methods described herein for treating or preventing (i.e., reducing the likelihood of occurrence, or reducing the severity) a chemotherapy or radiotherapy side effect. Removal or destruction of senescent cells may ameliorate acute toxicity, including acute toxicity comprising energy imbalance, of a chemotherapy or radiotherapy. Acute toxic side effects include but are not limited to gastrointestinal toxicity (e.g., nausea, vomiting, constipation, anorexia, diarrhea), peripheral neuropathy, fatigue, malaise, low physical activity, hematological toxicity (e.g., anemia), hepatotoxicity, alopecia (hair loss), pain, infection, mucositis, fluid retention, dermatological toxicity (e.g., rashes, dermatitis, hyperpigmentation, urticaria, photosensitivity, nail changes), mouth (e.g., oral mucositis), gum or throat problems, or any toxic side effect caused by a chemotherapy or radiotherapy.
[000273] Accordingly, in some embodiments, methods are provided herein for ameliorating (reducing, inhibiting, or preventing occurrence (i.e., reducing the likelihood of occurrence)) acute toxicity or reducing severity of a toxic side effect (i.e., deleterious side effect) of a chemotherapy or radiotherapy or both in a subj ect who receives the therapy, wherein the method comprises administering to the subject an agent that selectively kills, removes, or destroys or facilitates selective destruction of senescent cells. Administration of the Senolytic Small Molecule for treating or reducing the likelihood of occurrence, or reducing the severity of a chemotherapy or radiotherapy side effect may be accomplished by the same treatment courses described above for treatment/prevention of metastasis. As described for treating or preventing (i.e., reducing the likelihood of occurrence of) metastasis, the Senolytic Small Molecule is administered in a Therapeutically Effective Dose during the off-chemotherapy or off- radiotherapy time interval or after the chemotherapy or radiotherapy treatment regimen has been completed.
[000274] In some embodiments the methods of use of Senolytic Agents aim for reduction of the side effects of chemotherapy and radiotherapy comprise Senolytic Therapy as disclosed in the subject matter of this patent. Some examples of Treatment Regimes of Senolytic Therapy for reduction of the side effects of chemotherapy and radiotherapy include administering Therapeutically Effective Dose via Impulse Regime, Sustained Regime, and Gentle Regime or via any of the regimes disclosed in Schedule 1. In various embodiments of the Senolytic Therapy for reduction of the side effects of chemotherapy and radiotherapy certain parameters of the Therapy Regimes including, but not limited to, Therapeutically Effective Dose and/or combinations of Senolytic Agents, and/or the duration and number of treatment cycles and/or delivery methods or combinations thereof could be altered depending on the subject’s state of health and/or the subject’s response to the treatment. Said alterations of Therapy Regime are subject to the decision of the doctor overseeing the therapy particularly when the subject has other diseases or disorders.
[000275] During the therapy, a subject’s progress can be monitored by appropriate measurements including those presented herein or by detecting the senescent cell population on a biopsy sample taken from the subj ect at the beginning and throughout the period of the therapy and thus monitoring the senescent cell population decline. The number of cycles of a chemotherapy or radiotherapy or the total length of time of a chemotherapy or radiotherapy dose regime can vary depending on the subject's response to the cancer therapy. In some embodiments the senolytic therapy plan timeframe will be adjusted and aligned with said chemotherapy or radiotherapy treatment by a person skilled in the oncology art.
[000276] Compositions and methods of delivery for the Senolytic Small Molecule
[000277] In some embodiments, compositions comprising a Senolytic Small Molecule can be formulated in a manner appropriate for the delivery method by using techniques routinely practiced in the art. The composition may be in the form of a solid (e.g., tablet, capsule), semi solid (e.g., gel), liquid, or gas (aerosol). In some embodiments, the Senolytic Small Molecule (or a composition comprising same) is administered in a Therapeutically Effective Dose as a bolus infusion. In some embodiments when the Senolytic Small Molecule is delivered in a Therapeutically Effective Dose by infusion wherein the Senolytic Small Molecule is delivered to an organ or tissue comprising senescent cells or cancer tumors as an adjuvant drug to a chemotherapy to be killed in accordance with techniques routinely performed by a person skilled in the medical art.
[000278] Pharmaceutically-acceptable excipients are well-known in the pharmaceutical arts. Examples of pharmaceutically-acceptable excipients include sterile saline and phosphate buffered saline at physiological pH. Preservatives, stabilizers, dyes, buffers, and the like may be provided in the composition. In some embodiments, the type of excipient is selected based on the mode of administration, as well as the chemical composition of the active ingredient(s). In other embodiments, the Senolytic Small Molecule is formulated as a lyophilizate. A composition based on the disclosed subject matter may be lyophilized or otherwise formulated as a lyophilized product using one or more appropriate excipient solutions for solubilizing and/or diluting the agent(s) of the composition upon administration. In some embodiments, the Senolytic Small Molecule is encapsulated within liposomes using technology known and practiced in the art.
[000279] In some embodiments, compositions comprising the Senolytic Small Molecule is formulated and packaged for delivery for any appropriate manner of administration described in the disclosed subject matter and in the art.
[000280] A composition comprising the Senolytic Small Molecule(s) may be delivered to a subject in need thereof by any one of several routes known to a person skilled in the art. By way of non-limiting example, the composition may be delivered orally, intravenously, intraperitoneally, by infusion (e.g., a bolus infusion), subcutaneously, enteral, rectal, intranasal, by inhalation, buccal, sublingual, intramuscular, transdermal, intradermal, topically, intraocularly, vaginally, rectally, intrathecally, intracranially, or by some combination thereof. In some embodiments, administration of a dose, as described above, is via intravenous, intraperitoneal, directly into the target tissue or organ, or subcutaneous route. In some embodiments, a delivery method includes drug-coated or permeated stents for which the drug is the Senolytic Small Molecule. Formulations suitable for such delivery methods are described in greater detail herein. [000281] In some embodiments, the Senolytic Small Molecule (which may be combined with at least one therapeutically-acceptable excipient to form a composition comprising the Senolytic Small Molecule(s)) is administered in a Therapeutically Effective Dose directly to the target tissue or tumor or organ comprising senescent cells that contribute to manifestation of the disease or disorder. In some embodiments when treating osteoarthritis, at least one Senolytic Small Molecule is administered in a Therapeutically Effective Dose directly to an osteoarthritic joint (i.e., intra-articularly) of a subject in need thereof. In some embodiments, the Senolytic Small Molecule(s) may be administered in a Therapeutically Effective Dose to the joint via topical, transdermal, intradermal, or subcutaneous route. In some embodiments, methods are provided herein for treating a cardiovascular disease or disorder associated with arteriosclerosis, such as atherosclerosis by administering directly into an artery. In some embodiments, the Senolytic Small Molecule (which may be combined with at least one pharmaceutically- acceptable excipient to form a composition comprising the Senolytic Small Molecule(s)) for treating a senescence-associated pulmonary disease or disorder may be administered in a Therapeutically Effective Dose by inhalation, intranasally, by intubation, or intracheally, for example, to provide the Senolytic Small Molecule more directly to the affected pulmonary tissue. By way of another non-limiting example, the Senolytic Small Molecule (or composition comprising the Senolytic Small Molecule) may be delivered directly to the eye either by injection (e.g., intraocular or intravitreal) or by conjunctival application underneath an eyelid of a cream, ointment, gel, or eye drops. In some embodiments, the Senolytic Small Molecule or a composition comprising the Senolytic Small Molecule may be formulated as a timed release (also called sustained release, controlled release) composition or may be administered in a Therapeutically Effective Dose as a bolus infusion.
[000282] A composition comprising the Senolytic Small Molecule(s) (e.g., for oral administration or for injection, infusion, subcutaneous delivery, intramuscular delivery, intraperitoneal delivery or other method) may be in the form of a liquid. A liquid composition comprising the Senolytic Small Molecule(s) may include, for example, one or more of the following: a sterile diluent such as water, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils that may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents; antioxidants; chelating agents; buffers and agents for the adjustment of tonicity such as sodium chloride or dextrose. A parenteral composition can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. The use of physiological saline is preferred, and an injectable composition comprising the Senolytic Small Molecule(s) is preferably sterile. In another embodiment, for treatment of an ophthalmological condition or disease, a liquid composition comprising the Senolytic Small Molecule(s) may be applied to the eye in the form of eye drops. A liquid composition comprising the Senolytic Small Molecule(s) may be delivered orally.
[000283] For oral formulations, at least one of the Senolytic Small Molecules described herein can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, and if desired, with diluents, buffering agents, moistening agents, preservatives, coloring agents, and flavoring agents. The compounds may be formulated with a buffering agent to provide for protection of the compound from low pH of the gastric environment and/or an enteric coating. The Senolytic Small Molecule included in a composition comprising the Senolytic Small Molecule(s) may be formulated for oral delivery with a flavoring agent, e.g., in a liquid, solid or semi-solid formulation and/or with an enteric coating.
[000284] A composition comprising any one of the Senolytic Small Molecules described herein may be formulated for sustained or slow release (also called timed release or controlled release). Such compositions comprising the Senolytic Small Molecule may generally be prepared using well known technology and administered in a Therapeutically Effective Dose by, for example, oral, rectal, intradermal, or subcutaneous implantation, or by implantation at the desired target site. Sustained-release formulations may contain the compound dispersed in a carrier matrix and/or contained within a reservoir surrounded by a rate controlling membrane. Excipients for use within such formulations are biocompatible, and may also be biodegradable; preferably the formulation provides a relatively constant level of active component release. The amount of active agent contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release, and the nature of the condition, disease or disorder to be treated or prevented.
[000285] In some embodiments, the compositions comprising the Senolytic Small Molecule are formulated for transdermal, intradermal, or topical administration. Said compositions can be administered in a Therapeutically Effective Dose using a syringe, bandage, transdermal patch, insert, or syringe-like applicator, as a powder/talc or other solid, liquid, spray, aerosol, ointment, foam, cream, gel, paste. This preferably is in the form of a controlled release formulation or sustained release formulation administered in a Therapeutically Effective Dose topically or injected directly into the skin adjacent to or within the area to be treated (intradermally or subcutaneously). The active compositions comprising the Senolytic Small Molecule can also be delivered via iontophoresis. Preservatives can be used to prevent the growth of fungi and other microorganisms. Suitable preservatives include, but are not limited to, benzoic acid, butylparaben, ethyl paraben, methyl paraben, propylparaben, sodium benzoate, sodium propionate, benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetypyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, thimerosal, and combinations thereof.
[000286] In some embodiments, compositions comprising the Senolytic Small Molecule can be formulated as emulsions for topical application. An emulsion contains one liquid distributed within the body of a second liquid. The emulsion may be an oil-in-water emulsion or a water- in-oil emulsion. Either or both of the oil phase and the aqueous phase may contain one or more surfactants, emulsifiers, emulsion stabilizers, buffers, and other excipients. The oil phase may contain other oily pharmaceutically-approved excipients. Suitable surfactants include, but are not limited to, anionic surfactants, non-ionic surfactants, cationic surfactants, and amphoteric surfactants. Compositions comprising the Senolytic Small Molecule for topical application may also include at least one suitable suspending agent, antioxidant, chelating agent, emollient, or humectant.
[000287] In some embodiments, ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents. Liquid sprays may be delivered from pressurized packs, for example, via a specially shaped closure. Oil-in-water emulsions can also be used in the compositions comprising the Senolytic Small Molecule, patches, bandages and articles. These systems are semi solid emulsions, micro-emulsions, or foam emulsion systems.
[000288] In some embodiments, the Senolytic Small Molecule(s) can be formulated with oleaginous bases or ointments to form a semisolid composition with a desired shape. In addition to the Senolytic Small Molecule, these semisolid compositions comprising the Senolytic Small Molecule can contain dissolved and/or suspended bactericidal agents, preservatives and/or a buffer system. A petrolatum component that may be included may be any paraffin ranging in viscosity from mineral oil that incorporates isobutylene, colloidal silica, or stearate salts to paraffin waxes. Absorption bases can be used with an oleaginous system. Additives may include cholesterol, lanolin (lanolin derivatives, beeswax, fatty alcohols, wool wax alcohols, low HLB (hydrophobellipophobe balance) emulsifiers, and assorted ionic and nonionic surfactants, singularly or in combination.
[000289] In some embodiments, a composition comprising any one of the Senolytic Small Molecules described herein may be formulated for sustained or slow release (which may also be called timed release or controlled release). Controlled or sustained release transdermal or topical formulations can be achieved by the addition of time-release additives, such as polymeric structures, matrices, that are available in the art. For example, the compositions comprising the Senolytic Small Molecule may be administered in a Therapeutically Effective Dose through use of hot-melt extrusion articles, such as bioadhesive hot-melt extruded film. The formulation can comprise a cross-linked polycarboxylic acid polymer formulation. A cross- linking agent may be present in an amount that provides adequate adhesion to allow the system to remain attached to target epithelial or endothelial cell surfaces for a sufficient time to allow the desired release of the compound.
[000290] In some embodiments, an insert, transdermal patch, bandage or article comprise a mixture or coating of polymers that provide release of the active agents at a constant rate over a prolonged period of time. In some embodiments, the article, transdermal patch or insert comprises water-soluble pore forming agents, such as polyethylene glycol (PEG) that can be mixed with water insoluble polymers to increase the durability of the insert and to prolong the release of the active ingredients.
[000291] Transdermal devices (inserts, patches, bandages) may also comprise a water insoluble polymer. In some embodiments, rate controlling polymers are useful for administration to sites where pH change can be used to effect release. Said rate controlling polymers are applied using a continuous coating film during the process of spraying and drying with the active compound. In one embodiment, the coating formulation is used to coat pellets comprising the active ingredients that are compressed to form a solid, biodegradable insert.
[000292] In some embodiments, a polymer formulation is utilized to provide controlled or sustained release including bioadhesive polymers. By way of example, a sustained-release gel and the compound may be incorporated in a polymeric matrix, such as a hydrophobic polymer matrix. Examples of a polymeric matrix include a microparticle. The microparticles can be microspheres, and the core may be of a different material than the polymeric shell. Alternatively, the polymer may be cast as a thin slab or film, a powder produced by grinding or other standard techniques, or a gel such as a hydrogel. In some embodiments, said polymer is in the form of a coating or part of a bandage, stent, catheter, vascular graft, or other device to facilitate delivery of the Senolytic Small Molecule. The matrices can be formed by solvent evaporation, spray drying, solvent extraction and other methods known to those skilled in the art.
[000293] In some embodiments of a method described herein for treating a cardiovascular disease associated with or caused by arteriosclerosis, one or more Senolytic Small Molecule(s) may be delivered directly into a blood vessel (e.g., an artery) via a stent. In some embodiments, a stent is used for delivering the Senolytic Small Molecule to an atherosclerotic blood vessel (an artery). Several methods are described in the art for preparing drug-coated and drug- embedded stents. In some embodiments the Senolytic Small Molecule may also be incorporated into the stent (for example as a coating or pores in the metal stent itself). In some embodiments, the Senolytic Small Molecule may be formulated within liposomes and applied to a stent. Placement of stents in an atherosclerotic artery is performed by a person skilled in the medical art.
[000294] In some embodiments, the Senolytic Small Molecule is administered in a Therapeutically Effective Dose to a subject who has an ophthalmic senescence-associated or disease or disorder may be delivered intraocularly or intravitreally. In other some embodiments, the Senolytic Small Molecule(s) may be administered in a Therapeutically Effective Dose to the eye by a conjunctival route, applying the Senolytic Small Molecule to the mucous membrane and tissues of the eyelid, either upper, lower, or both. Any of these administrations may be bolus infusions.
[000295] In some embodiments, the Senolytic Small Molecule(s) is administered in a Therapeutically Effective Dose in the forms that will promote intracellular delivery. In some embodiments, the Senolytic Small Molecule(s) is administered in a Therapeutically Effective Dose in the forms that will be recognized by the person in the skilled art.
[000296] As will be recognized by those skilled in the art in some embodiments the Senolytic Small Molecule(s) could be co-administered with enzyme inhibitors to alleviate proteolytic degradation thereby increasing bioavailability. In some embodiments of the Senolytic Small Molecule(s) is co-administered with enzyme inhibitor. [000297] As will be recognized by those skilled in the art in some embodiments, the Senolytic Small Molecule(s) could be co-administered with absorption enhancers such as chitin and its derivatives such as chitosan to enhance absorption of hydrophilic drug molecules. In some embodiments of the Senolytic Small Molecule(s) is co-administered with absorption enhancers such as chitin and its derivatives (i.e. chitosan).
[000298] The following embodiments demonstrate additional aspects of the disclosed subject matter.
[000299] The first embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule exhibits maximized interference with the CR3 domain of Fox04.
[000300] The second embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule exhibits reduced direct interaction with the p53DBD compared to that of the endogenous Fox04.
[000301] The third embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule preferably exhibits a reduced interference with the CR3 domains of FoxOl or Fox03 compared to that of said Fox04.
[000302] The forth embodiment is the method of the Senolytic Agents including but not limited to those listed in the Table 1, wherein the small molecule exhibits reduced interference with DNA compared to that of said Fox04.
[000303] The fifth embodiment is the method according to any one of the first to forth embodiments, wherein the senescent cell is characterized as expressing the senescence- associated secretory phenotype (SASP).
[000304] The sixth embodiment is the method according to any one of the first to forth embodiments, wherein the method comprises treatment of a senescence-associated disease or disorder.
[000305] The seventh embodiment is the method according to any one of the first to forth embodiments, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the small molecule is administered before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent.
[000306] The eight embodiment is the method according to any one of the first to forth embodiments, wherein the said cancer is characterized as resistant to therapy.
[000307] The ninth embodiment is the method according to any one of the first to forth embodiments, wherein said therapy-resistant cancer comprises is metastatic melanoma, breast cancer, or glioblastoma, and wherein the therapy to which the cancer is resistant is radiation therapy or chemotherapy.
[000308] The tenth embodiment is the method according to any one of the first to forth embodiments, wherein the subject comprises a human characterized as suffering from, or expected to suffer from chronic inflammatory diseases or a senescence related disease or disorder.
[000309] The eleventh embodiment is the method according to any one of the first to forth embodiments, wherein the method is effective to remove cells from the subject that express pl6INK4a, wherein the subject is characterized as suffering from, or expected to suffer from a senescence-associated disease or disorder.
[000310] The twelfth embodiment is the method according to any one of the first to forth embodiments, wherein the method is effective to alter levels of the Serine-46 phosphorylated p53 foci in the subject, wherein the subject is characterized as suffering from, or expected to suffer from, a senescence-associated disease or disorder.
[000311] The thirteenth embodiment is the method according to any one of the first to forth embodiments, wherein the method comprises administering the small molecules according to the Therapeutically Regime or an Impulse Regime, Sustained Regime, a Gentle Shock Regime, or combinations thereof.
[000312] The fourteenth embodiment is the method of inducing the apoptosis of a senescent cell in a subject, wherein the method uses one or more of the Senolytic Small Molecules listed in the Table 1. [000313] The fifteenth embodiment is the method of inducing the apoptosis of a senescent cell in a subject, wherein the method uses one or more compounds that carry a similar scaffold to the Senolytic Small Molecules listed in the Table 1.
[000314] The sixteenth embodiment is the method of use of the Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
[000315] The seventeenth embodiment is the method of use of the compounds that carry a similar chemical scaffold to any of the Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
[000316] The eighteenth embodiment is the method of use of the HIV reverse transcriptase inhibitors listed in the Table 1 for treatment of senescent related diseases and disorders.
[000317] The nineteenth embodiment is the method of use of the compounds that carry a similar chemical scaffold to the HIV reverse transcriptase inhibitors listed in the Table 1 for treatment of senescent related diseases and disorders.
[000318] The twentieth embodiment is the method of use of the reductase inhibitor listed in the Table 1 for treatment of senescent related diseases and disorders.
[000319] The twenty first embodiment is the method of use of the compounds that carry a similar chemical scaffold to the reductase inhibitor listed in the Table 1 for treatment of senescent related diseases and disorders.
[000320] The twenty second embodiment is the method of use of the anti-cancer Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
[000321] The twenty third embodiment is the method of use of the compounds that carry a similar chemical scaffold to the anti-cancer Senolytic Small Molecules listed in the Table 1 for treatment of senescent related diseases and disorders.
[000322] The twenty forth embodiment is the method according to any one of claims 1-10, wherein causing the Senolytic Small Molecule to interfere with the CR3 domain of Fox04 of the senescent cell comprises administering a pharmaceutical composition comprising the Senolytic Small Molecule according to any one of the embodiments. [000323] The twenty fifth embodiment is the method according to any one of the embodiments, wherein a small molecule exhibits maximized interference with the CR3 domain of F ox04.
[000324] The twenty sixth embodiment is the method according to any one of the embodiments, wherein a small molecule exhibits reduced direct interaction with the p53DBD compared to that of the endogenous Foxo4.
[000325] The twenty seventh embodiment is the method according to any one of the embodiments, wherein a small molecule preferably exhibits a reduced interference with the CR3 domains of FoxOl or Fox03 compared to that of said Fox04.
[000326] The twenty eighth embodiment is the method according to any one of the embodiments, wherein a small molecule exhibits reduced interference with DNA compared to that of said Fox04.
[000327] The twenty ninth embodiment is the method according to any one of the embodiments, wherein the senescent cell is characterized as expressing the senescence- associated secretory phenotype (SASP).
[000328] The thirtieth embodiment is the method according to any one of the embodiments, wherein the method comprises administering Therapeutic Regimes for treatment of a senescence-associated disease or disorder. Another embodiment is the method of use of the Senolytic Small Molecules, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the Senolytic Agent is administered before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent.
[000329] The thirty first embodiment is the method according to any one of the embodiments, wherein the said cancer is characterized as resistant to therapy.
[000330] The thirty second embodiment is the method according to any one of the embodiments, wherein said therapy-resistant cancer comprises is metastatic melanoma, breast cancer, or glioblastoma, and wherein the therapy to which the cancer is resistant to radiation therapy or chemotherapy. [000331] The thirty third embodiment is the method according to any one of the embodiments, wherein the subject comprises a human characterized as suffering from, or expected to suffer from chronic inflammatory diseases or a senescence related disease or disorder.
[000332] The thirty forth embodiment is the method according to any one of the embodiments, wherein the method is effective to remove cells from the subject that express pl6INK4a, wherein the subject is characterized as suffering from, or expected to suffer from a senescence- associated disease or disorder.
[000333] The thirty fifth embodiment is the method according to any one of the embodiments, wherein the method is effective to alter levels of the Serine-46 phosphorylated p53 foci in the subject, wherein the subject is characterized as suffering from, or expected to suffer from, a senescence-associated disease or disorder.
[000334] The thirty sixth embodiment is the method according to any one of the embodiments, wherein the method comprises Senolytic Therapy using one or more Senolytic Small Molecules. Another embodiment is the method use of the Senolytic Small Molecules, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the Senolytic Agent is administered as Therapeutic Regimes before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent or during an immunotherapy.
[000335] As will be recognized by those skilled in the art, the Senolytic Small Molecule(s) and the disclosed method described in the present application can be modified and varied over a tremendous range of applications to produce a wide range of Senolytic Small Molecules which may be directed to specific therapies which target those subsets of senescent cells and accordingly the scope of patented subject matter is not limited by any of the specific exemplary teachings given. It is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.
[000336] None of the description in the present application should be read as implying that any particular element, step, or function is an essential element which must be included in the claim scope: THE SCOPE OF PATENTED SUBJECT MATTER IS DEFINED ONLY BY THE ALLOWED CLAIMS. Moreover, none of these claims are intended to invoke paragraph six of 35 USC section 112 unless the exact words“means for” are followed by a participle.

Claims

What is claimed is:
1. A Senolytic Small Molecule listed in the Table 1 for use in inducing the apoptosis of a senescent cell in a subject.
2. A compound having a similar scaffold to the Senolytic Small Molecules listed in the Table 1, for use in inducing the apoptosis of a senescent cell in a subject.
3. Senolytic Small Molecules listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
4. A compound having a similar chemical scaffold to any of the Senolytic Small Molecules listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
5. A HIV reverse transcriptase inhibitor with the ZINC IDs of 95566015 and 377845189 listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
6. A compound having a similar chemical scaffold to the HIV reverse transcriptase inhibitors listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
7. A reductase inhibitor with the ZINC ID of 13513540 listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
8. A compound having a similar chemical scaffold to the reductase inhibitors listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
9. An anti-cancer Senolytic Small Molecule with the ZINC IDs of 22863770 and 5390317 listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
10. A compound having a similar chemical scaffold to the anti-cancer Senolytic Small Molecules listed in the Table 1 for use in the treatment of senescent related diseases and disorders.
11. A pharmaceutical composition comprising the Senolytic Small Molecule listed in the Table 1 or a compound having a similar scaffold to the Senolytic Small Molecules listed in the Table 1 according to any one of claims 1-10.
12. The use according to any one of claims 1-10, wherein the Senolytic Small Molecule is caused to interfere with the CR3 domain of Fox04 of the senescent cell by administering the pharmaceutical composition according to claim 11 to the subject.
13. The use according to any one of claims 1-10 and 12, wherein the Senolytic Small Molecule exhibits maximized interference with the CR3 domain of Fox04.
14. The use according to any one of claims 1-10, 12 and 13, wherein the Senolytic Small Molecule exhibits reduced direct interaction with the p53DBD compared to that of the endogenous Foxo4.
15. The use according to any one of claims 1-10, and 12-14, wherein the Senolytic Small Molecule exhibits a reduced interference with the CR3 domains of FoxOl or Fox03 compared to that of said Fox04.
16. The use according to any one of claims 1-10 and 12-15, wherein the Senolytic Small Molecule exhibits reduced interference with DNA compared to that of said Fox04.
17. The use according to any one of claims 1-10 and 12-16, wherein the senescent cell is characterized as expressing the senescence-associated secretory phenotype (SASP).
18. The use according to any one of claims 1-10 and 12-17, further comprising administering Therapeutic Regimes for the treatment of a senescence-associated disease or disorder.
19. The use according claim 18, wherein the disease or disorder is cancer, and wherein the subject is a mammal, preferably a human, and wherein the Senolytic Agent is administered according to the Therapeutic Regime before, during, and/or after subjecting the subject to radiation therapy, and/or before, during or after administering to the subject at least one chemotherapeutic agent or during an immunotherapy.
20. The use according to any one of claims 18-19, wherein said cancer is characterized as resistant to therapy.
21. The use according to any one of claims 18-20, wherein said therapy-resistant cancer comprises is metastatic melanoma, breast cancer, or glioblastoma, and wherein the therapy to which the cancer is resistant to is radiation therapy or chemotherapy.
22. The use according to any one of claims 1-10 and 12-21, wherein the subject comprises a human characterized as suffering from, or expected to suffer from chronic inflammatory diseases or a senescence related disease or disorder.
23. The use according to any one of claims 1-10 and 12-22, wherein the treatment is effective to remove cells from the subject that express pl6INK4a, wherein the subject is characterized as suffering from, or expected to suffer from a senescence-associated disease or disorder.
24. The use according to any one of claims 1-10 and 12-23, wherein the treatment is effective to alter levels of the Serine-46 phosphorylated p53 foci in the subject, wherein the subject is characterized as suffering from, or expected to suffer from, a senescence- associated disease or disorder.
25. The use according to any one of claims 1-10 and 12-24, wherein the treatment comprises Senolytic Therapy using one or more Senolytic Small Molecules.
PCT/EP2020/059436 2019-04-02 2020-04-02 Repurposing small molecules for senescence-related diseases and disorders WO2020201444A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962828230P 2019-04-02 2019-04-02
US62/828,230 2019-04-02

Publications (1)

Publication Number Publication Date
WO2020201444A1 true WO2020201444A1 (en) 2020-10-08

Family

ID=70289741

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/059436 WO2020201444A1 (en) 2019-04-02 2020-04-02 Repurposing small molecules for senescence-related diseases and disorders

Country Status (1)

Country Link
WO (1) WO2020201444A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021052962A1 (en) * 2019-09-16 2021-03-25 Instituto de Medicina Molecular João Lobo Antunes Treatment of spinal cord injury
CN115561468A (en) * 2022-10-18 2023-01-03 深圳市瑞格生物科技有限公司 Method for evaluating risk of having tumor or specific tumor

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180015137A1 (en) * 2015-01-23 2018-01-18 Erasmus University Medical Center Rotterdam Anti-senescence compounds and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180015137A1 (en) * 2015-01-23 2018-01-18 Erasmus University Medical Center Rotterdam Anti-senescence compounds and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CARREGAL ANA PAULA ET AL: "Docking-based virtual screening of Brazilian natural compounds using the OOMT as the pharmacological target database", JOURNAL OF MOLECULAR MODELING, SPRINGER BERLIN HEIDELBERG, BERLIN/HEIDELBERG, vol. 23, no. 4, 11 March 2017 (2017-03-11), pages 1 - 9, XP036211470, ISSN: 1610-2940, [retrieved on 20170311], DOI: 10.1007/S00894-017-3253-8 *
LI GUANGYUN ET AL: "Kukoamine A attenuates insulin resistance and fatty liver through downregulation of Srebp-1c", BIOMEDICINE AND PHARMACOTHERAPY, ELSEVIER, FR, vol. 89, 28 February 2017 (2017-02-28), pages 536 - 543, XP029987356, ISSN: 0753-3322, DOI: 10.1016/J.BIOPHA.2017.02.024 *
WANG, F. ET AL.: "Biochemical and structural characterization of an intramolecular interaction in FOX03a and its binding with p53", J MOL BIOL, vol. 384, no. 3, 2008, pages 590 - 603, XP025885485, DOI: 10.1016/j.jmb.2008.09.025

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021052962A1 (en) * 2019-09-16 2021-03-25 Instituto de Medicina Molecular João Lobo Antunes Treatment of spinal cord injury
CN115561468A (en) * 2022-10-18 2023-01-03 深圳市瑞格生物科技有限公司 Method for evaluating risk of having tumor or specific tumor
CN115561468B (en) * 2022-10-18 2024-02-02 深圳市瑞格生物科技有限公司 Method for assessing risk of suffering from tumor or specific tumor

Similar Documents

Publication Publication Date Title
JP6688224B2 (en) Treatment of osteoarthritis in joints by administration of means that inhibit MDM2
JP2021175754A (en) Combination of hdac inhibitor and anti-pd-1 antibody for treatment of cancer
EA013966B1 (en) Use of erythropoietin for treatment of diabetes melitus
JP2000510460A (en) Treatment of epithelial malignancies using squalamine in combination with other anticancer drugs
WO2019126739A1 (en) Pyrvinium pamoate anti-cancer therapies
EA032345B1 (en) Method of treating cancer using coenzyme q10
DK3137090T3 (en) USE OF GINSENOSIDE M1 FOR TREATMENT OF IGA NEPHROPATHY
WO2020201444A1 (en) Repurposing small molecules for senescence-related diseases and disorders
DE60021360T2 (en) VEGF ANGIOGENIC GROWTH FACTORS FOR THE TREATMENT OF PERIPHERAL NEUROPATHY
US20200255489A1 (en) Novel Senolytic Peptides
TWI798320B (en) Therapeutic agent for nervous system disease
US20200031873A1 (en) Repurposing cell penetrating peptides and their novel derivatives and iopromide and iodo-aryl carbonates for treatment of senescence-related diseases and disorders
JP2006513223A (en) Inhibition of melanogenesis and melanoma metastasis by p-aminobenzoic acid (PABA)
JP2014526518A (en) Compounds and methods for treating heart failure or neuronal damage
KR102520382B1 (en) Acyl sulfonamides as BCL family antagonists for use in the clinical management of conditions induced or mediated by senescent cells and for the treatment of cancer
US20220088015A1 (en) Methods and pharmaceutical compositions for the treatment of age-related cardiometabolic diseases
TWI776451B (en) Combinations of bcl-2/bcl-xl inhibitors and related uses
CN117597131A (en) Novel composition for treating coronavirus pneumonia comprising taurodeoxycholic acid or pharmaceutically acceptable salt thereof as active ingredient
JP7406192B2 (en) Fibrosis treatment drug
US10688128B2 (en) Use of Z-butylidenephthalide in activating autoimmune system
JP2022523465A (en) Fibroblasts and their microvesicles to reduce the toxicity associated with cancer immunotherapy
JP6559357B2 (en) Use of butylidenephthalide
JP2021507934A (en) Use of compounds in the manufacture of therapeutic agents for cerebral microvascular disease
US20230008508A1 (en) Combination therapies for the treatment of cancer
CN108685896B (en) Application of oroxylin A in preparation of medicine for treating and/or preventing chronic peripheral vascular occlusive diseases

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: 20719324

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20719324

Country of ref document: EP

Kind code of ref document: A1