Mitochondrial-enhanced therapy for primary mitochondrial diseases

文档序号:816424 发布日期:2021-03-26 浏览:56次 中文

阅读说明:本技术 用于原发性线粒体疾病的线粒体增强疗法 (Mitochondrial-enhanced therapy for primary mitochondrial diseases ) 是由 娜塔莉·伊夫吉奥哈纳 乌列·哈拉维 史密尔·布克施班 诺亚·谢尔 莫里亚·布鲁金 于 2019-07-22 设计创作,主要内容包括:本发明提供了富集有功能性线粒体的人类干细胞,其中健康的功能性外源线粒体占线粒体富化的人类干细胞中总线粒体的至少3%并少于33%。还公开了生产此类细胞的方法及其用于治疗原发性线粒体疾病的用途。(The invention provides human stem cells enriched for functional mitochondria wherein healthy functional exogenous mitochondria comprise at least 3% and less than 33% of the total mitochondria in the mitochondria-enriched human stem cells. Also disclosed are methods of producing such cells and their use for treating primary mitochondrial diseases.)

1. A therapeutic agent for use in a human patient in need of such treatmentA pharmaceutical composition for the development of a mitochondrial disease, disorder or symptom thereof, said composition comprising at least 10% in a pharmaceutically acceptable liquid medium capable of supporting cell survival5To 2x107(ii) individual human stem cells per kilogram patient body weight, wherein the human stem cells are enriched in freeze-thawed, healthy functional human extramitochondrial free of pathogenic mutations in mitochondrial DNA, and wherein the healthy functional human extramitochondrial comprises at least 3% and less than 33% of total mitochondria in the mitochondria-enriched human stem cells.

2. The pharmaceutical composition of claim 1, wherein the enriching comprises introducing mitochondria at a dose of at least 0.088 up to 176 milliunits CS activity per million cells in the stem cells.

3. The pharmaceutical composition of claim 2, wherein the enriching comprises contacting the stem cells with mitochondria at a dose of 0.88 up to 17.6 milliunits CS activity per million cells.

4. The pharmaceutical composition of any one of claims 1 to 3, wherein the healthy functional human allogenic mitochondria are syngeneic or allogeneic.

5. The pharmaceutical composition of any one of claims 1 to 4, wherein the human stem cell is CD34+

6. The pharmaceutical composition of any one of claims 1 to 5, wherein the primary mitochondrial disease or disorder is associated with a mutation in mitochondrial DNA.

7. The pharmaceutical composition of claim 6, wherein the primary mitochondrial disease or disorder associated with mutations in mitochondrial DNA is selected from Pearson syndrome, Kearns-Sayre syndrome, mitochondrial encephalopathy lactic acidosis and stroke-like attack (MELAS) syndrome, Leber's Hereditary Optic Neuropathy (LHON), neuropathy, ataxia and retinitis pigmentosa (NARP) syndrome, myoclonic epilepsy with fluffy-like-red-fiber (MERRF) syndrome, maternal hereditary diabetes and deafness (MIDD), Alpers-like syndrome, Chronic Progressive External Ophthalmoplegia (CPEO), mitochondrial DNA-related forms of Congenital Lactic Acidosis (CLA), Mitochondrial DNA Depletion Syndrome (MDDS) and mitochondrial DNA-related forms of Leigh syndrome.

8. The pharmaceutical composition of any one of claims 1 to 5, wherein the primary mitochondrial disease or disorder is associated with a mutation in nuclear DNA.

9. The pharmaceutical composition of claim 8, wherein the primary mitochondrial disease or disorder associated with mutations in nuclear DNA is selected from the group consisting of mitochondrial neurogastrointestinal encephalopathy (MNGIE) syndrome, Alpers syndrome, Friedrich's Ataxia (FA), progressive extraocular Paralysis (PEO), sideroblasts anemia, ataxia-mediated neuropathy syndrome (ANS), mendelian neurodegenerative mitochondrial disease, 3-methylpentenedionate urethritis (MEG) with deafness (D), encephalopathy (E), and Leigh-like disease (L) syndrome (MEGDEL), Sengers syndrome, mini-morphic nephropathy syndrome (MCNS), nuclear DNA-related forms of congenital acidosis (CLA), Mitochondrial DNA Depletion Syndrome (MDDS), and nuclear DNA-related forms of Leigh syndrome.

10. The pharmaceutical composition of any one of claims 1 to 9, wherein the primary mitochondrial disease or disorder is associated with an organ selected from kidney, liver, brain, muscle, pancreas, eye, and any combination thereof.

11. The pharmaceutical composition of any one of claims 1 to 10, wherein the symptom is selected from impaired walking ability, impaired motor skills, impaired language skills, impaired memory, impaired weight gain, growth retardation, low blood alkaline phosphatase levels, low blood magnesium levels, high blood creatinine levels, low blood bicarbonate levels, low blood alkali excess levels, high urine glucose/creatinine ratios, high urine chloride/creatinine ratios, high urine sodium/creatinine ratios, high blood lactate levels, high urine magnesium/creatinine ratios, high urine potassium/creatinine ratios, high urine calcium/creatinine ratios, diabetes, magnesium urine, high blood urea levels, low C-peptide levels, high HbA1C levels, hypoparathyroidism, ptosis, hearing loss, cardiac conduction disorders, seizures, stroke-like symptoms, stroke-related disorders, stroke-related to-related conditions, EEG is impaired, blood aspartate Aminotransferase (AST) levels are high, blood alanine Aminotransferase (ALT) levels are high, ATP levels and oxygen consumption in lymphocytes are low.

12. The pharmaceutical composition of any one of claims 1 to 11, wherein the pharmaceutical composition is administered to a specific tissue or organ.

13. The pharmaceutical composition of any one of claims 1 to 11, wherein the pharmaceutical composition is administered by systemic administration.

14. The pharmaceutical composition of claim 13, comprising about 106Individual mitochondrially enriched human stem cells per kilogram patient body weight.

15. The pharmaceutical composition of any one of claims 1 to 12, comprising a total of about 5x105To 5x109Individual mitochondrially enriched human stem cells.

16. The pharmaceutical composition of any one of claims 1 to 15, wherein the mitochondrially enriched human stem cell has at least one of the following:

corresponding levels in stem cells prior to enrichment relative to mitochondria

(i) Increased mitochondrial DNA content;

(ii) increased CS activity levels;

(iii) increased levels of at least one mitochondrial protein selected from SDHA and COX 1;

(iv) increased O2A consumption rate;

(v) increased ATP production rate; or

(vi) Any combination thereof.

17. The pharmaceutical composition of any one of claims 1 to 16, wherein the human stem cells are obtained or derived from the patient prior to enrichment with the exogenous mitochondria.

18. The pharmaceutical composition of any one of claims 1 to 16, wherein the human stem cells are obtained or derived from a donor different from the patient prior to enrichment with the exogenous mitochondria.

19. The pharmaceutical composition of claim 18, wherein the donor is at least partially HLA matched to the patient.

20. The pharmaceutical composition of any one of claims 1 to 19, wherein the human stem cells are hematopoietic stem cells.

21. The pharmaceutical composition of any one of claims 1 to 19, wherein the human stem cell is a mesenchymal stem cell.

22. The pharmaceutical composition of any one of claims 1 to 19, wherein the human stem cell is a Pluripotent Stem Cell (PSC) or an Induced Pluripotent Stem Cell (iPSC).

23. The pharmaceutical composition of any one of claims 2 to 22, wherein the human stem cells have undergone at least one freeze-thaw cycle prior to introduction of the freeze-thawed, healthy, functional human extramitochondrial into the human stem cells.

24. The pharmaceutical composition of any one of claims 1 to 23, wherein the human stem cells are isolated, derived or obtained from cells of bone marrow, adipose tissue, oral mucosa, skin fibroblasts, blood or umbilical cord blood.

25. The pharmaceutical composition of any one of claims 1 to 24, wherein the healthy functional exogenous mitochondria are isolated or obtained from a placenta, placental cells grown in culture, or blood cells.

26. The pharmaceutical composition of any one of claims 1 to 25, wherein the human stem cells have undergone at least one freeze-thaw cycle after being enriched with the healthy functional human ex vivo mitochondria.

27. The pharmaceutical composition of any one of claims 1 to 26, wherein the healthy, functional, exogenous mitochondria comprise between 5% and 30% of total mitochondria.

28. The pharmaceutical composition of claim 27, wherein the healthy, functional, exogenous mitochondria comprise between 10% and 30% of total mitochondria.

29. An ex vivo method of enriching human stem cells with healthy functional human extramitochondrial, the method comprising the steps of:

(i) providing a first composition comprising a plurality of isolated or partially purified human stem cells from a patient having a mitochondrial disease, disorder or symptom thereof or from a donor;

(ii) providing a second composition comprising a plurality of isolated, freeze-thawed, healthy, functional human extramitochondrial obtained from a donor that does not have a pathogenic mutation in mitochondrial DNA;

(iii) freeze-thawed, healthy, functional human allomitochondria of the first composition of human stem cells and the second composition are activated at 0.088-176mU CS activity/106(ii) the ratio of individual stem cells, thereby providing a third composition; and

(iv) incubating the third composition under conditions that allow the freeze-thawed, healthy, functional, human allogenic mitochondria to enter the human stem cells, thereby enriching the human stem cells with the healthy, functional, human allogenic mitochondria, thereby providing a fourth composition comprising mitochondria-enriched human stem cells;

wherein the healthy functional human extramitochondrial accounts for at least 3% and less than 33% of total mitochondria in the fourth composition.

30. The method of claim 29, wherein the conditions that allow healthy functional human exogenous mitochondria to enter human stem cells comprise incubating the human stem cells with the healthy functional human exogenous mitochondria for a time in the range of 0.5 to 30 hours at a temperature in the range of 16 to 37 ℃.

31. The method of claim 29 or claim 30, wherein prior to incubation, the method further comprises a single centrifugation of the human stem cells and healthy functional human exogenous mitochondria at a centrifugal force of greater than 2500 xg.

32. The method of any one of claims 29 to 31, wherein the human stem cell is CD34+

33. The method of any one of claims 29 to 32, wherein the level of mitochondrial enrichment in mitochondrially enriched human stem cells is determined from at least one of:

(i) the level of endogenous and exogenous mitochondrial DNA in the case of allogeneic mitochondria;

(ii) the level of citrate synthase activity;

(iii) levels of succinate dehydrogenase complex flavoprotein subunit a (sdha) or cytochrome C oxidase (COX1) protein;

(iv) oxygen (O)2) A consumption rate;

(v) (ii) rate of ATP production; or

(vi) Any combination thereof.

34. The method of any one of claims 29 to 33, further comprising freezing the mitochondrially enriched human stem cells, and optionally further comprising thawing the mitochondrially enriched human stem cells.

35. The method of any one of claims 29 to 34, further comprising expanding the stem cells prior to or after enrichment with the healthy functional human ex vivo mitochondria.

36. A composition comprising a plurality of human stem cells enriched in healthy functional human extramitochondrial obtained by the method of any one of claims 29 to 35, wherein the healthy functional human extramitochondrial accounts for at least 3% and less than 33% of total mitochondria in the mitochondria-enriched human stem cells.

37. A method of treating a primary mitochondrial disease, disorder or symptom thereof in a human patient in need thereof, comprising the step of parenterally administering to the patient a pharmaceutical composition comprising at least about 5x105To 5x109A human stem cell, wherein the human stem cell is enriched in a freeze-thawed, functional human extramitochondrial free of pathogenic mutations in mitochondrial DNA, wherein the functional human extramitochondrial healthy comprises at least 3% and less than 33% of total mitochondria in the mitochondrially enriched human stem cell.

38. A method of treating a primary mitochondrial disease or disorder or symptoms thereof in a patient in need of treatment comprising administering a pharmaceutical composition according to any one of claims 1 to 28.

Technical Field

The present invention relates to mammalian stem cells, more particularly human stem cells enriched for exogenous functional human mitochondria. The invention also relates to methods for their production and therapeutic methods that utilize such enriched cells.

Background

Mitochondria are membrane-bound organelles present in most eukaryotic cells, ranging in diameter from 0.5 to 1.0 μm. Mitochondria are present in almost all eukaryotic cells, and their number and location vary with cell type. Mitochondria contain their own dna (mtdna) and their own mechanisms for synthesizing RNA and proteins. mtDNA contains only 37 genes, and therefore most gene products in mammals are encoded by nuclear DNA.

Mitochondria perform a number of essential tasks in eukaryotic cells, such as pyruvate oxidation, the krebs cycle, and the metabolism of amino acids, fatty acids, and steroids. However, the main function of mitochondria is to utilize the electron transport chain and the oxidative phosphorylation system ("respiratory chain") to generate energy as Adenosine Triphosphate (ATP). Other processes in which mitochondria are involved include thermogenesis, calcium ion storage, calcium signaling, programmed cell death (apoptosis) and cell proliferation. Thus, a number of diseases and disorders associated with mitochondrial dysfunction or dysfunction are known in the art that require treatment.

Intracellular ATP concentrations are typically 1-10 mM. ATP can be produced by redox reactions using monosaccharides and complex sugars (carbohydrates) or lipids as energy sources. For complex fuels to be used for the synthesis of ATP, it is first necessary to break them down into smaller, simpler molecules. Complex carbohydrates are hydrolyzed into monosaccharides such as glucose and fructose. Fat (triglycerides) is metabolized to give fatty acids and glycerol.

The overall process of oxidation of glucose to carbon dioxide is known as cellular respiration, and about 30 ATP molecules can be produced from a single glucose molecule. ATP may be produced by a number of different cellular processes. The three major pathways for energy production in eukaryotic organisms are glycolysis and citric acid cycle/oxidative phosphorylation (both components of cellular respiration) and β -oxidation. This ATP production by non-photosynthetic aerobic eukaryotes occurs mostly in mitochondria, which can account for nearly 25% of the total typical cell volume.

Mitochondrial diseases are a class of diseases caused by dysfunctional mitochondria. Primary mitochondrial diseases may be caused by mutations in mitochondrial DNA that affect mitochondrial function or by mutations in genes of nuclear DNA whose products are imported into the mitochondria (mitochondrial proteins). Mitochondrial diseases have unique characteristics, both because the mode of the disease is usually inherited and because mitochondria are critical to cellular function. A subset of these diseases with symptoms of neuromuscular disease is commonly referred to as mitochondrial myopathy. Unlike primary mitochondrial diseases, secondary mitochondrial dysfunction, also known as acquired mitochondrial dysfunction, can be caused by genes of nuclear DNA that are not directly involved in the mitochondrial oxidative phosphorylation cascade. The affected genes neither encode mitochondrial proteins nor affect OXPHOS by affecting the production of the complex machinery required to operate the oxidative phosphorylation (OXPHOS) process. Secondary mitochondrial dysfunction may be accompanied by a number of diseases or disorders such as fatty liver disease, myocardial infarction and stroke, and may also be obtained following adverse environmental or drug-related effects that may cause oxidative stress. The latter may cause mtDNA alterations and/or dysfunctional mitochondria, as seen in various other processes that adversely affect mitochondria, such as aging, inflammatory responses, mitochondrial toxic drugs, and the like.

Mitochondrial disease may become clinically apparent after the number of affected mitochondria reaches a certain level; this phenomenon is called the "heterogeneity threshold". Mitochondrial DNA mutations occur frequently due to inefficient error checking capabilities, and because DNA is naked and not as protected as nuclear histones. This means that mitochondrial DNA disorders can occur spontaneously and relatively frequently. Defects in enzymes that control mitochondrial DNA replication, all of which are encoded by genes in nuclear DNA, may also cause mitochondrial DNA mutations. Most mitochondrial function and biogenesis are controlled by nuclear DNA. Human mitochondrial DNA encodes only 13 respiratory chain proteins, while the majority of the 1,500 proteins and components targeted to mitochondria are estimated to be nuclear-encoded. Defects in nuclear-encoded mitochondrial genes are associated with a wide range of clinical disease phenotypes including anemia, dementia, epilepsy, diabetes, myopathy, hypertension, lymphoma, retinopathy, seizures, and neurodevelopmental disorders.

Pearson Syndrome (PS) is a mitochondrial disease characterized by bone marrow failure, anemia, and pancreatic dysfunction. Other clinical features are growth arrest, pancreatic fibrosis with insulin dependent diabetes mellitus and exocrine pancreatic insufficiency, renal insufficiency, muscle and nervous system injury. Few patients who survive into adulthood often develop symptoms of Kearns-Sayre syndrome (KSS).

Renal fanconi syndrome or fanconi syndrome is a syndrome of insufficient reabsorption of the proximal tubules of the kidney. The syndrome may be caused by a variety of different underlying congenital or acquired diseases, toxicities or adverse drug reactions. This results in the trafficking of various small molecules of metabolism into the urine, rather than reabsorption from the renal tubule fluid.

KSS is a mitochondrial myopathy, a more severe syndrome variant of chronic progressive extraocular paralysis (CPEO), a syndrome characterized by muscles involved in controlling eye and eyelid movement in isolation. KSS causes ptosis and ophthalmoplegia. KSS includes a combination of CPEO and pigmented retinopathy in the eye, as well as cardiac conduction abnormalities. Other symptoms may include cerebellar ataxia, proximal muscle weakness, deafness, diabetes, growth hormone deficiency, and hypoparathyroidism.

Leber Hereditary Optic Neuropathy (LHON) or Leber optic atrophy is a degeneration of the mitochondrial inheritance (from mother to offspring) of Retinal Ganglion Cells (RGCs) and their axons, which causes acute or subacute loss of central vision, primarily affecting young adult males. However, LHON is only transmitted through the mother, as it is mainly caused by mutations in the mitochondrial (not nuclear) genome, and only the ovum contributes mitochondria to the embryo. LHON is usually caused by one of three pathogenic mitochondrial dna (mtdna) point mutations. These mutations were mutated from G to A at nucleotide 11778, G to A at nucleotide 3460 and T to C at nucleotide 14484 in the ND4, ND1 and ND6 subunit genes of complex I of the oxidative phosphorylation chain in mitochondria, respectively. These mutations can lead to a reduction in cellular energy production, which in turn leads to cellular damage and death of certain optic nerve cells. At present, experts are unable to determine which family members will develop symptoms, although on average 50% of men with LHON mutations and 15% of women with LHON mutations will lose vision during their lifetime.

Mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes, abbreviated as MELAS, are members of the mitochondrial cytopathic family that also includes MERRF and Leber hereditary optic neuropathy. The disease may manifest itself in both sexes. MELAS is caused by genetic mutations in mitochondrial DNA. Some of the genes affected in MELAS (MT-ND1, MT-ND5) encode proteins that are part of NADH dehydrogenase (also known as Complex I) in mitochondria, which dehydrogenase contributes to the conversion of oxygen and monosaccharides into energy. Other genes (MT-TH, MT-TL1 and MT-TV) encode mitochondrial specific transfer RNA (tRNA). Mutations in MT-TL1 caused more than 80% of all MELAS cases. They impair the mitochondria' ability to make protein, use oxygen, and generate energy.

Mitochondrial Respiratory Chain Disorders (MRCD) are a heterogeneous group of disorders that share a cellular bioenergy mechanism in that they affect the molecular defect of the mitochondrial oxidative phosphorylation system (OXPHOS). Although they tend to affect primarily the nervous system and skeletal muscles, clinically they often involve multiple tissues. Heart disease manifests itself frequently and includes hypertrophic or dilated cardiomyopathy and defects in cardiac conduction as part of a multi-system mitochondrial disorder in adults or infants, or less frequently, as isolated clinical conditions. In certain embodiments, the mitochondrial disease is Mitochondrial Respiratory Chain Disease (MRCD).

Stem cells are generally cells that can differentiate into other types of cells and/or can divide to produce more stem cells of the same type. In mammals, the major types of stem cells are embryonic stem cells and adult stem cells. There are at least three known sources of adult stem cells in humans: bone marrow stem cells, adipose tissue stem cells, and blood stem cells. Other stem cells include Mesenchymal Stem Cells (MSCs), tissue-specific stem cells and induced pluripotent stem cells (ipscs).

WO 2013/002880 describes compositions and methods comprising bioenergy agents for restoring the quality of aged oocytes, enhancing oocytes or improving their derivatives (e.g. cytoplasm or isolated mitochondria) for use in procedures for enhancing fertility.

WO 2013/035101 to the present inventors relates to mitochondrial compositions and methods of treatment using them and discloses partially purified functional mitochondrial compositions and methods of using the compositions, including treating conditions that benefit from increased mitochondrial function by administering the compositions to a subject in need thereof.

WO 2016/008937 relates to a method of transferring mitochondrial cells isolated from a donor cell population into a recipient cell population. The method shows improved transport efficiency for a certain amount of mitochondria.

US 2012/0107285 relates to mitochondrial enhancement of cells. Certain embodiments include, but are not limited to, methods of modifying stem cells or methods of administering modified stem cells to at least one biological tissue.

WO 2016/135723 relates to human bone marrow cells enriched in at least 50% functional mitochondria, methods for their production and methods of treatment using such cells.

There remains a need in the art for new methods of increasing mitochondrial function in cells and organs affected by a variety of different primary mitochondrial diseases and disorders.

Disclosure of Invention

Mitochondrial-enhanced therapy was first used to ameliorate the deficiencies of various physiological parameters in children suffering from severe effects caused by defective mitochondria. Although it has been hypothesized that the positive effects of mitochondrial-enhancing therapy can restore defective mitochondrial function, it has never been successfully implemented in human juvenile patients. It is noteworthy that it is now disclosed that even a low level of healthy mitochondrial enrichment can successfully provide a very beneficial long-lasting improvement in the health of patients and a significant improvement in the physiological parameters of various organs and tissues.

Previously, it has been shown using animal model systems that it is easy to increase the mitochondrial content of host cells by more than 50% or more than 100%. As exemplified below, it has now been found that even a small increase in mitochondria from the donor to the recipient can achieve the desired clinical result.

The present invention provides mammalian stem cells enriched for exogenous functional mitochondria and methods of treating various primary mitochondrial diseases. In particular, the invention provides compositions comprising human stem cells that have been enriched for functional mitochondria obtained from healthy donors. The invention also provides methods of treating a subject having a primary mitochondrial disorder using allogeneic or autologous "mitochondria-enriched" stem cells.

Providing stem cells from a subject with a mitochondrial disease, treating ex vivo and returning to the same subject provides a great benefit compared to other methods involving allogeneic cell therapy. For example, the methods provided herein eliminate the need for a long, costly and not always successful process of screening a population and finding donors that match the patient's Human Leukocyte Antigen (HLA). The method also advantageously eliminates the need for life-long immunosuppressive therapy for preventing rejection of the allogeneic cell population. Thus, the present invention advantageously provides a unique method of ex vivo orthotherapy, wherein cells are harvested from a patient's body, treated ex vivo with exogenous (e.g., allogeneic) mitochondria, and returned to the same patient. Furthermore, the invention relates to the administration of stem cells which are empirically distributed in different tissues and organs of the body and which enhance mitochondrial function at these sites.

The present invention is based, in part, on the surprising discovery that treatment of adolescents with primary mitochondrial disorders with mitochondria-enriched stem cells increases mitochondrial function and levels in target tissues and organs and ameliorates a wide variety of adverse conditions and symptoms associated with mitochondrial dysfunction.

It has also been surprisingly found, as exemplified herein for the first time, that enriched human stem cells are effective in treating a variety of different diseases and conditions in human patients, even if the mitochondrial content of these cells is only moderately increased after mitochondrial enrichment. Although WO 2016/135723, which is among certain inventors of the present invention, relates to mitochondrial enrichment of at least 50% of bone marrow cells, it has surprisingly been found that a mitochondrial enrichment of about 5% to about 45% of human stem cells is sufficient to provide significant improvements in a number of clinical parameters in human patients that persist for a long period of time.

The compositions and methods provided herein can be considered a form of mitochondrial "potentiation therapy". According to the principles of the present invention, the reduction in the number of functional mitochondria and/or the reduction in mitochondrial function can be alleviated by the addition of stem cells enriched in wild-type, healthy functional mitochondria. The stem cells enriched in intact functional mitochondria are fused or entered into the tissues and organs of a patient, increasing both mitochondrial copy number per cell/tissue/organ and mitochondrial function. Furthermore, cells exhibiting low levels of mitochondrial function may be replaced by the addition of stem cells enriched for functional mitochondria in accordance with the principles of the present invention. It is postulated that the enriched stem cells may differentiate into the same type of cells as the cells that have been damaged or have low function, thereby improving dysfunction or restoring function.

In one aspect, the invention provides a method of treating a primary mitochondrial disease, disorder or symptom thereof in a human patient in need thereof, comprising the step of parenterally administering to the patient a pharmaceutical composition comprising at least about 5x105To 5x109A human stem cell, wherein the human stem cell is enriched in a freeze-thawed, healthy, functional, human exogenous mitochondria that are free of pathogenic mutations in mitochondrial DNA, and wherein the healthy, functional, exogenous mitochondria comprise at least 3% and less than 33% of total mitochondria in the mitochondria-enriched human stem cell.

In another aspect, the invention provides a pharmaceutical composition for treating a primary mitochondrial disease, disorder or symptom thereof in a human patient in need thereof, the composition comprising at least 10% in a pharmaceutically acceptable liquid medium capable of supporting cell survival5To 2x107(ii) individual human stem cells per kilogram patient body weight, wherein the human stem cells are enriched in freeze-thawed, healthy, functional human extramitochondrial free of pathogenic mutations in mitochondrial DNA, wherein the healthy, functional human extramitochondrial comprises at least 3% and less than 33% of total mitochondria in the mitochondria-enriched human stem cells.

In certain embodiments, the enriching comprises introducing mitochondria at a dose of at least 0.088 up to 176 milliunits CS activity per million cells in the stem cells.

In further embodiments, the enriching comprises contacting the stem cells with mitochondria at a dose of 0.88 up to 17.6 milliunits CS activity per million cells. In certain embodiments, the dose of isolated mitochondria is added to the recipient cell at a desired concentration. The ratio of the number of mitochondrial donor cells to the number of mitochondrial acceptor cells is higher than 2:1 (donor cells compared to acceptor cells). In typical embodiments, the ratio is at least 5 or at least 10 or higher. In particular embodiments, the ratio of donor cells to recipient cells from which mitochondria are collected is at least 20, 50, 100, or possibly even higher. Each possibility is a separate embodiment.

In certain embodiments, the human stem cell is CD34+

In certain embodiments, the healthy functional human allogenic mitochondria are allogeneic mitochondria. In other embodiments, the healthy functional human extramitochondrial is syngeneic.

In certain embodiments, the primary mitochondrial disease or disorder is associated with a mutation in mitochondrial DNA. In certain embodiments, the primary mitochondrial disease or disorder associated with mutations in mitochondrial DNA is selected from Pearson Syndrome (PS), Kearns-Sayre syndrome (KSS), mitochondrial encephalopathy, lactic acidosis and stroke-like attack (MELAS) syndrome, Leber Hereditary Optic Neuropathy (LHON), neuropathy, ataxia and retinitis pigmentosa (NARP) syndrome, myoclonic epilepsy with fleshy red fiber (MERRF) syndrome, maternal hereditary diabetes and deafness (MIDD), coats-like syndrome, chronic progressive extraocular paralysis (CPEO), mitochondrial DNA-related forms of Congenital Lactic Acidosis (CLA), Mitochondrial DNA Depletion Syndrome (MDDS) and mitochondrial DNA-related forms of Leigh syndrome. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the primary mitochondrial disease or disorder is associated with a mutation in nuclear DNA encoding a gene product essential for mitochondrial function. In certain embodiments, the primary mitochondrial disease or disorder associated with a mutation in nuclear DNA is selected from the group consisting of mitochondrial neurogastrointestinal encephalopathy (MNGIE) syndrome, Alpers syndrome, Friedrich's Ataxia (FA), progressive extraocular Paralysis (PEO), sideroblasts anemia, Ataxia Neuropathy Syndrome (ANS), mendelian neurodegenerative mitochondrial disease, 3-methylpentene-dionuria (MEG) with deafness (D), encephalopathy (E), and Leigh-like disease (L) syndrome (MEGDEL), Sengers syndrome, mini-morbid nephropathy syndrome (MCNS), nuclear DNA-related forms of Congenital Lactic Acidosis (CLA), nuclear DNA-related forms of Mitochondrial DNA Depletion Syndrome (MDDS), and nuclear DNA-related forms of Leigh syndrome. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the primary mitochondrial disease or disorder is associated with an organ selected from the group consisting of kidney, brain, muscle, pancreas, eye, and any combination thereof. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the symptom of impaired mitochondrial function is selected from impaired walking ability, impaired motor skills, impaired language skills, impaired memory, impaired weight gain, growth retardation, low blood alkaline phosphatase levels, low blood magnesium levels, high blood creatinine levels, low blood bicarbonate levels, low blood excess levels, high urine glucose/creatinine ratios, high urine chloride/creatinine ratios, high urine sodium/creatinine ratios, high blood lactate levels, high urine magnesium/creatinine ratios, high urine potassium/creatinine ratios, high urine calcium/creatinine ratios, diabetes, magnesium urine, high blood urea levels, low C-peptide levels, high HbA1C scores, hypoparathyroidism, ptosis, hearing loss, cardiac conduction disorders, seizure, stroke-like seizures, impaired EEG, impaired memory, impaired weight gain, impaired growth of body weight, impaired blood glucose/creatinine ratios, high urine chloride/creatinine ratios, high urine sodium/creatinine ratios, high urine lactate levels, high urine magnesium/creatinine ratios, high urine potassium/creatinine ratios, high urine calcium/creatinine ratios, high urine, Blood AST/ALT is high, and ATP content and oxygen consumption in lymphocytes are low. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the pharmaceutical composition is administered to a specific tissue or organ.

In certain embodiments, the pharmaceutical composition is administered by systemic administration. In certain embodiments, the pharmaceutical composition comprises about 106Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises a total of about 5x105To 5x109Individual mitochondrially enriched human stem cells.

In certain embodiments, the mitochondrially enriched human stem cell has at least one of the following: (ii) an increased mitochondrial DNA content relative to the corresponding level in the stem cell prior to mitochondrial enrichment; (ii) increased CS activity levels; (iii) increased levels of at least one mitochondrial protein selected from SDHA and COX 1; (iv) increased O2A consumption rate; (v) increased rate of ATP production; or (vi) any combination thereof. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the human stem cells are obtained or derived from the patient prior to enrichment with the exogenous mitochondria. In certain embodiments, the human stem cells are obtained or derived from a donor different from the patient prior to enrichment with the exogenous mitochondria.

In certain embodiments, the donor of the stem cell is at least partially HLA matched to the patient.

In certain embodiments, the mitochondrially enriched human stem cell is a mitochondrially enriched human progenitor cell. In certain embodiments, the mitochondrially enriched human stem cell is a hematopoietic stem cell. In certain embodiments, the mitochondrially enriched human stem cell is a mesenchymal stem cell. In certain embodiments, the mitochondrially enriched human stem cell is a Pluripotent Stem Cell (PSC) or an Induced Pluripotent Stem Cell (iPSC).

In certain embodiments, the human stem cells have undergone at least one freeze-thaw cycle prior to introducing the freeze-thawed, healthy, functional human exogenous mitochondria into the human stem cells. In certain embodiments, the method comprises (a) freezing the human stem cells, (b) thawing the human stem cells, and (c) introducing freeze-thawed, healthy, functional exogenous mitochondria into the human stem cells.

In certain embodiments, the human stem cells are isolated, derived or obtained from cells of the bone marrow. In other embodiments, the human stem cells are isolated, derived or obtained from adipose tissue, oral mucosa, skin fibroblasts, blood or umbilical cord blood. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the healthy functional exogenous mitochondria are isolated or obtained from a placenta, placental cells grown in culture, or blood cells. Each possibility represents a separate embodiment of the invention. In certain embodiments, the healthy functional exogenous mitochondria are isolated or obtained from a human placenta, human placental cells grown in culture, or human blood cells. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the human stem cells have undergone at least one freeze-thaw cycle after enrichment with the healthy functional human extramitochondrial. In certain embodiments, the above method further comprises the following additional steps prior to administering the human stem cells enriched for healthy functional exogenous mitochondria to the patient: (a) freezing the human stem cells enriched for healthy functional exogenous mitochondria, and (b) thawing the human stem cells enriched for healthy functional exogenous mitochondria.

In certain embodiments, the healthy, functional, exogenous mitochondria comprise between 5% and 30% of total mitochondria. In certain embodiments, the healthy, functional, exogenous mitochondria comprise at least 10% and less than 30% of total mitochondria. In certain embodiments, the healthy, functional, exogenous mitochondria comprise at least 10% and less than 25% of total mitochondria.

In another aspect, the present invention also provides an ex vivo method of enriching human stem cells with healthy functional human extramitochondrial, the method comprising the steps of: (i) providing a first composition comprising a plurality of isolated or partially purified human stem cells from a patient having a mitochondrial disease, disorder or symptom thereof or from a donor; (ii) providing a second composition comprising a plurality of isolated, freeze-thawed, healthy, functional human extramitochondrial obtained from a donor that does not have a pathogenic mutation in mitochondrial DNA; (iii) freeze-thawed, healthy, functional human allomitochondria of the first composition of human stem cells and the second composition are activated at 0.088-176mU CS activity/106(ii) the ratio of individual stem cells, thereby providing a third composition; and (iv) incubating the third composition under conditions that allow the freeze-thawed, healthy, functional human allogenic mitochondria to enter the human stem cells, thereby enriching the human stem cells with the healthy, functional human allogenic mitochondria, thereby providing a fourth composition comprising mitochondria-enriched human stem cells; whereinThe total mitochondria of said fourth composition comprise at least 3% and less than 33% of said healthy functional human extramitochondrial.

In certain embodiments, the conditions that allow healthy functional human exogenous mitochondria to enter human stem cells comprise incubating the human stem cells with the healthy functional human exogenous mitochondria for a time in the range of 0.5 to 30 hours at a temperature in the range of 16 to 37 ℃. In certain embodiments, the conditions that allow healthy functional human exogenous mitochondria to enter human stem cells comprise incubating the human stem cells with the healthy functional human exogenous mitochondria in a culture medium at a temperature in the range of 16 to 37 ℃ for a time in the range of 0.5 to 30 hours in an environment that supports cell survival. According to certain embodiments, the medium is saline containing human serum albumin. In certain embodiments, the conditions for incubation include a 5% CO2Of the atmosphere (c). In certain embodiments, the conditions for incubating do not include adding CO above the level present in air2

In certain embodiments, the method further comprises centrifuging the human stem cells and healthy functional exogenous mitochondria before, during, or after the incubating. In certain embodiments, the method further comprises a single centrifugation of the human stem cells and healthy functional human exo-source mitochondria at a centrifugal force above 2500xg prior to incubation.

In certain embodiments, the mitochondria that have undergone a freeze-thaw cycle exhibit a comparable rate of oxygen consumption after thawing as compared to a control mitochondria that has not undergone a freeze-thaw cycle.

In certain embodiments, the methods further comprise freezing the mitochondria-enriched human stem cells, and optionally further comprise thawing the mitochondria-enriched human stem cells.

In further embodiments, the human stem cells are expanded before or after mitochondrial enhancement.

In certain aspects and embodiments, the present invention provides a composition comprising a plurality of human stem cells enriched in healthy functional exogenous mitochondria obtained by the methods described above in various embodiments thereof, wherein the healthy functional exogenous mitochondria comprise at least 3% and less than 33% of total mitochondria in the mitochondria-enriched human stem cells.

In another aspect, the invention also provides a method of treating a primary mitochondrial disease or disorder or symptoms thereof in a human patient in need thereof, comprising the step of administering to said patient a pharmaceutical composition comprising a mitochondria-enriched human stem cell as described above.

Further embodiments and a full scope of applicability of the present invention will become apparent from the detailed description given hereinafter. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. Other limitations and disadvantages of these systems will become apparent to one of skill in the art, through comparison of conventional and traditional methods with some aspects of the present invention as set forth in the remainder of the present application with reference to the drawings.

Drawings

FIG. 1 shows fluorescence confocal microscopy obtained CD34 incubated with GFP-labeled mitochondria isolated from HeLa-TurboGFP-mitochondrial cells+Photomicrographs of the cells.

Fig. 2A is a bar graph showing copy number of C57BL mtDNA in FVB/N bone marrow cells after incubation with various concentrations (0.044, 0.44, 0.88, 2.2, 4.4, 8.8, 17.6 milliunits CS activity) of exogenous mitochondria from C57BL mice compared to untreated cells (NTs).

Figure 2B is a bar graph showing the level of mtDNA-encoded (COX1) protein in FVB/N bone marrow cells after incubation with various concentrations (0.044, 0.44, 0.88, 2.2, 4.4, 8.8, 17.6 milliunits of CS activity) of exogenous mitochondria from C57BL mice, normalized to Janus levels, compared to untreated cells (NTs).

Figure 2C is a bar graph showing the amount of nuclear-encoded (SDHA) protein in FVB/N bone marrow cells normalized to Janus levels after incubation of the cells with various concentrations (0.044, 0.44, 0.88, 2.2, 4.4, 8.8, 17.6 milliunits of CS activity) of exogenous mitochondria from C57BL mice compared to untreated cells (NTs).

Fig. 3A is a bar graph showing a comparison of CS activity in murine BM cells after enrichment with increasing amounts of GFP-labeled mitochondria.

FIG. 3B is a bar graph showing cytochrome c reductase activity in these cells (black bars) compared to activity in GFP-labeled mitochondria (grey bars).

Figure 4 is a bar graph showing the level of C57BL mtDNA in the bone marrow of FVB/N mice at various time points post IV injection of bone marrow cells enriched for exogenous mitochondria from C57BL mice.

FIG. 5 is a dot-matrix diagram showing healthy donor CD34 after MAT with two different placenta-derived mitochondrial batches (PLC1 and PLC2)+Percentage of placental haplotypes in cells.

Figure 6A is a bar graph showing a comparison of CS activity in control untreated human BM cells and centrifuged or non-centrifuged human BM cells incubated with GFP-labeled mitochondria isolated from human placental cells.

Figure 6B is a bar graph showing ATP levels in control untreated human BM cells and centrifuged human BM cells incubated with GFP-labeled mitochondria isolated from human placental cells.

FIG. 7A is a graphical illustration of mtDNA deletion in cord blood cells of a patient with Pearson's syndrome and a southern blot analysis showing the deletion.

Figure 7B is a bar graph showing human mtDNA copy number in bone marrow of NSGS mice 2 months after mitochondrial-enhanced therapy with human mitochondrial enriched cord blood cells of pearson syndrome (UCB + Mito) compared to mice injected with non-enhanced cord blood cells (UCB).

FIG. 8 is a bar graph showing the level of FVB/N ATP8 mutant mtDNA in bone marrow of FVB/N mice 1 month after administration of stem cells enriched with healthy functional mitochondria obtained from C57/BL placenta.

Fig. 9A is a plan view of different stages of treatment for a patient with Pearson Syndrome (PS) provided by the present invention.

Fig. 9B is a bar graph showing the change in MET score over time before and after therapy for PS patients treated by the methods provided in the present invention.

Fig. 9C is a bar graph showing the lactate levels found in the blood of PS patients treated by the methods provided in the present invention as a function of time before (B) and after therapy.

Fig. 9D is a line graph showing the standard deviation scores for weight and height of PS patients treated by the methods provided in the present invention as a function of time before and after therapy.

Fig. 9E is a line graph showing the change in alkaline phosphatase (ALP) levels over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 9F is a line graph showing the long-term elevation of blood Red Blood Cell (RBC) levels in PS patients before and after the therapy provided by the present invention.

Fig. 9G is a line graph showing the long-term elevation of blood Hemoglobin (HGB) levels in PS patients before and after the therapy provided by the present invention.

Fig. 9H is a line graph illustrating the long-term rise in blood Hematocrit (HCT) levels in PS patients before and after the therapy provided by the present invention.

Fig. 9I is a bar graph showing the change in blood magnesium levels before and after therapy in PS patients treated by the methods provided in the present invention before and after magnesium supplementation over time.

Fig. 9J is a line graph showing the change in creatinine levels over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 9K is a line graph showing the change in bicarbonate levels over time before and after therapy for PS patients treated by the methods provided in the present invention.

Fig. 9L is a line graph showing the change in base excess levels over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 9M is a bar graph showing the change in the ratio of glucose to creatinine in urine over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 9N is a bar graph showing the change in the ratio of potassium to creatinine over time before and after therapy in the urine of PS patients treated by the methods provided in the present invention.

Figure 9O is a bar graph showing the change in the ratio of chloride to creatinine over time before and after therapy in the urine of PS patients treated by the methods provided in the present invention.

Figure 9P is a bar graph showing the change in the ratio of sodium to creatinine over time before and after therapy in the urine of PS patients treated by the methods provided in the present invention.

Fig. 10A is a line graph showing the change in the amount of normal mtDNA before and after therapy in 3 PS patients (pt.1, pt.2 and pt.3) treated by the method provided in the present invention, measured by digital PCR of the deleted region (in each patient), compared with 18S genomic DNA representing the normal mtDNA amount per cell, and normalized according to baseline.

Fig. 10B is a line graph showing the level of heterogeneity (presence of deleted mtDNA compared to total mtDNA) in 3 PS patients (pt.1, pt.2, and pt.3) at baseline after MAT. The dashed line represents the baseline for each patient.

FIG. 11A is another plan view of different stages of treatment for a patient with Pearson Syndrome (PS) further provided by the present invention.

Fig. 11B is a bar graph showing the variation of lactate levels in the blood over time before (B) and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 11C is a bar graph showing the change in sitting trial score over time before and after therapy for PS patients treated by the methods provided in the present invention.

Fig. 11D is a bar graph showing the 6 minute walk test score of PS patients treated by the methods provided in the present invention as a function of time before and after therapy.

Figure 11E is a bar graph showing the ergometer score of three consecutive replicates (R1, R2, R3) of PS patients treated by the methods provided in the present invention as a function of time before and after therapy.

Fig. 11F is a bar graph showing the change in urinary magnesium to creatinine ratio over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 11G is a bar graph showing the change in urinary potassium to creatinine ratio over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 11H is a bar graph showing the change in urinary calcium to creatinine ratio over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 11I is a bar graph showing the ratio of ATP8 to 18S copy number in urine of PS patients treated by the methods provided in the present invention as a function of time before and after therapy.

Fig. 11J is a bar graph showing ATP levels in lymphocytes of PS patients treated by the methods provided in the present invention as a function of time before and after therapy.

FIG. 12A is yet another plan view of different stages of treatment for a patient with Pearson Syndrome (PS) and a patient with Kearns-Sayre syndrome (KSS) further provided by the present invention.

Fig. 12B is a bar graph illustrating the variation of lactate levels in the blood over time before (B) and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 12C is a bar graph showing the AST and ALT levels over time before and after therapy for PS patients treated by the methods provided in the present invention.

Fig. 12D is a bar graph showing the change in triglyceride, total cholesterol, and VLDL cholesterol levels over time before and after therapy in PS patients treated by the methods provided in the present invention.

Fig. 12E is a bar graph showing the change in hemoglobin A1C (HbA1C) score over time before and after therapy for PS patients treated by the methods provided in the present invention.

Fig. 12F is a line graph showing the change in sitting trial score over time before and after therapy for PS patients (pt.3) treated by the methods provided by the present invention.

Fig. 12G is a line graph showing the change in 6-minute walk test score before and after therapy for PS patients (pt.3) treated by the methods provided herein over time.

FIG. 13 is a bar graph showing ATP levels in peripheral blood of KSS patients treated by methods provided herein before and after therapy.

Detailed Description

The present invention provides a cell platform, more particularly a stem cell platform, for targeted and/or systemic delivery of therapeutically significant amounts of exogenous functional healthy mitochondria. The invention also provides methods of producing such cell platforms and methods of using them in the treatment of mitochondrial diseases.

It has now been shown for the first time that even human stem cells moderately enriched with healthy functional exogenous mitochondria can achieve systemic delivery of healthy functional mitochondria to organs, tissues and cells in vivo in patients with mitochondrial diseases and disorders of diverse pathology.

Providing stem cells moderately enriched for functional mitochondria enables improved therapy of primary mitochondrial diseases in humans that have not been available to date. For example, primary mitochondrial diseases associated with mutations in mitochondrial DNA, such as Pearson Syndrome (PS) and Kearns-Sayre syndrome (KSS), can now be treated by transplanting stem cells that are only moderately enriched for functional mitochondria into the tissues or organs affected by the disease, resulting in long-term elimination of the disease. In the case where the disease-affected cells are stem cells themselves, the administered enriched stem cells may replace the affected cells, again resulting in long-term elimination of the disease. In other examples, where the primary mitochondrial disease is associated with a mutation in nuclear DNA and the affected cells are stem cells or are derived from stem cells, the administered stem cells can replace the affected cells, again resulting in long-term elimination of the disease. It should be emphasized that the present invention provides for the first time stem cell-based means and methods for the sustained correction of the pathological state of primary mitochondrial diseases and the long-term elimination of these diseases in humans, all that is required is a low to moderate mitochondrial enrichment of these cells prior to administration.

The present invention is also based on several surprising findings, one of which is that a single administration of mitochondrially enriched human stem cells is sufficient to improve the overall physiological and cognitive state of a human patient, including the function of the pancreas of organs such as kidney, liver, brain, muscle, for at least one year, as determined by the results of various clinical parameters, without repeated intervention. Although a single round of therapy is sufficient to achieve long-term effects in a variety of different organs and conditions, there is still the possibility that more rounds of treatment will be required to maintain at least some of these effects.

In one aspect, the invention provides a method of treating a primary mitochondrial disease, disorder or symptom thereof in a subject in need thereof, the method comprising the step of administering to the patient a pharmaceutical composition comprising a plurality of stem cells enriched in healthy, functional, exogenous mitochondria that are free of pathogenic mutations in mitochondrial DNA. In certain embodiments, the subject is a mammalian subject and the stem cells are mammalian stem cells. In certain embodiments, the subject is a human subject and the stem cells are human stem cells.

In another aspect, the invention provides a pharmaceutical composition for treating a primary mitochondrial disease, disorder or symptom thereof in a human patient in need thereof, the composition comprising a plurality of human stem cells enriched in freeze-thawed, healthy, functional, exogenous mitochondria that are free of pathogenic mutations in mitochondrial DNA, wherein the healthy, functional, exogenous mitochondria comprise at least 3% and less than 33% of the total mitochondria in the mitochondria-enriched human stem cells.

In certain embodiments, the pharmaceutical composition comprises at least 105To 4x107Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises at least 105To 2x107Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises at least 5x105To 1.5x107Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises at least 106To 107Individual mitochondrially enriched human stem cells per kilogram patient body weight. In other embodiments, the pharmaceutical composition comprises at least 105Or at least 106Individual mitochondrially enriched human stem cells per kilogram patient body weight. Each possibility represents a separate embodiment of the invention. In certain embodiments, the pharmaceutical composition comprises at least 5x10 in total5Up to 5x109Individual mitochondrially enriched human stem cells. In certain embodiments, the pharmaceutical composition comprises a total of at least 106Up to 109Individual mitochondrially enriched human stem cells. In other embodiments, the pharmaceutical composition comprises at least 2x10 in total6Up to 5x108Individual mitochondrially enriched human stem cells.

In another aspect, the invention provides a composition comprising a plurality of human CD34 enriched for healthy, functional exogenous mitochondria+Stem cells, wherein said healthy functional exogenous mitochondria comprise mitochondria-enriched human CD34+At least 3% and less than 33% of total mitochondria in the stem cell.

In another aspect, the invention provides a pharmaceutical composition for the treatment of a primary mitochondrial disease or disorder or a symptom thereof, wherein the medicament isThe composition comprises a plurality of human CD34 enriched with healthy functional exogenous mitochondria+Stem cells, wherein said healthy functional exogenous mitochondria comprise mitochondria-enriched human CD34+At least 3% and less than 33% of total mitochondria in the stem cell. As used herein, the term "method" refers to manners, means, techniques and procedures for accomplishing a given task, including those that are known, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmaceutical, biological, biochemical and medical arts.

As used herein, the term "treating" includes the alleviation, alleviation or amelioration of at least one symptom associated with or caused by a disease or disorder. As used herein, the term "treatment" also includes prophylactic (e.g., prophylactic), palliative and curative treatment.

As used herein, the term "pharmaceutical composition" refers to any composition comprising at least one biologically active agent. The term "pharmaceutical composition" as used herein also refers to a composition comprising an active pharmaceutical ingredient to be delivered to a subject, e.g., to achieve a therapeutic, prophylactic, diagnostic, preventative, or prognostic effect. The term "pharmaceutical composition" as used herein also refers to any composition comprising human stem cells, optionally further comprising a vehicle or carrier that maintains the cells therein in a viable state. In certain embodiments, the pharmaceutical composition comprises an active pharmaceutical ingredient and a pharmaceutically acceptable carrier. As used herein, the term "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, which are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, and combinations thereof.

As used herein, the term "bioactive agent" refers to any molecule capable of eliciting a response in a biological system, such as living cells, tissues, organs, and humans. Non-limiting examples of biologically active agents according to the present invention include cells, intact mitochondria, mitochondrial DNA, and mitochondrial proteins. In accordance with the principles of the present invention, a plurality of human stem cells enriched for healthy functional exogenous mitochondria without pathogenic mutations in mitochondrial DNA are bioactive agents.

As used herein, the term "stem cell" generally refers to any type of stem cell. Stem cells are undifferentiated cells that can differentiate into other types of cells and can divide to produce more stem cells of the same type. The stem cells may be totipotent or pluripotent. As used herein, the term "human stem cell" generally refers to all stem cells naturally occurring in humans, as well as all stem cells produced or derived ex vivo and compatible with humans. Like stem cells, "progenitor cells" have a tendency to differentiate into a particular type of cell, but are already more specific than stem cells and are pushed to differentiate into its "target" cells. The most important difference between stem cells and progenitor cells is that stem cells can replicate indefinitely, while progenitor cells can divide only a limited number of times. As used herein, the term "human stem cell" also includes "progenitor cells" and "stem cells that are not fully differentiated.

In accordance with the principles of the present invention, stem cells are enriched with healthy, functional exogenous mitochondria prior to administration to a patient in need thereof, in order to increase the number and/or function of mitochondria therein. Without being bound by any theory or mechanism, the increased number and/or function of mitochondria in the administered stem cells results in a variety of different therapeutic effects, as first exemplified herein, in human patients.

The terms "functional mitochondria," "healthy functional mitochondria," and "healthy functional exogenous mitochondria" are used interchangeably herein and refer to mitochondria that do not have pathogenic mutations in the mitochondrial DNA, which exhibit normal, non-pathological levels of activity. Mitochondrial activity can be measured by various methods known in the art, such as tetramethylrhodamine ethyl perchlorate (TMRE) staining, O2Consumption, ATP production and CS activity level.

In the embodiments exemplified hereinafter, the mitochondria are human mitochondria.

The terms "healthy donor" and "healthy subject" are used interchangeably and refer to a subject that does not have the disease or disorder to be treated.

In certain embodiments, the healthy functional exogenous mitochondria are syngeneic or allogeneic.

As used herein, the term "enrichment" refers to any action performed ex vivo that increases the mitochondrial content of human cells, e.g., the number of intact, functionally healthy mitochondria. In accordance with the principles of the present invention, healthy functional exogenous mitochondria are introduced into human stem cells, thereby enriching these cells with healthy functional exogenous mitochondria. It will be appreciated that this enrichment alters the mitochondrial content of the human stem cells: original timeHuman stem cells have essentially one host/autologous/endogenous population of mitochondria, while human stem cells enriched for exogenous mitochondria have essentially two populations of mitochondria-one population being host/autologous/endogenous mitochondria and the other population being introduced mitochondria (i.e., exogenous mitochondria). Thus, the term "enriched" refers to the state of the cell after receiving/incorporating exogenous mitochondria. Determining the number of and/or ratio between the two mitochondrial populations is straightforward, as the two populations differ in several respects, for example in their mitochondrial DNA. Thus, the phrase "human stem cells enriched in healthy functional human mitochondria" is equivalent to the phrase "human stem cells comprising endogenous mitochondria and healthy functional exogenous mitochondria". For example, a human stem cell comprising at least 1% and less than 33% of total mitochondria of healthy functional exogenous mitochondria is considered to comprise host/autologous/endogenous mitochondria and healthy functional exogenous mitochondria in a ratio of 99:1 to 67: 33. For example, "3% of total mitochondria" means that the original (endogenous) mitochondrial content after enrichment is 97% of total mitochondria and the introduced (exogenous) mitochondria is 3% of total mitochondria, which is equivalent to(3/97 ═ 3.1% enrichment. Another example, "33% of total mitochondria" means that after enrichment the original (endogenous) mitochondrial content is 67% of total mitochondria and the introduced (exogenous) mitochondria is 33% of total mitochondria, which is equivalent to an enrichment of (33/67 ═ 49.2%.

In certain embodiments, the healthy, functional, exogenous mitochondria comprise at least 1% and less than 33% of total mitochondria in the stem cell. In certain embodiments, the healthy, functional, exogenous mitochondria comprise between about 1%, 3%, 5%, 7%, 10%, 15%, or 20% to about 25%, 27%, 29%, or 31% of total mitochondria. Each possibility represents a separate embodiment of the invention. As used herein, the term "about" means 10% above or below the given number. For example, about 10% means 9%, 9% to 11%, or 11%. Generally, numerical values used herein refer to ± 10% of the stated numerical value.

In certain embodiments, the healthy, functional, exogenous mitochondria comprise at least 1% of the total mitochondria of the stem cell, wherein the stem cell is not a bone marrow cell or a cell derived or derived therefrom. In certain embodiments, the healthy, functional, exogenous mitochondria comprise between about 1% to about 99% of the total mitochondria and any subrange thereof of the stem cell, wherein the stem cell is not a bone marrow cell or a cell derived or derived therefrom. In certain embodiments, the healthy, functional, exogenous mitochondria comprise at least about 1%, 3%, 5%, 7%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the total mitochondria of the stem cell, wherein the stem cell is not a bone marrow cell or a cell derived or derived therefrom. Each possibility represents a separate embodiment of the invention.

The degree to which the stem cells are enriched for functional mitochondria can be determined by functional and/or enzymatic assays, including but not limited to oxygen (O)2) Consumption rate, level of citrate synthase or activity, rate of Adenosine Triphosphate (ATP) production. In the alternative, the stem cells are used with healthy donorsEnrichment of mitochondria can be confirmed by detection of mitochondrial DNA of the donor. According to certain embodiments, the degree to which the stem cells are enriched for functional mitochondria can be determined by the level of heterogeneity variation and/or by the mtDNA copy number per cell. Each possibility represents a separate embodiment of the invention.

TMRM (tetramethylrhodamine methyl ester) or related TMRE (tetramethylrhodamine ethyl ester) is a cell-penetrating fluorogenic dye commonly used to assess mitochondrial function in living cells by identifying changes in mitochondrial membrane potential. According to certain embodiments, the level of enrichment may be determined by staining with TMRE or TMRM. According to certain embodiments, enrichment of the stem cells with healthy functional mitochondria can be determined by conventional assays well known in the art. In certain embodiments, the level of mitochondrial enrichment in the mitochondria-enriched human stem cell is determined by: (i) levels of host/endogenous mitochondrial DNA and exogenous mitochondrial DNA; (ii) (ii) a level of a mitochondrial protein selected from Citrate Synthase (CS), COX1, SDHA, and any combination thereof; (iii) (ii) a level of CS activity; or (iv) any combination of (i), (ii), and (iii). Each possibility represents a separate embodiment of the invention. In certain embodiments, the level of mitochondrial enrichment in the mitochondria-enriched human stem cell is determined by at least one of: (i) levels of host (endogenous) defective mitochondrial DNA and healthy exogenous mitochondrial DNA in the case of allogeneic mitochondria; (ii) the level of citrate synthase activity; (iii) levels of succinate dehydrogenase complex flavoprotein subunit a (sdha) or cytochrome C oxidase (COX1) protein; (iv) oxygen (O)2) A consumption rate; (v) rate of Adenosine Triphosphate (ATP) production; or (vi) any combination thereof. Each possibility represents a separate embodiment of the invention.

It should be understood that, as used herein, the phrase "human stem cell enriched with healthy functional exogenous mitochondria" refers to a human stem cell comprising healthy functional mitochondria, wherein said healthy functional mitochondria have a different origin from said human stem cell, i.e. these mitochondria are obtained/derived/isolated from an external source. The presence of "foreign", "foreign" or "non-primitive" healthy functional mitochondria in human stem cells serves as evidence that these cells are enriched for said mitochondria. One of ordinary skill in the art will know how to determine, based on methods well known in the art, that human stem cells contain exogenous mitochondria from different origins (see, e.g., Zander j. et al, Forensic sci. int. genet.,2017, vol.29, page 242-249). These methods may be based on, for example, genetic differences within a human stem cell or between different mitochondrial populations within a plurality of human stem cells. For example, in humans, mitochondrial DNA encodes 37 genes (Nature.290(5806): 457-65), so by sequencing the mtDNA one can readily determine the presence of 1, 2 or more different mtDNA populations within a human stem cell or within a plurality of human stem cells. In certain embodiments, the level of mitochondrial enrichment in a mitochondrially enriched human stem cell is determined by sequencing at least a statistically representative portion of total mitochondrial DNA in the cell and determining the relative levels of host/endogenous mitochondrial DNA and exogenous mitochondrial DNA. In certain embodiments, the level of mitochondrial enrichment in the mitochondria-enriched human stem cells is determined by Single Nucleotide Polymorphism (SNP) analysis. In certain embodiments, the largest population of mitochondria and/or the largest population of mitochondrial DNA is the host/endogenous population of mitochondria and/or the host/endogenous population of mitochondrial DNA; and/or the second large population of mitochondria and/or the second large population of mitochondrial DNA is the exogenous population of mitochondria and/or the exogenous population of mitochondrial DNA. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the enriching of the stem cells with healthy functional human exogenous mitochondria comprises washing the mitochondria-enriched stem cells after incubating the human stem cells with the healthy functional human exogenous mitochondria. This step provides a composition of mitochondrially enriched stem cells that is substantially free of cell debris or mitochondrial membrane residues and mitochondria that do not enter the stem cells. In certain embodiments, washing comprises centrifuging the mitochondrially enriched stem cells after incubating the human stem cells with the healthy functional human exo-source mitochondria. According to some embodiments, the pharmaceutical composition comprising the mitochondrially enriched human stem cell is separated from free mitochondria, i.e. mitochondria or other cell debris that has not entered the stem cell. According to certain embodiments, the pharmaceutical composition comprising mitochondrially enriched human stem cells does not comprise a detectable amount of free mitochondrion.

As used herein and in the claims, the term "mitochondrial disease" and the term "primary mitochondrial disease" are used interchangeably. As used herein, the term "primary mitochondrial disease" refers to a mitochondrial disease that is diagnosed by known or undisputed pathogenic mutations in the mitochondrial DNA or by mutations in the genes of the nuclear DNA whose gene products are imported into the mitochondria. According to some embodiments, the primary mitochondrial disease is a congenital disease. According to certain embodiments, the primary mitochondrial disease is not secondary mitochondrial dysfunction. The terms "secondary mitochondrial dysfunction" and "acquired mitochondrial dysfunction" are used interchangeably throughout this application.

According to certain embodiments, the primary mitochondrial disease is characterized by sub-normal mitochondrial parameters in the affected cells. According to certain embodiments, the affected cells have (i) a sub-normal rate of oxygen (O2) consumption; (ii) a sub-normal level or activity of citrate synthase; (iii) a sub-normal rate of Adenosine Triphosphate (ATP) production; or (iv) any combination of (i), (ii), and (iii).

As used herein, the term "subnormal oxygen (O)2) Consumption rate "means substantially lower than control oxygen (O)2) Consumption rate of oxygen (O)2) Consumption rate, the control oxygen (O)2) The rate of depletion is derived from or corresponds to oxygen (O) found in the corresponding cell or corresponding mitochondria of the subject or subjects not suffering from a mitochondrial disorder2) The rate of consumption.

As used herein, the term "sub-normal level or activity level of citrate synthase" refers to a level or activity of citrate synthase that is substantially lower than a control level or activity level of citrate synthase derived from or corresponding to the level or activity of citrate synthase in a subject or subjects not suffering from a mitochondrial disorder.

As used herein, the term "sub-normal rate of Adenosine Triphosphate (ATP) production" refers to a rate of Adenosine Triphosphate (ATP) production that is substantially lower than a control rate of Adenosine Triphosphate (ATP) production that is derived from or corresponds to the rate of Adenosine Triphosphate (ATP) production found in a respective cell or a respective mitochondrion of a subject or subjects that does not have a mitochondrial disorder.

In certain embodiments, the term "substantially lower" as used herein refers to a statistically significant reduction below normal. In certain embodiments, the term "substantially lower" as used herein refers to a pathological reduction, i.e. to a level where at least one pathological symptom associated with said substantially lower value becomes apparent.

In certain embodiments, the term "sub-normal" as used herein refers to a value that is at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, or at least 10-fold lower than the corresponding value found in the corresponding cell or corresponding mitochondria of a subject or subjects not suffering from a mitochondrial disorder. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the primary mitochondrial disease or disorder is associated with a mutation in mitochondrial DNA. As used herein, the phrase "associated with a mutation in mitochondrial DNA" generally means that the etiology of the mitochondrial disease or disorder is at least partially operably linked to a mutation or set of mutations in the coding region in mitochondrial DNA that encodes for the mitochondrial molecule.

In certain embodiments, the mitochondrial disease or disorder associated with a mutation in mitochondrial DNA is selected from Pearson Syndrome (PS), Kearns-Sayre syndrome (KSS), mitochondrial encephalopathy lactic acidosis and stroke-like attack (MELAS) syndrome, Leber's Hereditary Optic Neuropathy (LHON), neuropathy, ataxia and retinitis pigmentosa (NARP) syndrome, myoclonic epilepsy with hirsute-like red fiber (MERRF) syndrome, maternal hereditary diabetes and deafness (MIDD), Alpers-like syndrome, Chronic Progressive External Ophthalmoplegia (CPEO), mitochondrial DNA-related forms of Congenital Lactic Acidosis (CLA), Mitochondrial DNA Depletion Syndrome (MDDS), and mitochondrial DNA-related forms of Leigh syndrome. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the primary mitochondrial disease or disorder is associated with a mutation in nuclear DNA. As used herein, the phrase "associated with a mutation in nuclear DNA" generally means that the cause of the mitochondrial disease or disorder is at least partially operably linked to a mutation or set of mutations in the coding region in nuclear DNA that encodes for a mitochondrial molecule. The term "mitochondrial molecule" generally refers to any molecule that is delivered to and/or active in and/or present in healthy functional mitochondria. These molecules may be nucleic acid molecules, protein molecules, enzyme molecules, etc. In certain embodiments, the primary mitochondrial disease or disorder is associated with a mutation in a gene of nuclear DNA that directly encodes an OXPHOS protein or indirectly affects OXPHOS function by affecting the generation of complex mechanisms required to operate the OXPHOS process.

In certain embodiments, the primary mitochondrial disease or disorder associated with a mutation in nuclear DNA is selected from the group consisting of mitochondrial neurogastrointestinal encephalopathy (MNGIE) syndrome, Alpers syndrome, Friedrich's Ataxia (FA), progressive extraocular muscle Paralysis (PEO), sideroblasts anemia, Ataxia Neuropathy Syndrome (ANS), mendelian neurodegenerative mitochondrial disease, 3-methylpentene-dionuria (MEG) with deafness (D), encephalopathy (E), and Leigh-like disease (L) (MEGDEL) syndrome, Sengers syndrome, mini-morbid nephropathy syndrome (MCNS), nuclear DNA-related forms of Congenital Lactic Acidosis (CLA), nuclear DNA-depleted syndrome (MDDS), and nuclear DNA-related forms of Leigh syndrome. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the ANS is selected from the group consisting of mitochondrial recessive ataxia syndrome (MIRAS), spinocerebellar ataxia with epilepsy (SCAE), sensorineural neuropathy dysarthria and ophthalmoplegia (SANDO) and myoclonic epilepsy, myopathic sensory ataxia (mema). Each possibility represents a separate embodiment of the invention.

In certain embodiments, the mitochondrial disease or disorder is associated with acquired mitochondrial dysfunction. As used herein, the phrase "associated with acquired mitochondrial dysfunction" generally means that the mitochondrial disease or disorder becomes symptomatic over adulthood, worsens over time, and/or is not necessarily operably linked to a mutation or group of mutations in the coding region encoding the mitochondrial molecule in mitochondrial or nuclear DNA.

In certain embodiments, the mitochondrial disease or disorder associated with acquired mitochondrial dysfunction becomes symptomatic during adulthood. In certain embodiments, the mitochondrial disease or disorder associated with acquired mitochondrial dysfunction is exacerbated over time. In certain embodiments, the mitochondrial disease or disorder associated with acquired mitochondrial dysfunction is not operably linked or only partially linked to a mutation or set of mutations in the coding region in mitochondrial or nuclear DNA that encodes for a mitochondrial molecule. In certain embodiments, the patient is an adult. In certain embodiments, the patient is over 20 years of age. In certain embodiments, the patient is over 30, 40, 50, 60, or 70 years of age.

In certain embodiments, the mitochondrial disease or disorder is associated with an organ selected from kidney, liver, brain, muscle, pancreas, eye, and any combination thereof. In certain embodiments, the mitochondrial disease or disorder is associated with the kidney. In certain embodiments, the mitochondrial disease or disorder is associated with the liver. In certain embodiments, the mitochondrial disease or disorder is associated with the brain. In certain embodiments, the mitochondrial disease or disorder is associated with muscle. In certain embodiments, the mitochondrial disease or disorder is associated with the heart. In certain embodiments, the mitochondrial disease or disorder is associated with the pancreas. In certain embodiments, the mitochondrial disease or disorder is associated with the eye.

In certain embodiments, the symptom is selected from the group consisting of impaired walking ability, impaired motor skills, impaired language skills, impaired memory, impaired weight gain, growth retardation, low blood alkaline phosphatase levels, low blood magnesium levels, high blood creatinine levels, low blood bicarbonate levels, low blood alkaloid excess levels, high urine glucose/creatinine ratios, high urine chloride/creatinine ratios, high urine sodium/creatinine ratios, high blood lactate levels, high urine magnesium/creatinine ratios, high urine potassium/creatinine ratios, high urine calcium/creatinine ratios, diabetes, magnesium urine, high blood urea levels, low C-peptide levels, high HbA1C levels, hypoparathyroidism, ptosis, hearing loss, cardiac conduction disorders, low ATP content and low oxygen consumption in lymphocytes. Each possibility represents a separate embodiment of the invention. It is understood that the definition of a symptom as "high" and "low" corresponds to "detectably higher than normal" and "detectably lower than normal", respectively, wherein the normal level is the corresponding level in a plurality of subjects not suffering from a mitochondrial disorder.

In certain embodiments, the pharmaceutical composition is administered to a specific tissue or organ. In certain embodiments, the pharmaceutical composition comprises at least 104Individual mitochondrially enriched human stem cells. In certain embodiments, the pharmaceutical composition comprises about 104To about 108Individual mitochondrially enriched human stem cells.

In certain embodiments, the pharmaceutical composition is administered by parenteral administration. In certain embodiments, the pharmaceutical composition is administered by systemic administration. In certain embodiments, the pharmaceutical composition is administered by intravenous injection. In certain embodiments, the pharmaceutical composition is administered by intravenous infusion. In certain embodiments, the pharmaceutical composition comprises at least 105Individual mitochondrially enriched human stem cells. In certain embodiments, the pharmaceutical composition comprises about 106To about 108Individual mitochondrially enriched human stem cells. In certain embodiments, the medicament isThe composition comprises at least about 105-2×107Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises at least about 105Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises about 105To about 2X107Individual mitochondrially enriched human stem cells per kilogram patient body weight. In certain embodiments, the pharmaceutical composition comprises about 106To about 5X106Individual mitochondrially enriched human stem cells per kilogram patient body weight.

In certain embodiments, the human stem cells are obtained or derived from the patient prior to enrichment. In further embodiments, the human stem cells obtained or derived from the patient prior to enrichment have (i) a sub-normal oxygen (O2) consumption rate; (ii) a sub-normal level or activity of citrate synthase; (iii) a sub-normal rate of Adenosine Triphosphate (ATP) production; or (iv) any combination of (i), (ii), and (iii).

In certain embodiments, the mitochondrially enriched human stem cells are obtained or derived from a donor different from the patient prior to enrichment.

In certain embodiments, the donor is matched to at least a portion of a Human Leukocyte Antigen (HLA) of the patient. In certain embodiments, the above methods further comprise the step of administering to the patient an agent that prevents, delays, minimizes or abrogates an adverse immunogenic response between the patient and the mitochondrially enriched human stem cells. Each possibility represents a separate embodiment of the invention. In certain embodiments, the adverse immunogenic response is graft versus host disease (GvHD).

In certain embodiments, the human stem cell is CD34+. In certain embodiments, the human stem cells are hematopoietic stem cells. In certain embodiments, the human stem cell is a mesenchymal stem cell. In certain embodiments, the human stem cell is a Pluripotent Stem Cell (PSC) or an Induced Pluripotent Stem Cell (iPSC). The term "Pluripotent Stem Cell (PSC)" as used herein refers toCells that are capable of multiplying indefinitely and producing a wide variety of cell types in the body. As used herein, the term "induced pluripotent stem cell (iPSc)" refers to a class of pluripotent stem cells that can be generated from human adult somatic cells. In certain embodiments, the PSC is a non-embryonic stem cell. In certain embodiments, it is expressly understood that human embryonic stem cells are expressly excluded from the scope of the claims. As used herein, the term "Embryonic Stem Cell (ESC)" refers to a type of totipotent stem cell derived from the internal cell mass of the blastocyst. Totipotent stem cells are cells that can give rise to every other cell type in the body.

As used herein, the term "CD 34+Cells "refer to stem cells characterized as being positive for CD34, regardless of their origin. The term also refers to hematopoietic stem cells characterized as being CD34 positive, obtained from stem cells or mobilized from the bone marrow or obtained from cord blood. As used herein, the term "CD 34+ cells" refers to cells that express the surface marker protein CD 34. Expression of CD34 can be determined by immunofluorescence analysis or FACS analysis using antibodies against CD 34.

In certain embodiments, the CD34+ cells are umbilical cord cells. In certain embodiments, the CD34+ cells are bone marrow cells. In certain embodiments, the CD34+ cells are hematopoietic cells. In certain embodiments, the CD34+ cells are mesenchymal stem cells. In certain embodiments, the CD34+ cells are endothelial progenitor cells. In certain embodiments, the CD34+ cells are endothelial cells of blood vessels. In certain embodiments, the CD34+ cell is a mast cell. In certain embodiments, the CD34+ cells are a sub-population of dendritic cells (which are factor XIIIa negative). In certain embodiments, the CD34+ cells are long term hematopoietic stem cells (LT-HSCs). In certain embodiments, the CD34+ cells are human HSC cells. In certain embodiments, the CD34+ cells are allogeneic to the patient, wherein the CD34+ cells are HLA matched to the patient. In certain embodiments, the CD34+ cells are HLA matched to the patient. In certain embodiments, the CD34+ cells are autologous to the patient.

In certain embodiments, the mitochondrially enriched human stem cell is obtained by introducing freeze-thawed, healthy, functional, exogenous mitochondria into the human stem cell. In certain embodiments, the above methods further comprise the preliminary step of isolating, deriving or obtaining a human stem cell and introducing healthy, functional exogenous mitochondria into the human stem cell, thereby producing a mitochondria-enriched human stem cell. In certain embodiments, the above method further comprises the step of selecting CD34 positive cells from the healthy, functional, exogenous mitochondria prior to introducing the cells into the human stem cells. Selection of CD34 positive cells can be performed by methods known in the art including, but not limited to, CliniMACS or Prodigy systems (Miltenyi).

In certain embodiments, the human stem cell has undergone at least one freeze-thaw cycle prior to introducing the freeze-thawed, healthy, functional exogenous mitochondria into the human stem cell. In certain embodiments, the method comprises: (a) freezing the human stem cells, (b) thawing the human stem cells, and (c) introducing healthy, functional, exogenous mitochondria into the human stem cells.

In certain embodiments, the method further comprises expanding the stem cells before or after enriching the healthy, functional, exogenous mitochondria. In certain embodiments, the method further comprises expanding the cells by culturing the stem cells in the first composition in a medium capable of expanding stem cells or a proliferation medium. In other embodiments, the method further comprises expanding the cells by culturing the mitochondrially enriched stem cells in the fourth composition in a medium capable of expanding stem cells or a proliferation medium. The term "culture or propagation medium" as used throughout this application is a fluid medium such as cell culture medium, cell growth medium, buffer that provides nutrients to the cells. The term "pharmaceutical composition" as used throughout the present application and claims encompasses fluid carriers such as cell culture media, cell growth media, buffers that provide nutrients to the cells.

The possibility of freezing human stem cells prior to enrichment with healthy functional exogenous mitochondria is advantageous in accordance with the principles of the present invention because it provides, for example, sufficient time to test certain attributes of the human stem cells prior to enrichment with healthy functional exogenous mitochondria and/or improves the shelf life of the human stem cells and/or allows for easy distribution of the human stem cells.

The possibility of freezing human stem cells after enrichment of healthy functional exogenous mitochondria is advantageous according to the principles of the present invention, because it provides, for example, sufficient time to test certain properties of the enriched human stem cells after enrichment of healthy functional exogenous mitochondria and/or increases the shelf life of the enriched human stem cells and/or allows for easy distribution of the enriched human stem cells.

In certain embodiments, the human stem cells are isolated, derived, or obtained from cells of bone marrow, adipose tissue, oral mucosa, skin fibroblasts, blood, or umbilical cord blood. Each possibility represents a separate embodiment of the invention. In certain embodiments, the human stem cells are not isolated, derived, or obtained from bone marrow or cells of bone marrow.

In certain embodiments, the above methods further comprise the preliminary step of isolating or obtaining healthy functional exogenous mitochondria from a suitable source and introducing the healthy functional exogenous mitochondria into a human stem cell, thereby producing a mitochondria-enriched human stem cell. In certain embodiments, the method comprises: (a) freezing the healthy functional exogenous mitochondria, (b) thawing the healthy functional exogenous mitochondria, and (c) introducing the healthy functional exogenous mitochondria into the human stem cell.

In accordance with the principles of the present invention, the possibility of freezing healthy functional exogenous mitochondria prior to enriching the human stem cells is critical to the process of mitochondria enhancement therapy because it provides, for example, sufficient time to test the function and/or certain attributes of the healthy functional exogenous mitochondria prior to enriching the human stem cells, as well as improving the shelf life of the healthy functional exogenous mitochondria and/or allowing for easy distribution of the healthy functional exogenous mitochondria.

Without wishing to be bound by any theory or mechanism, mitochondria that have undergone a freeze-thaw cycle exhibit a comparable rate of oxygen consumption after thawing as compared to control mitochondria that have not undergone a freeze-thaw cycle.

According to certain embodiments, the freeze-thaw cycle comprises freezing the functional mitochondria for at least 24 hours prior to thawing. According to other embodiments, the freeze-thaw cycle comprises freezing the functional mitochondria for at least 1 month, several months, or longer prior to thawing. Each possibility represents a separate embodiment of the invention. According to another embodiment, the oxygen consumption of the functional mitochondria after the freeze-thaw cycle is equal to or higher than the oxygen consumption of the functional mitochondria prior to the freeze-thaw cycle.

As used herein, the term "freeze-thaw cycle" refers to freezing the functional mitochondria to a temperature below 0 ℃, maintaining the mitochondria at a temperature below 0 ℃ for a defined period of time, and thawing the mitochondria to room or body temperature or any temperature above 0 ℃ that is capable of treating the stem cells with the mitochondria. Each possibility represents a separate embodiment of the invention. As used herein, the term "room temperature" generally refers to a temperature between 18 ℃ and 25 ℃. As used herein, the term "body temperature" refers to a temperature between 35.5 ℃ and 37.5 ℃, preferably 37 ℃. In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are functional mitochondria.

In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are frozen at a temperature of-70 ℃ or less. In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are frozen at a temperature of-20 ℃ or less. In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are frozen at a temperature of-4 ℃ or less. According to another embodiment, the freezing of the mitochondria is gradual. According to some embodiments, the freezing of mitochondria is performed by flash freezing. As used herein, the term "flash freezing" refers to the rapid freezing of the mitochondria by subjecting them to ultra-low temperatures.

In another embodiment, the mitochondria undergoing a freeze-thaw cycle are frozen for at least 30 minutes prior to thawing. According to another embodiment, the freeze-thaw cycle includes freezing the functional mitochondria for at least 30, 60, 90, 120, 180, 210 minutes prior to thawing. Each possibility represents a separate embodiment of the invention. In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are frozen for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 24, 48, 72, 96, or 120 hours prior to thawing. Each freezing time represents a separate embodiment of the present invention. In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are frozen for at least 4, 5, 6, 7, 30, 60, 120, 365 days prior to thawing. Each freezing time represents a separate embodiment of the present invention. According to another embodiment, the freeze-thaw cycle includes freezing the functional mitochondria for at least 1, 2, 3 weeks prior to thawing. Each possibility represents a separate embodiment of the invention. According to another embodiment, the freeze-thaw cycle comprises freezing the functional mitochondria for at least 1, 2, 3, 4, 5, 6 months prior to thawing. Each possibility represents a separate embodiment of the invention.

In another embodiment, the mitochondria that have undergone a freeze-thaw cycle are frozen at-70 ℃ for at least 30 minutes prior to thawing. Without wishing to be bound by any theory or mechanism, the possibility of freezing mitochondria and thawing them after a long period of time enables easy storage and use of the mitochondria and reproducible results even after long storage times.

According to one embodiment, the thawing is performed at room temperature. In another embodiment, the thawing is performed at body temperature. According to another embodiment, the thawing is performed at a temperature that enables administration of said mitochondria according to the method of the invention. According to another embodiment, the thawing is performed gradually.

According to another embodiment, the mitochondria undergoing a freeze-thaw cycle are frozen in a freezing buffer. According to another embodiment, the mitochondria undergoing a freeze-thaw cycle are frozen in a separation buffer. As used herein, the term "isolation buffer" refers to the buffer in which the mitochondria of the invention are isolated. In a non-limiting example, the separation buffer is a sucrose buffer. Without wishing to be bound by any mechanism or theory, freezing mitochondria in the isolation buffer saves time and isolation steps, as there is no need to replace the isolation buffer with a freezing buffer prior to freezing or replace the freezing buffer after thawing.

According to another embodiment, the freezing buffer comprises a cryoprotectant. According to some embodiments, the cryoprotectant is a sugar, an oligosaccharide or a polysaccharide. Each possibility represents a separate embodiment of the invention. According to another embodiment, the sugar concentration in the freezing buffer is a sugar concentration sufficient for protecting mitochondrial function. According to another embodiment, the separation buffer comprises a sugar. According to another embodiment, the sugar concentration in the separation buffer is a sugar concentration sufficient for protecting mitochondrial function. According to another embodiment, the sugar is sucrose.

In certain embodiments, the healthy functional exogenous mitochondria are isolated or obtained from a placenta, placental cells grown in culture, or blood cells. Each possibility represents a separate embodiment of the invention.

According to certain aspects, the present invention provides a method of treating a primary mitochondrial disease, disorder or symptom thereof in a human patient in need thereof, the method comprising administering to the human patient a pharmaceutical composition comprising a plurality of human CD34+A step of administering a pharmaceutical composition of stem cells to said patient, wherein said human CD34+The stem cells are enriched in healthy functional exogenous mitochondria that are freeze-thawed, free of pathogenic mutations in mitochondrial DNA, and wherein the healthy functional exogenous mitochondria comprise mitochondrially enriched human CD34+At least 3% and less than 33% of total mitochondria in the stem cell.

In another aspect, the present invention also provides an ex vivo method of enriching human stem cells for healthy, functional, exogenous mitochondria, the method comprising the steps of: (i) providing a first composition comprising a plurality of human stem cells from a patient having a primary mitochondrial disease, disorder or symptom thereof; (ii) providing a second composition comprising a plurality of isolated healthy functional exogenous mitochondria obtained from a donor that does not have a pathogenic mutation in mitochondrial DNA; (iii) contacting the human stem cells of the first composition with healthy, functional, exogenous mitochondria of the second composition, thereby providing a third composition; and (iv) incubating the third composition under conditions that allow the healthy functional exogenous mitochondria to enter the human stem cell, thereby enriching the human stem cell for the healthy functional exogenous mitochondria, thereby providing a fourth composition comprising a mitochondria-enriched human stem cell; wherein the healthy functional exogenous mitochondria comprise at least 3% and less than 33% of the total mitochondria of the fourth composition.

As used herein, the term "ex vivo method" refers to any method that includes steps that are performed exclusively outside the human body. In particular, ex vivo methods involve manipulation of cells in vitro, which are subsequently reintroduced or transplanted into the subject to be treated.

The term "contacting" refers to bringing the composition of mitochondria and cells into sufficient proximity to facilitate entry of the mitochondria into the cells. The term "introducing" mitochondria into a stem cell can be used interchangeably with the term "contacting".

According to some embodiments, the method of enriching a human stem cell for healthy, functional, exogenous mitochondria does not comprise measuring the membrane potential of the cell.

In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.044 up to 176 milliunits CS activity per million cells into the stem cell. In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.088 up to 176 milliunits CS activity per million cells into the stem cell. In other embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.2 up to 150 milliunits CS activity per million cells into the stem cell. In other embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.4 up to 100 milliunits CS activity per million cells into the stem cell. In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.6 up to 80 milliunits CS activity per million cells into the stem cell. In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.7 up to 50 milliunits CS activity per million cells into the stem cell. In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.8 up to 20 milliunits CS activity per million cells into the stem cell. In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.88 up to 17.6 milliunits CS activity per million cells into the stem cell. In certain embodiments, enriching the stem cell for healthy functional exogenous mitochondria comprises introducing a dose of mitochondria of at least 0.9 up to 15 milliunits CS activity per million cells into the stem cell.

Mitochondrial dose can be expressed in terms of units of CS activity or mtDNA copy number or other quantifiable measure of the amount of healthy, functional mitochondria as explained herein. "units of CS activity" is defined as the amount of substrate that is capable of converting 1 micromole in 1 minute in a 1mL reaction volume.

In another aspect, the invention also provides a method of treating a primary mitochondrial disease or disorder or symptoms thereof in a human patient in need thereof, comprising the step of administering to the patient a pharmaceutical composition comprising a mitochondrially enriched human stem cell as described above.

As used herein, the phrase "stem cells obtained from a patient having a mitochondrial disease" refers to cells that, when isolated from the patient, are stem cells in the patient.

As used herein, the phrase "stem cells derived from a patient having a mitochondrial disease" refers to cells that are not stem cells in the patient and have been manipulated to become stem cells. The phrase also includes stem cells of a certain type that have been manipulated to become stem cells of a different type. As used herein, the term "manipulation" refers to reprogramming somatic cells to an undifferentiated state and into induced pluripotent stem cells (iPSc) using any of the methods known in the art (Yu j. et al, Science,2007, vol.318(5858), pp 1917-1920), and optionally further reprogramming the iPSc to become cells of a desired lineage or population (Chen m. et al, IOVS,2010, vol.51(11), pp 5970-5978), such as bone marrow cells (Xu y. et al, 2012, PLoS ONE, vol.7(4), pp e 34321).

As used herein, the term "patient with a mitochondrial disorder" refers to a human subject diagnosed as having, suspected of having, or in a risk group for developing a mitochondrial disorder. Since certain mitochondrial diseases are inherited, genetic carriers of mitochondrial diseases or offspring of subjects diagnosed with mitochondrial diseases are considered as risk groups for developing mitochondrial diseases.

As used herein, the term "subject/donor not suffering from a mitochondrial disorder" refers to a human subject that has not been diagnosed as suffering from a mitochondrial disorder, is not suspected of having a mitochondrial disorder, and/or is not in a risk group for developing a mitochondrial disorder. The term also includes subjects without mutations in mitochondrial DNA and/or subjects without mutations in nuclear DNA encoding molecules (e.g., proteins or RNA molecules) that are transferred to mitochondria.

As used herein, the term "isolated healthy functional human exo-mitochondria" refers to intact mitochondria obtained or derived from cells obtained from a subject not suffering from a mitochondrial disease. The term also includes functional mitochondria obtained from subjects without mutations in mitochondrial DNA. In certain embodiments, the mitochondria are exogenous mitochondria. The term "isolated" when used herein and in the claims in the context of mitochondria includes mitochondria that are at least partially purified from other components present in the source. In certain embodiments, the total amount of mitochondrial proteins in the second composition comprising a plurality of isolated healthy, functional, exogenous mitochondria is between 20% -80%, 20-70%, 40-70%, 20-40%, or 20-30% of the total amount of cellular proteins in the sample. Each possibility represents a separate embodiment of the invention. In certain embodiments, the total amount of mitochondrial proteins in the second composition comprising a plurality of isolated healthy, functional, exogenous mitochondria is between 20% and 80% of the total amount of cellular proteins in the sample. In certain embodiments, the total amount of mitochondrial proteins in the second composition comprising a plurality of isolated healthy, functional, exogenous mitochondria is between 20% and 80% of the combined weight of the mitochondria and other subcellular fractions. In other embodiments, the total amount of mitochondrial proteins in the second composition comprising a plurality of isolated healthy functional exogenous mitochondria is greater than 80% of the combined weight of the mitochondria and other subcellular fractions.

As used herein, the phrase "conditions that allow the human functional mitochondria to enter the human stem cells" generally refers to parameters such as time, temperature, and proximity between the mitochondria and the human stem cells. These conditions are provided by the present invention.

In certain embodiments, the human stem cells are incubated with the healthy, functional, exogenous mitochondria at a temperature in the range of about 16 to about 37 ℃ for a time in the range of 0.5 to 30 hours. In certain embodiments, the human stem cells are incubated with the healthy, functional, exogenous mitochondria for a time in the range of 1 to 30 or 5 to 25 hours. Each possibility represents a separate embodiment of the invention. In a particular embodiment, the incubation is performed for 20 to 30 hours. In certain embodiments, the incubation is performed for at least 1,5, 10, 15, or 20 hours. Each possibility represents a separate embodiment of the invention. In other embodimentsWherein the incubation is carried out for up to 5, 10, 15, 20 or 30 hours. Each possibility represents a separate embodiment of the invention. In a particular embodiment, the incubation is performed for 24 hours. In certain embodiments, the incubation is performed at room temperature (16 ℃ to 30 ℃). In other embodiments, the incubation is performed at 37 ℃. In certain embodiments, the incubation is at 5% CO2Is carried out in an atmosphere. In other embodiments, the incubating does not include adding CO above the level present in air2. In certain embodiments, the incubation is carried out until the mitochondrial content in the stem cells is increased by an average of 1% to 45% compared to their initial mitochondrial content.

By manipulating the conditions of the incubation, one can manipulate the characteristics of the product. In certain embodiments, the incubation is performed at 37 ℃. In certain embodiments, the incubation is performed for at least 6 hours. In certain embodiments, the incubation is performed for at least 12 hours. In certain embodiments, the incubation is performed for 12 to 24 hours. In certain embodiments, the incubation is 1x10 in an amount of exogenous mitochondria that have or exhibit 4.4 milliunits CS per serving5To 1X107The proportion of primary stem cells. In certain embodiments, the incubation is 1x10 in an amount of exogenous mitochondria that have or exhibit 4.4 milliunits CS per serving6The proportion of primary stem cells. In certain embodiments, the conditions are sufficient to increase mitochondrial content of the naive stem cell as determined by CS activity by at least about 3%, 5%, or 10%. Each possibility represents a separate embodiment of the invention.

As used herein, the term "mitochondrial content" refers to the amount of mitochondria within a cell. In certain embodiments, the incubation is performed in a medium that supports survival of the cells. In certain embodiments, the medium is Dulbecco's Modified Eagle Medium (DMEM). In other embodiments, the medium is saline containing HSA (human serum albumin). In certain embodiments, the brine contains between 1% and 10% HSA. In further embodiments, the brine contains between 3 and 6% HSA. In still other embodiments, the brineContains 4.5% HSA. In certain embodiments, the incubation of the stem cells with the healthy functional mitochondria is at a temperature in the range of 16 to 30 ℃, in saline containing between 3 to 6% HSA, without the addition of CO above the level present in air2For a time in the range of 15 to 30 hours.

In certain embodiments, the methods described above, in various embodiments thereof, further comprise centrifuging before, during, or after incubating the stem cells with the exogenous mitochondria. Each possibility represents a separate embodiment of the invention. In certain embodiments, the methods described above, in various embodiments thereof, comprise performing a single centrifugation step before, during, or after incubating the stem cells with the exogenous mitochondria. In certain embodiments, the centrifugal force is in the range of 1000g to 8500 g. In certain embodiments, the centrifugal force is in the range of 2000g to 4000 g. In certain embodiments, the centrifuge force is greater than 2500 g. In certain embodiments, the centrifugal force is in the range of 2500g to 8500 g. In certain embodiments, the centrifugal force is in the range of 2500g to 8000 g. In certain embodiments, the centrifugal force is in the range of 3000g to 8000 g. In other embodiments, the centrifugal force is in the range of 4000g to 8000 g. In a particular embodiment, the centrifugal force is 7000 g. In other embodiments, the centrifugal force is 8000 g. In certain embodiments, centrifugation is performed for a time in the range of 2 minutes to 30 minutes. In certain embodiments, centrifugation is performed for a time in the range of 3 minutes to 25 minutes. In certain embodiments, centrifugation is performed for a time in the range of 5 minutes to 20 minutes. In certain embodiments, centrifugation is performed for a time in the range of 8 minutes to 15 minutes.

In certain embodiments, centrifugation is performed at a temperature in the range of 4 to 37 ℃. In certain embodiments, centrifugation is performed at a temperature in the range of 4 to 10 ℃ or 16-30 ℃. Each possibility represents a separate embodiment of the invention. In a particular embodiment, centrifugation is performed at 2-6 ℃. In a particular embodiment, centrifugation is performed at 4 ℃. In certain embodiments, the methods described above, in various embodiments thereof, comprise a single centrifugation before, during, or after incubating the stem cells with the exogenous mitochondria, followed by resting the cells at a temperature below 30 ℃. In certain embodiments, the conditions that allow the human functional mitochondria to enter the human stem cell comprise a single centrifugation before, during, or after incubating the stem cell with the exogenous mitochondria, followed by resting the cell at a temperature in the range of 16 to 28 ℃.

In certain embodiments, the first composition is fresh. In certain embodiments, the first composition is frozen and then thawed prior to incubation. In certain embodiments, the second composition is fresh. In certain embodiments, the second composition is frozen and then thawed prior to incubation. In certain embodiments, the fourth composition is fresh. In certain embodiments, the fourth composition is frozen and then thawed prior to administration.

In certain embodiments, the stem cells in the fourth composition have (i) increased mitochondrial DNA content as compared to the stem cells in the first composition; (ii) increased content of at least one mitochondrial protein selected from CS, COX1 and SDHA; (iii) enhanced oxygen (O)2) A consumption rate; (iv) increased level of citrate synthase activity; (v) increased rate of Adenosine Triphosphate (ATP) production; or (vi) any combination of (i), (ii), (iii), (iv), and (v). Each possibility represents a separate embodiment of the invention. Methods for determining these various parameters are well known in the art.

As used herein, the term "increased mitochondrial DNA content" refers to a mitochondrial DNA content that is detectably higher than the mitochondrial DNA content in the first composition prior to mitochondrial enrichment. Mitochondrial DNA content can be measured by quantitative PCR of mitochondrial genes before and after mitochondrial enrichment and normalization to nuclear genes.

As used herein, the term "increased level of at least one mitochondrial protein" refers to a level of nuclear-or mitochondria-encoded mitochondrial proteins, such as CS, COX1, and SDHA, that is detectably higher than the level of the mitochondrial protein in the first composition prior to mitochondrial enrichment.

As used herein, the term "elevated oxygen (O)2) By consumption rate "is meant a rate detectably higher than the oxygen (O) in the first composition prior to mitochondrial enrichment2) Consumption rate of oxygen (O)2) The rate of consumption.

As used herein, the term "increased level or activity level of citrate synthase" refers to a level or activity of citrate synthase that is detectably higher than the level or activity of citrate synthase in the first composition prior to mitochondrial enrichment.

As used herein, the term "increased rate of Adenosine Triphosphate (ATP) production" refers to a rate of Adenosine Triphosphate (ATP) production that is detectably higher than the rate of Adenosine Triphosphate (ATP) production in the first composition prior to mitochondrial enrichment.

In certain embodiments, the term "detectably higher" as used herein refers to a statistically significant increase between the stated normal and increased values. In certain embodiments, the term "detectably higher" as used herein refers to a non-pathological increase, i.e., to a level where no pathological symptoms associated with the substantially higher values become apparent. In certain embodiments, the term "increased" as used herein refers to a value that is 1.05-fold, 1.1-fold, 1.25-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold or higher than the corresponding value present in the corresponding cells or corresponding mitochondria of a plurality of healthy subjects or in the stem cells of the first composition prior to mitochondrial enrichment. Each possibility represents a separate embodiment of the invention.

In particular cases, the same cells prior to mitochondrial enrichment were used as controls to measure CS and ATP activity and determine the level of enrichment.

Citrate Synthase (CS) is located in the mitochondrial matrix but is encoded by nuclear DNA. Citrate synthase participates in the first step of the krebs cycle and is commonly used as a quantitative enzymatic marker for the presence of intact mitochondria (Larsen s. et al, 2012, j. physiol., vol.590(14), p.3349-3360; Cook g. a. et al, biochim. biophysis. acta, 1983, vol.763(4), p.356-367). In certain embodiments, the mitochondrial content of the stem cells in the first or fourth composition is determined by determining the level of citrate synthase. In certain embodiments, the mitochondrial content of the stem cells in the first or fourth composition is determined by determining the level of citrate synthase activity. In certain embodiments, the mitochondrial content of the stem cells in the first or fourth composition is related to the level of citrate synthase. In certain embodiments, the mitochondrial content of the stem cell in the first or fourth composition is correlated with the level of citrate synthase activity. CS activity can be measured, for example, using CS activity kit CS0720 (Sigma).

Eukaryotic NADPH-cytochrome C reductase (cytochrome C reductase) is a flavoprotein localized to the endoplasmic reticulum. It transfers electrons from NADPH to several oxygenases, the most important of which is the enzyme of the cytochrome P450 family responsible for xenobiotic detoxification. Cytochrome C reductase is widely used as an endoplasmic reticulum marker. In certain embodiments, the second composition is substantially free of cytochrome C reductase or cytochrome C reductase activity. In certain embodiments, the fourth composition is not enriched for cytochrome C reductase or cytochrome C reductase activity as compared to the first composition.

In certain embodiments, the stem cell comprises a myeloblast. As used herein, the term "myeloblasts" refers to cells involved in bone marrow formation, e.g., in the production of bone marrow and all cells produced therefrom, i.e., all blood cells.

In certain embodiments, the stem cells comprise erythropoietic cells. As used herein, the term "erythropoietic cell" refers to a cell that is involved in erythropoiesis, e.g., in the production of red blood cells (erythrocytes).

In certain embodiments, the stem cells comprise pluripotent Hematopoietic Stem Cells (HSCs). As used herein, the term "pluripotent hematopoietic stem cell" or "hematopoietic cell" refers to a stem cell that produces all other blood cells by the hematopoietic process.

In certain embodiments, the stem cells comprise common myeloid progenitor cells, common lymphoid progenitor cells, or any combination thereof. As used herein, the term "common myeloid progenitor cell" refers to a cell that produces myeloid cells. As used herein, the term "common lymphoid lineage progenitor" refers to a cell that produces lymphocytes.

In certain embodiments, the pharmaceutical composition further comprises megakaryocytes, erythrocytes, mast cells, myeloblasts, basophils, neutrophils, eosinophils, monocytes, macrophages, Natural Killer (NK) cells, small lymphocytes, T lymphocytes, B lymphocytes, plasma cells, reticulocytes, or any combination thereof. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the stem cell comprises a mesenchymal stem cell. As used herein, the term "mesenchymal stem cell" refers to a pluripotent stromal cell that can differentiate into a variety of different cell types including osteoblasts (osteocytes), chondrocytes, myocytes (myocytes), and adipocytes.

In certain embodiments, the stem cells are comprised of myeloblasts. In certain embodiments, the stem cells are comprised of erythropoietic cells. In certain embodiments, the stem cells are comprised of pluripotent Hematopoietic Stem Cells (HSCs). In certain embodiments, the stem cells are comprised of common myeloid progenitor cells, common lymphoid progenitor cells, or any combination thereof. Each possibility represents a separate embodiment of the invention. In certain embodiments, the stem cell is comprised of a mesenchymal stem cell.

The hematopoietic progenitor antigen, CD34, also known as the CD34 antigen, is a protein encoded by the CD34 gene in humans. CD34 is a cluster of differentiation antigens in cell surface glycoproteins and acts as a cell-cell adhesion factor. In certain embodiments, the stem cell expresses the myeloid progenitor antigen CD34 (which is CD 34)+Of (d). In certain embodiments, the stem cells present the myeloid progenitor antigen CD34 on their outer membrane. In certain embodiments, the stem cell does not express the myeloid progenitor antigen CD34 (is CD 34)-Of (d). In certain embodiments, the stem cells do not have the myeloid progenitor antigen CD34 on their outer membrane.

In certain embodiments, the stem cells in the first composition are derived directly from a patient having a mitochondrial disorder. In certain embodiments, the stem cells in the first composition are derived directly from a subject not suffering from a mitochondrial disorder. As used herein, the term "directly derived from" refers to stem cells that are directly derived from other cells. In certain embodiments, the stem cells are derived from hematopoietic stem cells.

In certain embodiments, the stem cells in the first composition are derived indirectly from a patient having a mitochondrial disorder. In certain embodiments, the stem cells in the first composition are derived indirectly from a subject not suffering from a mitochondrial disorder. As used herein, the term "indirectly derived from" refers to stem cells that are derived from non-stem cells or other types of stem cells. In certain embodiments, the stem cells are derived from somatic cells that are manipulated to become induced pluripotent stem cells (ipscs).

In certain embodiments, the stem cells in the first composition are obtained directly from bone marrow of a patient having a mitochondrial disorder. In certain embodiments, the stem cells in the first composition are obtained directly from bone marrow of a subject that has not suffered from a mitochondrial disorder. As used herein, the term "directly obtained" refers to stem cells obtained from the bone marrow itself, for example, by means such as surgery or sucking through a needle with a syringe.

In certain embodiments, the stem cells in the first composition are obtained indirectly from bone marrow of a patient having a mitochondrial disorder. In certain embodiments, the stem cells in the first composition are obtained indirectly from the bone marrow of a subject that has not suffered from a mitochondrial disorder. As used herein, the term "indirectly obtained" refers to stem cells obtained from a location other than the bone marrow itself.

In certain embodiments, the stem cells in the first composition are obtained directly or indirectly from adipose tissue, oral mucosa, skin fibroblasts, blood, and/or umbilical cord blood. Each possibility is a separate embodiment.

In certain embodiments, the stem cells in the first composition are obtained from peripheral blood of a patient having a mitochondrial disorder. In certain embodiments, the stem cells in the first composition are obtained from peripheral blood of a subject who has not suffered from a mitochondrial disorder. As used herein, the term "peripheral blood" refers to blood circulating in the blood system.

In certain embodiments, the above method further comprises a preliminary step comprising administering to the subject having a mitochondrial disorder an agent that induces mobilization of stem cells into peripheral blood. In certain embodiments, the above methods further comprise a preliminary step comprising administering to a subject not suffering from a mitochondrial disorder an agent that induces mobilization of stem cells into peripheral blood.

In certain embodiments, the agent that induces mobilization of stem cells into peripheral blood is selected from the group consisting of granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), 1' - [1, 4-phenylenebis (methylene) ] bis [1,4,8, 11-tetraazacyclotetradecane ] (plexafop, CAS No. 155148-31-5), salts thereof, and any combination thereof. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the above method further comprises the step of isolating the stem cells from peripheral blood of a patient having a mitochondrial disorder. In certain embodiments, the above method further comprises the step of isolating the stem cells from peripheral blood of a subject not suffering from a mitochondrial disorder. As used herein, the term "isolated from peripheral blood" refers to the separation of stem cells from the other constituents of blood.

During apheresis, the blood of a donor or patient is passed through a device that separates out one particular constituent and returns the remaining components to circulation. It is therefore a medical procedure that is performed outside the body. In certain embodiments, the separation is performed by apheresis.

In certain embodiments, the above method further comprises concentrating the stem cells and functional mitochondria in the third composition prior to incubation. In certain embodiments, the above method further comprises concentrating the stem cells and functional mitochondria in the third composition during the incubating. In certain embodiments, concentration of the stem cells and functional mitochondria in the third composition during incubation is performed by continuous centrifugation.

In certain embodiments, the above methods further comprise centrifuging the third composition prior to incubating. In certain embodiments, the above methods further comprise centrifuging the third composition during the incubating. In certain embodiments, the above methods further comprise centrifuging the third composition after incubation.

It should be emphasized that any reference to any measurable characteristic or condition for a plurality of cells or mitochondria is to the measurable average characteristic or condition of the plurality of cells or mitochondria.

Heterogeneity is the presence of more than one type of mitochondrial DNA within a cell or individual. The level of heterogeneity is the ratio of mutant mtDNA molecules relative to wild type/functional mtDNA molecules and is an important factor in considering the severity of mitochondrial disease. A lower level of heterogeneity (sufficient numbers of mitochondria are functional) is associated with a healthy phenotype, while a higher level of heterogeneity (insufficient numbers of mitochondria are functional) is associated with pathology. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 1% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 3% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 5% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 10% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 15% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 20% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 25% less than the level of heterogeneity of stem cells in the first composition. In certain embodiments, the level of heterogeneity of stem cells in the fourth composition is at least 30% less than the level of heterogeneity of stem cells in the first composition.

In certain embodiments, the method further comprises freezing the fourth composition. In certain embodiments, the method further comprises freezing and then thawing the fourth composition.

As used herein, the term "autologous cells" or "autologous cells" refers to the patient's own cells. The term "allogeneic cells" refers to cells from different donor individuals. The term "autologous mitochondria" refers to mitochondria obtained from the same maternal genetically related cells. The term "allogeneic mitochondria" refers to mitochondria from a different donor individual that is not maternally genetically related to the subject to be treated.

The term "syngeneic" as used herein and in the claims refers to genetic identity or genetic near identity sufficient to allow for transplantation between individuals without rejection. In the case of mitochondria, the term "syngeneic" is used interchangeably herein with the term "autologous mitochondria" meaning the same maternal blood system.

The term "exogenous mitochondria" refers to mitochondria that are introduced into a target cell (i.e., a stem cell) from a source outside the cell. For example, in certain embodiments, the exogenous mitochondria can be derived or isolated from a cell different from the target cell. For example, exogenous mitochondria can be produced/manufactured in a donor cell, purified/isolated/obtained from the donor cell, and subsequently introduced into the target cell.

The term "endogenous mitochondria" refers to mitochondria that are produced/expressed/produced by a cell and are not introduced into the cell from an external source. In certain embodiments, the endogenous mitochondria can contain proteins and/or other molecules encoded by the genome of the cell. In certain embodiments, the term "endogenous mitochondria" is equivalent to the term "host mitochondria".

In certain embodiments, the healthy functional human allogenic mitochondria are autologous or allogeneic mitochondria.

In certain embodiments, the identification/discrimination of the endogenous mitochondria from the exogenous mitochondria after they have been introduced into the target cell can be performed by a variety of different means, including, for example, but not limited to: identifying differences in mitochondrial dna (mtdna) sequence between the endogenous and exogenous mitochondria, e.g., a different haplotype, identifying a particular mitochondrial protein originating from the tissue of the exogenous mitochondria, e.g., cytochrome P450 cholesterol side chain cleavage (P450SCC) from placenta, UCP1 from brown adipose tissue, etc., or any combination thereof.

In certain embodiments, the above methods further comprise the step of administering to said patient an agent that promotes mitochondrial biogenesis. As used herein, the term "mitochondrial biogenesis" refers to the growth and division of mitochondria. In certain embodiments, the agent that promotes mitochondrial biogenesis is Erythropoietin (EPO) or a salt thereof. In certain embodiments, the pharmaceutical agent is selected from the group consisting of recombinant human erythropoietin and isolated human erythropoietin.

In certain embodiments, the above methods further comprise the step of administering to the patient an agent that prevents, delays, minimizes or abrogates an adverse immunogenic response between the patient and the stem cells. In certain embodiments, the adverse immunogenic response is graft versus host disease (GvHD). In certain embodiments, the GvHD is an acute form of the disease (aGvHD). In certain embodiments, the GvHD is a chronic form of the disease (cGvHD).

In certain embodiments, the above method further comprises the preliminary step of administering a pre-transplant conditioner to the patient prior to administering the pharmaceutical composition. As used herein, the term "pre-transplant conditioner" refers to any agent capable of killing stem cells in the human subject. In certain embodiments, the pre-transplant conditioner is busulfan.

As used herein, the term "mutation" refers to an insertion, deletion or substitution of at least one nucleotide in mitochondrial or nuclear DNA. In certain embodiments, the mutation is a pathological mutation.

In certain embodiments, the pharmaceutical composition is administered topically. In certain embodiments, administration of the pharmaceutical composition to a subject is via direct administration to the bone marrow of the subject. In certain embodiments, the administration of the pharmaceutical composition to a subject is to a tissue or organ. In certain embodiments, the administration of the pharmaceutical composition to a subject is to the eye. Vitreous humor is a transparent, colorless, gelatinous mass that fills the space between the lens and retina in the eye. In certain embodiments, the administration of the pharmaceutical composition to a subject is to the vitreous humor of the eye. In certain embodiments, administration of the pharmaceutical composition to a subject is by direct intramuscular injection. In certain embodiments, the pharmaceutical composition is administered systemically. In certain embodiments, administration of the pharmaceutical composition to a subject is by a route selected from intravenous, intraarterial, intramuscular, subcutaneous, and direct injection into a tissue or organ. Each possibility represents a separate embodiment of the invention.

In certain embodiments, the functional mitochondria are obtained from a human cell or human tissue selected from the group consisting of a placenta, placental cells grown in culture, and blood cells. Each possibility represents a separate embodiment of the invention.

According to another embodiment, the integrity of the mitochondrial membrane may be determined by any method known in the art. In a non-limiting example, integrity of mitochondrial membranes is measured using tetramethylrhodamine methyl ester (TMRM) or tetramethylrhodamine ethyl ester (TMRE) fluorescent probes. Each possibility represents a separate embodiment of the invention. The TMRM or TMRE stained mitochondria were observed under the microscope and shown to have an intact outer mitochondrial membrane. As used herein, the term "mitochondrial membrane" refers to a mitochondrial membrane selected from the group consisting of the inner mitochondrial membrane, the outer mitochondrial membrane, and both.

While the invention has been described with reference to certain embodiments, it will be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the scope of the invention. In addition, many modifications may be made to adapt a particular situation or material to the teachings of the invention without departing from its scope. Therefore, it is intended that the invention not be limited to the particular embodiment disclosed, but that the invention will include all embodiments falling within the scope of the appended claims.

The following examples are provided to provide a more complete understanding of the invention. The specific techniques, conditions, materials, proportions and reported data set forth to illustrate the principles of the invention are exemplary and should not be construed as limiting the scope of the invention.

Examples

+Example 1 mitochondrial Rapid entry into CD34 cells following MAT program

CD34 from healthy donors+Cells were treated with Mitotracker Orange (MTO) and washed, then MAT was performed using mitochondria isolated from HeLa-TurboGFP-mitochondrial cells (CellTrend GmbH). Cells were fixed with 2% PFA for 10 min and DAPI. The cells were scanned using a confocal microscope equipped with a 60X/1.42 oil immersion objective.

As can be seen in fig. 1, the exogenous mitochondria entered CD34 as soon as 0.5 hour after MAT+Cells (bright, almost white spots inside the cells) and continued entry at 8 and 24 hours tested.

Example 2 mitochondrial enhancement therapy in mice

Different mouse cells were cultured in DMEM containing 10% FCS at 37 ℃ and 5% CO2The isolated mitochondria were incubated for 24 hours under atmosphere in order to increase their mitochondrial content and activity. Table 1 depicts representative results of the mitochondrial enhancement process, as determined by the relative increase in CS activity of the cell after the process compared to the CS activity of the cell before the process.

Table 1.

FVB/N bone marrow cells (carrying a mutation in mtDNA ATP 8) were incubated with C57/BL Wild Type (WT) mitochondria isolated from placenta at various doses (0.044, 0.44, 0.88, 2.2, 4.4, 8.8, 17.6 milliunits of CS activity per 1M cells in 1 mL). As can be seen in fig. 2A, dPCR using WT specific sequences showed that WT mtDNA increased in a dose dependent manner for most doses. The enriched cells also showed dose-dependent increases in mtDNA-encoded (COX1) (fig. 2B) and nuclear-encoded (SDHA) (fig. 2C) protein content.

Mouse bone marrow cells (10)6) The GFP-labeled mitochondria (17milliU or 34milliU, indicating the level of citrate synthase activity as a marker of mitochondrial content) that were not treated or isolated from mouse myeloma cells were incubated for 24 hours. The cells were mixed with mitochondria, centrifuged at 8000g and resuspended. After 24 hours incubation, the cells were washed twice with PBS and the levels of Citrate Synthase (CS) activity (fig. 3A) and cytochrome c reductase activity (fig. 3B) were measured as previously described in WO 2016/135723 using CS0720 and CY0100 kits (Sigma), respectively.

The results demonstrated in figure 3 clearly demonstrate that the functional mitochondrial composition used in the above experiments enriched bone marrow cells with mitochondria but not ER.

To investigate the in vivo effect of mitochondrial-enhancing therapy, FVB/N bone marrow cells (1X 10) enriched with C57/BL placental mitochondria with 4.4 milliunits of CS activity6) IV was injected into FVB/N mice. Bone marrow was collected from mice 1 day, 1 week, 1 month and 3 months after the treatment and WT mtDNA levels were detected using dPCR. As can be seen in fig. 4, significant amounts of WT mtDNA were detected in bone marrow 1 day after treatment.

Example 3 MAT leads to the coexistence of exogenous and endogenous mtDNA within cells

Healthy donor CD34+Mitochondrial enhancement of cells was performed using two different placenta-derived mitochondrial batches and the cells were washed extensively after 24h incubation. Illumina-based sequencing of mtDNA revealed the presence of both metastatic and endogenous mitochondria within the same cell.

As can be seen in fig. 5, two MAT experiments from two different placentas produced similar percent enhancement.

Example 4 mitochondria can enter human bone marrow cells

Human CD34+Cells (1.4X 10)5ATCC PCS-800-012), either untreated or incubated with GFP-labeled mitochondria isolated from human placental cells for 20 hours. Mitochondria were mixed with the cells, centrifuged at 8000g and resuspended prior to plating the cells. After incubation, the cells were washed twice with PBS and CS activity was measured using CS0720 Sigma kit (fig. 6A). ATP content was measured as previously described in WO 2016/135723 using ATPLite (Perkin Elmer) (FIG. 6B).

The results demonstrated in figure 6 clearly show that by interacting and co-incubating with isolated human mitochondria, the mitochondrial content of human bone marrow cells can be increased many-fold, to an extent that exceeds the capacity of human or murine fibroblasts or murine bone marrow cells.

Example 5 bone marrow from NSGS mice engrafted with human umbilical cord blood contained more 2 months after MAT Human mtDNA

Incubating umbilical cord blood cells of a patient with Pearson syndrome with 0.88mU of human mitochondria for 24hr, removing the medium, washing and resuspending the cells in 4.5% HSAIn (1). The enriched cells IV were injected into NSGS mice (100,000 CD34 per mouse)+A cell).

Fig. 7A is a graphical illustration of mtDNA deletion in umbilical cord blood cells of a patient with pearson syndrome, showing the 4978kb deleted UCB mtDNA region (left panel), and showing southern blot analysis of the deletion (right panel).

Bone marrow was collected from mice 2 months after MAT and the copy number of WT mtDNA without deletion was analyzed in dPCR using primers and probes identifying WT mtDNA sequence without deletion of UCB.

As can be seen in fig. 7B, 2 months after mitochondrial potentiation therapy, the bone marrow of the mice contained-100% more human mtDNA than bone marrow of mice injected with non-enhanced cord blood cells.

Example 6 in vivo safety and biodistribution animal models

Mitochondria were introduced into bone marrow cells from two different backgrounds of control healthy mice: the source of mitochondria is from mice with different mtDNA sequences (Jenuth JP et al, Nature Genetics,1996, Vol.14, p. 146-.

Mitochondria were isolated from wild type mouse (C57BL) placenta. Bone marrow cells were isolated from FVB/N mice. The mutated FVB/N bone marrow cells (10)6) The healthy functional C57BL mitochondria (4.4 milliunits) were loaded and IV administered to FVB/N mice.

The method comprises the following steps: (1) mitochondria were isolated from the placenta of C57BL mice, frozen and thawed at-80 ℃, or used freshly; (2) obtaining bone marrow cells from mtDNA-mutated FVB/N mice; (3) contacting the mitochondria with bone marrow cells, centrifuging at 8000g for 5 minutes, resuspending and incubating for 24 hours; (4) the bone marrow cells were washed twice with PBS and injected into the tail vein of FVB/N mice. At various time points after transplantation, e.g., 24 hours, one week, one month, and 3 months, tissues (blood, bone marrow, lymphocytes, brain, heart, kidney, liver, lung, spleen, skeletal muscle, eye, ovary/testis) were collected and DNA was extracted for further sequence analysis.

The reduction in FVB/N levels in bone marrow 1 month post-transplantation is depicted in FIG. 8.

Example 7 juvenile onset of drug development with Pearson Syndrome (PS) and Kidney Vanconi syndrome (FS) +Homeopathic treatment of autologous CD34 cells of MNV-BLD (blood-derived mitochondria)

Patient 1 was a 6.5 year old male patient diagnosed with PS with a deletion at nucleotide 5835-9753 in their mtDNA. Before mitochondrial enhanced therapy (MAT), he was 14.5KG in weight and was unable to walk more than 100 meters or climb stairs. His growth was significantly delayed for 3 years prior to treatment, and his weight at baseline was-4.1 Standard Deviation Score (SDS), height was-3.2 SDS (relative to the population), and was not improved despite feeding through the gastrostomy tube (G-tube) for more than one year. He had renal failure (GFR 22ml/min) and proximal glomerulopathy, requiring electrolyte supplementation. He had hypoparathyroidism requiring calcium supplementation and had incomplete right bundle branch block (ICRBB) on the electrocardiogram.

Mobilization of Hematopoietic Stem and Progenitor Cells (HSPCs) was performed by subcutaneous administration of GCSF alone for 5 days. Leukocyte apheresis was performed through the peripheral venous interface using the Spectra Optia system (TerumoBCT) according to institutional guidelines. Mobilized peripheral blood-derived cells were positively selected for CD34 using CliniMACS CD34 reagent according to the manufacturer's instructions. Mitochondria were isolated from maternal Peripheral Blood Mononuclear Cells (PBMCs) by differential centrifugation using 250mM sucrose buffer, ph 7.4. For MAT, autologous CD34 was added+Cells were incubated with the healthy mitochondria from the patient's mother (mitochondria at 1X10 with an amount of 4.4 milliunits Citrate Synthase (CS) per serving6Individual cells) resulted in a 1.56-fold increase in the mitochondrial content of the cells (a 56% increase in mitochondrial content as evidenced by CS activity). Incubation with mitochondria was performed for 24 hours at r.t. in saline containing 4.5% HSA. The enriched cells were suspended in a saline solution containing 4.5% human serum albumin. 1.1X 10 infusion by time line IV as shown in FIG. 9A6Autologous CD34 enriched with healthy mitochondria+Cells per kilogram body weight, said patient receivingIs treated by a single round.

Figure 9B demonstrates the level of aerobic task Metabolic Equivalent (MET) score of the patients as a function of time after cell therapy. The data indicate that the aerobic MET score of the patients increased significantly over time, from 5 (moderate intensity activities such as walking and cycling) to 8 (high intensity activities such as running, jogging and skipping).

Figure 9C illustrates lactate levels found in the blood of the patient as a function of time after IV injection. Blood lactate, which is lactic acid that appears in the blood as a result of anaerobic metabolism at the time of mitochondrial damage or when the delivery of oxygen to tissues is insufficient to support normal metabolic demand, is one of the hallmarks of mitochondrial dysfunction. As can be seen in fig. 4C, patient 1 had decreased blood lactate levels to normal after MAT.

Table 2 shows the change in the pediatric mitochondrial disease scale (IPMDS) -quality of life (QoL) questionnaire results of the patients over time after cell therapy. In both categories, "complaints and symptoms" and "physical examination", 0 means the relevant attribute "normal", and the aggravated condition is scored 1 to 5 according to severity.

Table 2.

Before treatment +6 months
Complaints and symptoms 24 11
Physical examination 13.4 4.6

It should be noted that the patient did not gain weight in the 3 years prior to treatment, i.e. there was no weight gain since the age of 3.5 years. The data presented in figure 4D shows growth measured by standard deviation scores of the patient's weight and height, starting 4 years before MAT and covering the follow-up period. The data indicate that there is an increase in the weight and height of the patient at about 9 months or 15 months after a single treatment, respectively.

Another evidence of growth in the patient is from his alkaline phosphatase level. The alkaline phosphatase level test (ALP test) measures the amount of alkaline phosphatase in the blood stream. Having a lower than normal level of ALP in the blood may indicate malnutrition, which may be caused by a deficiency of certain vitamins and minerals. The data presented in fig. 9E indicate that a single treatment was sufficient to increase the alkaline phosphatase level of the patient from 159 to 486IU/L in only 12 months.

As can be seen in fig. 9F-H, treatment resulted in significant improvements in red blood cell levels (fig. 9F), hemoglobin levels (fig. 9G), and hematocrit levels (fig. 9H). These results show that a single treatment is sufficient to ameliorate the symptoms of anemia. Figure 9I shows the level of magnesium in the blood of the patients as a function of time after magnesium supplementation and cell therapy. The data indicate that the patient's blood magnesium levels increase significantly over time such that magnesium supplementation is no longer required. Achieving high magnesium levels without magnesium supplementation is evidence of improved magnesium absorption and reabsorption in the renal proximal tubule.

Figure 9J shows the change in creatinine levels in the blood of the patient over time before and after cell therapy. The data indicate that the patient's creatinine levels are normal initially (below 1mg/dL), but over time his condition worsens about 12 months prior to treatment. Reaching high creatinine levels is a marker for renal failure. After initiation of cell therapy, his condition became stable and prevented further deterioration (shown by the dashed line).

As can be further seen in fig. 9K to 9L, cell therapy also resulted in significant improvements in bicarbonate (fig. 9J) and base excess (fig. 9L) levels without supplementation of bicarbonate.

As can be seen in fig. 9M-9P, a single treatment also resulted in significant reductions in the levels of several indicators of renal tubule disease, such as glucose levels in urine (fig. 9M) and levels of certain salts (fig. 9N-potassium; fig. 9O-chloride; fig. 9P-sodium).

A genetic indicator of success of the therapy used is the occupancy of normal mtDNA compared to total mtDNA. As shown in fig. 10A (pt.1), the occupancy of normal mtDNA in the patients increased from about 1 at baseline to as high as 1.6(+ 60%) in only 4 months and to 1.9(+ 90%) after 20 months from treatment. Notably, normal mtDNA levels are above baseline levels at most time points. As shown in fig. 10B (pt.1), there was a reduction in the level of heterogeneity (less mtDNA present for deletions) following MAT, which had a relatively high level of heterogeneity at baseline. This persists throughout the follow-up.

According to the reports of the neurologist in the hospital, neurological improvement was demonstrated in the patient after autologous cell transplantation with healthy mitochondria (without deletion mutations). The patient improves his walking skills, ability to climb stairs and ability to use scissors and painting. Substantial improvements were also noted in his ability to execute commands, reaction time, and motor and language skills. In addition, the patient's mother reports an improvement in the patient's memory.

As indicated by the data presented above, the single round of treatment provided by the present invention successfully treated PS, FS, improved renal function, and increased occupancy of normal mtDNA in peripheral blood. Evidence of this combination of beneficial effects was further found in the normal weight gain and cognitive status of said patients for healthy subjects of the same age.

Example 8 Green plants with Pearson Syndrome (PS)MNV-BLD (blood-derived mitochondria) enriched for adolescent use +Somatic) homeopathic treatment of autologous CD34 cells

Patient 2 was a 7 year old female patient diagnosed with PS, with a 4977 nucleotide deletion in her mtDNA. The patient also suffered from anemia, endocrine pancreatic insufficiency, and she had suffered from insulin-dependent diabetes for 4 years. Patients have high lactate levels (>25mg/dL), low body weight, and eating and weight gain problems. The patient also suffers from hypermagnesias (high levels in urine and low levels of magnesium in blood). The patient had memory and learning problems, astigmatism and low mitochondrial activity in peripheral lymphocytes, as measured by TMRE, ATP levels and O2Consumption rate (relative to a healthy mother).

Mobilization of Hematopoietic Stem and Progenitor Cells (HSPCs), leukocyte apheresis, and CD34 positive selection were performed similarly to patient 1 (example 3), except that plerixafor (n-2) was added on day-1 prior to leukocyte apheresis. Mitochondria were isolated from maternal Peripheral Blood Mononuclear Cells (PBMCs) by differential centrifugation using 250mM sucrose buffer, pH 7.4. For MAT, autologous CD34 was added+Cells were incubated with the healthy mitochondria from the patient's mother (mitochondria at 1X10 with an amount of 4.4 milliunits Citrate Synthase (CS) per serving6Individual cells) resulted in a 1.62-fold increase in the mitochondrial content of the cells (a 62% increase in mitochondrial content as evidenced by CS activity). Incubation with mitochondria was performed for 24 hours at r.t. in saline containing 4.5% HSA. It should be noted that CD34 from the patient was obtained after mitochondrial enrichment+The colony formation rate of the cells is improved by 26 percent.

1.8X 10 infusion by time line IV as shown in FIG. 11A6Autologous CD34 enriched with healthy mitochondria+Patient 2 (15 KG on the day of treatment) was treated per KG of body weight of cells.

Figure 11B illustrates lactate levels found in the patient's blood as a function of time after i.v. injection. Blood lactate, which is lactic acid that appears in the blood as a result of anaerobic metabolism at the time of mitochondrial damage or when the delivery of oxygen to tissues is insufficient to support normal metabolic demand, is one of the hallmarks of mitochondrial dysfunction. The data indicate that blood lactate levels significantly decrease over time.

Fig. 11C and 11D show the results of the "sit-up test" and "6 minute walk" tests of the patients as a function of time after i.v. injection.

Figure 11E shows the results of the ergometer test performed on the right leg muscle of the patient as a function of time after i.v. injection. Three consecutive replicates were recorded in each experiment. The data indicate that the patient's muscle capacity improves over time, both in terms of increased muscle strength and decreased fatigue.

Figures 11F-11H show the ratio of magnesium, potassium and calcium, respectively, found in the urine of the patient compared to creatinine as a function of time after i.v. injection.

Figure 11I shows the gene ratio between ATP8 and 18S in the urine of the patient as a function of time after i.v. injection.

Fig. 11J shows ATP content in lymphocytes of the patients as a function of time after i.v. injection. The control is the ATP content in lymphocytes of the mother of the patient as donor of mitochondria.

Fig. 10A (pt.2) shows the occupancy of normal mtDNA as a function of time after i.v. injection. As can be seen in fig. 10A (pt.2), the occupancy of normal mtDNA increased from baseline of about 1 to as high as 2(+ 100%) for only 1 month and remained relatively high until 10 months post-treatment. Notably, normal mtDNA levels were above baseline levels at all time points.

Fig. 10B (pt.2) shows the level of heterogeneity as a function of time after MAT. It can be seen that in patient 2 there is a reduction in heterogeneity (less mtDNA present missing) after MAT. This persists throughout the follow-up.

Example 9 juvenile onset of Pearson Syndrome (PS) and Fanconi Syndrome (FS) with MNV- +Sympathy of autologous CD34 cells of BLD (blood-derived mitochondria)Therapy

Patient 3 was a female patient 10.5 years old diagnosed with PS with a deletion at nucleotide 12113-14421 in its mtDNA. The patient also suffers from anemia and fanconi syndrome that progresses to stage 4 renal insufficiency. The patient was treated with dialysis three times a week. In the last two months, patients also suffer from severe visual impairment, narrowing of the visual field and loss of near vision. The patient is completely unable to perform any physical activity (unable to walk, sitting in a stroller). The patient has high lactate levels (>50mg/dL) and pancreatic disorders treated with insulin. Brain MRI shows many lesions and atrophic areas. The patient only eats through gastrostomy. The patient has memory and learning problems. The patient has low mitochondrial activity in peripheral lymphocytes, as measured by tetramethylrhodamine ethyl ester (TMRE), ATP levels, and O2Consumption rate (relative to healthy mothers) determined experimentally.

Mobilization of Hematopoietic Stem and Progenitor Cells (HSPCs) and leukapheresis and CD34 positive selection were performed similarly to patient 1 (example 3), except that plerixafor (n ═ 1) was added on day-1 prior to leukapheresis. Leukemia is performed by permanent dialysis catheters. Mitochondria were isolated from maternal Peripheral Blood Mononuclear Cells (PBMCs) by differential centrifugation using 250mM sucrose buffer, pH 7.4. For MAT, autologous CD34 was added+Cells were incubated with the healthy mitochondria from the patient's mother (mitochondria at 1X10 with an amount of 4.4 milliunits Citrate Synthase (CS) per serving6Individual cells) resulting in a 1.14-fold increase in the mitochondrial content of the cells (14% increase in mitochondrial content as evidenced by CS activity). Cells were incubated with mitochondria for 24 hours at r.t. in saline containing 4.5% HSA. It should be noted that CD34 from the patient was obtained after mitochondrial enrichment+The colony formation rate of the cells is improved by 52 percent.

By infusion according to timeline IV shown in FIG. 12A, 2.8X 106Autologous CD34 enriched with healthy mitochondria from her mother+Patient 3(21KG) was treated per KG of body weight.

Figure 12B illustrates the lactate levels found in the blood of the patient as a function of time before and after therapy.

Fig. 12C shows the level of AST and ALT liver enzymes in the blood of the patients before and after cell therapy as a function of time. Achieving low liver enzyme levels in the blood is evidence of reduced liver damage.

Figure 12D shows the change in the levels of triglycerides, total cholesterol and Very Low Density Lipoprotein (VLDL) cholesterol in the blood of the patient over time before and after cell therapy. Achieving low triglyceride, total cholesterol, and VLDL cholesterol levels in the blood is evidence of increased liver function and improved lipid metabolism.

Glycated hemoglobin (also sometimes referred to as hemoglobin A1c, HbA1c, A1C, Hb1c, Hb1c, or HGBA1C) is a form of hemoglobin that is measured primarily to identify a three month average plasma glucose concentration. Since the lifespan of red blood cells is 4 months (120 days), the test is limited to a three month average. Fig. 12E shows the results of the A1C test on the patients before and after therapy as a function of time.

Fig. 12F and 12G show the results of the "sitting test" (12F) and the "6 min walking" (12G) tests of the patients as a function of time after i.v. injection, showing an improvement of both parameters at 5 months after treatment.

Fig. 10A (pt.3) shows the occupancy of normal mtDNA as a function of time after i.v. injection. As can be seen in fig. 10A (pt.3), the occupancy of normal mtDNA increased by 50% at 7 months post-treatment. Notably, normal mtDNA levels are above baseline levels at most time points.

Fig. 10B (pt.3) shows the level of heterogeneity as a function of time after MAT. It can be seen that in patient 3, which had a relatively low level of heterogeneity at baseline, there was a reduction in heterogeneity (less mtDNA present missing) following MAT. This persists throughout the follow-up.

Taken together, the results presented above demonstrate enhancement of autologous CD34 by enrichment of exogenous healthy functional mitochondria+HSPC, even on low to medium linesMitochondrial enrichment (14% exemplified herein) can also stop disease progression in patients with PS and can lead to improvement of many symptoms.

Example 10 teenagers with Kearns-Sayre syndrome (KSS) use MNV-BLD enriched (blood from) +Mitochondria of origin) of autologous CD34 cells

Patient 4 was a 14 year old, 19.5kg female patient diagnosed with Kearns-Sayre syndrome, experiencing tubular vision, ptosis, ophthalmoplegia, and retinal atrophy. The patient has vision problems, CPEO, seizures, pathological EEG, severe myopathy that fails to sit or walk, arrhythmia. The patient has a deletion of 7.4Kb in their mitochondrial DNA, including the following genes: TK, NC8, ATP8, ATP6, CO3, TG, ND3, TR, ND4L, TH, TS2, TL2, ND5, ND6, TE, NC9 and CYB.

Mobilization of Hematopoietic Stem and Progenitor Cells (HSPCs) and leukapheresis and CD34 positive selection were performed similarly to patient 3 (example 5). For MAT, autologous CD34 was added+Cells were compared with healthy mitochondria (mitochondria in an amount of 4.4 milliunits Citrate Synthase (CS) per serving, 1X 10) from the patient's mother6Individual cells) were incubated in saline containing 4.5% HSA at r.t. for 24 hours. The enrichment resulted in a 1.03-fold increase in the mitochondrial content of the cells (a 3% increase in mitochondrial content as evidenced by CS activity).

Patient 4 was used 2.2 × 10 by following the timeline shown in fig. 12A6Autologous CD34 enriched with healthy mitochondria+Cells/kg body weight were treated.

Surprisingly, CD34 enriched with only 3% healthy mitochondria was used+The patient's EEG showed significant improvement 4 months after the single treatment, with no seizures. At 5 months after treatment, the patient suffered a disease-related Atrioventricular (AV) block and was fitted with a pacemaker. The patient recovered and continued to improve. ATP levels in peripheral blood were measured 6 months after treatment and showed an increase of approximately 100% compared to pre-treatment, as shown in figure 13. In the treatment ofAfter the last 7 months, the patient can sit up alone, walk with assistance, talk, have a better appetite and gain weight of 3.6 KG.

Example 11 treatment of patients with mitochondrial disorders Using human Stem cells enriched for human mitochondria

A patient is diagnosed with a mitochondrial disease based on one or more mutations in their mitochondrial or nuclear DNA, the symptoms they experience, or both.

Patients are treated with autologous or allogeneic human stem cells enriched with healthy functional mitochondria obtained or isolated from healthy donors according to a timeline appropriate to their age, weight and clinical condition. The administered human stem cells are prepared by incubating naive human stem cells with healthy functional mitochondria.

Monitoring the clinical condition of the patient before, during and/or after the therapy. The physiological and/or cognitive clinical condition of the patient may be determined by one of the following tests: weeksler preschool and primary school intelligence scale (WPPSI V3), International Pediatric Mitochondrial Disease Scale (IPMDS) questionnaire, physical examination, neuropsychological examination (e.g., name list memory test according to developmental neuropsychological assessment NEPSY II (NEPSY II-2 nd edition), numerical span test according to Wechsler's children's intelligence scale (WIS) 4 th edition, and Visual Motor Integration (VMI) according to the beer-Buktenica Vis-motion integration developmental test: management, Scoring and instruction manual (6 th edition)), whole blood cell count, blood gas, blood biochemistry, manual differential blood testing, urine biochemistry, weight gain, respiratory function, and normal mitochondrial DNA content.

The scope of the invention is not limited by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the claims.

66页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于治疗组织坏死或改善心脏功能的药物

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类