Method and system to modify an individual's gut-brain axis to provide neurocognitive protection

Information

  • Patent Grant
  • 11980643
  • Patent Number
    11,980,643
  • Date Filed
    Tuesday, August 15, 2023
    10 months ago
  • Date Issued
    Tuesday, May 14, 2024
    a month ago
Abstract
The present invention is directed to beneficially directing the bidirectional communication that exists between the brain and the gut so as to influence brain physiology, psychological responses and ultimately behavior in a positive manner by regulating an individual's mood, psychological symptoms, such as anxiety and depression and stress-related changes in brain function, by modifying the gut microbiome of an individual. An various embodiments, an individual's microbiome is modified in a manner to reduce the likelihood of stress, to reduce or treat symptoms affecting mental health and to promote the mental health of the individual. In certain embodiments, administration to an individual of at least three different bacteria species are combined to modify an individual's microbiome, selected from the group of Lacticaseibacillus paracasei, Coprococcus, Roseburia, Bifidobacterium, L. casei and Faecalibacterium prausnitzii, to reduce adverse psychological and/or physiological responses attendant to psychological stress. Other embodiments reduce certain bacterial populations, such as Veillonella and Megasphaer, while in other embodiments, bacteria that produce hydrogen sulfide, methane and acetate are reduced to beneficially affect an individual's mental health.
Description
FIELD OF THE INVENTION

The present invention is directed to beneficially directing the bidirectional communication that exists between the brain and the gut so as to influence brain physiology, psychological responses and ultimately behavior in a positive manner by regulating an individual's mood, psychological symptoms, such as anxiety and depression and stress-related changes in brain function. This is achieved by modifying the gut microbiome of an individual in a manner to reduce the likelihood of stress, to reduce or treat symptoms affecting mental health and to promote the mental health of the individual. In certain embodiments, administration to an individual of a combination of at least three different bacteria species is done to modify an individual's microbiome, such bacteria selected from the group of Lacticaseibacillus paracasei, Coprococcus, Roseburia, Bifidobacterium, L. casei and Faecalibacterium prausnitzii, so as to reduce adverse psychological and/or physiological responses attendant to psychological stress. Other embodiments reduce certain bacterial populations in the individual, such as Veillonella and Megasphaer, while in other embodiments, bacteria that produce hydrogen sulfide, methane and acetate are reduced to beneficially affect an individual's mental health.


BACKGROUND OF THE INVENTION

An individual's mental health is closely related to emotional, psychological, physiological, physical and social well-being and one's mental health are factors which can determine how an individual handles stress. Mental health affects a wide range of disorders related to anxiety, mood, psychosis, eating behavior, impulse control and addiction, personality, sociability, dissociation, obsessive-compulsive and post-traumatic stress. One's mental health may also be affected by certain diseases and disorders, such as Parkinson's disease, epilepsy and multiple sclerosis, cognitive impairment, dementia and Alzheimer's disease. Major depression and cognitive impairment can mimic that observed in dementia. Moreover, stress is related to an individual's dealing with external and internal challenges and often triggers the release of stress hormones that play a major role in stress adaptation. The HPA axis is a major part of the neuroendocrine system and presents a complex set of interactions between the hypothalamus, the pituitary and the adrenal glands. The HPA axis is involved in numerous processes such as e.g. energy balance, immune system and mood, and also controls reactions to stress, where a hormonal cascade may be activated. The secretion of corticotrophin-releasing hormone (CRH) from the hypothalamus can trigger the release of adrenocorticotrophic hormone (ACTH) from the pituitary gland, which then provokes the release of cortisol from the cortex of the suprarenal gland.


Left untreated, chronic stress may result in an individual suffering from anxiety, sleep problems, muscle pain, high blood pressure, a weakened immune system, heart disease, depression and obesity. Exposure to stress results in alterations within the gut-brain axis, ultimately leading to the development of a broad array of gastrointestinal disorders including inflammatory bowel disease (IBD), irritable bowel syndrome (IBS) and other functional gastrointestinal diseases, food antigen-related adverse responses, peptic ulcers and gastroesophageal reflux disease (GERD). Stress can also affect an individual's gut health, leading to alterations in gastrointestinal motility and secretion, an increase in intestinal permeability, adverse effects on one's gastrointestinal mucosa and mucosal blood flow, as well as an alteration in the indvidual's gut microbial composition.


There are a variety of medications currently employed to treat symptoms of mental illnesses, with almost all having negative side effects such as nausea, increased appetite and weight gain, fatigue and gastrointestinal symptoms.


More than 1 kg of bacteria normally resides in the gut, an equivalent in weight to the whole brain, and represents more organisms than there are cells in the human body. The brain is exceptionally demanding in terms of energy metabolism. Approximately 20% of the calories consumed are devoted to our cerebral faculties, with the lion's share provided in the form of glucose. The variety of genes in gut bacteria is greater than 100 times the quantity of the human genome. It is therefore not surprising that a range of physiologic conditions in the body and the brain are increasingly linked to the status of the gut microbiome. Around 1 billion of the world population is reported to suffer from emotional, psychological and neurological imbalances, substance use disorders and cognitive, psychosocial and intellectual disabilities. It is therefore imperative to understand the role of gut microbes in mental disorders. Depression represents the number one cause of disability worldwide and is often fatal. Inflammatory processes have been implicated in the pathophysiology of depression. It is now well established that dysregulation of both the innate and adaptive immune systems occur in depressed patients and hinder favorable prognosis, including antidepressant responses.


According to the World Health Organization, major depressive disorder (MDD) is a complex debilitating psychiatric disorder that is estimated to account for approximately 10% of worldwide disability. Classic symptoms include depressed mood, anxiety, anhedonia, and cognitive impairments that profoundly affect patients' quality of life. Despite major investments over the last decades into understanding the etiology, progression, and biology of this disorder, its molecular and cellular bases remain poorly defined. There is an increasing emphasis on the fact that depression does not affect brain function exclusively, but manifests as a whole-body disorder affecting almost all of the major corporeal systems.


Antidepressant treatments classically involve the manipulation of the serotonergic and nor-adrenergic systems. However, these antidepressants are suboptimal, as they have a slow onset of action and adverse side effects that sometimes reduce patient compliance and thus limit their efficacy. It is estimated that approximately one third of MDD patients enter remission after first-line antidepressant treatment.


The etiological factors that are responsible for depression include stress and the hypothalamic-pituitary-adrenal (HPA) axis, inflammation and aberrant immune system activation, and the gut microbiome. In psychology there is a growing appreciation of the role of the microbiota-gut-brain axis in psychopathology.


According to the World Health Organization, approximately 4.4% of the world's population is affected by depression and it is the largest contributor to global disability and “non-fatal health loss”, as well as the major contributor to suicide deaths. Individuals suffering from depression often show typical symptoms of sadness, loss of interest and pleasure, feelings of low self-worth, guilt and tiredness, disturbed sleep, and poor concentration.


SUMMARY OF THE INVENTION

Various embodiments of the present invention are directed to modifying the gut-brain axis in a manner that beneficially affects the bidirectional communication that exists between the brain and the gut so as to influence brain physiology, psychological responses and ultimately behavior in a positive manner by regulating an individual's mood, psychological symptoms, such as anxiety and depression and stress-related changes in brain function.


In various embodiments, an individual's microbiome is modified in a manner to reduce the likelihood of stress, to reduce or treat symptoms affecting mental health and to promote the mental health of the individual. In certain embodiments, administration to an individual of at least three different bacteria species are combined to modify an individual's microbiome, e.g. selected from the group of Lacticaseibacillus paracasei, Coprococcus, Roseburia, and Faecalibacterium prausnitzii, to reduce adverse psychological and/or physiological responses attendant to psychological stress. In even more preferred embodiments, certain bacterial populations are reduced, such as Veillonella and Megasphaera. In still other embodiments, reduction in the levels of bacteria that produce hydrogen sulfide, methane and acetate is achieved to beneficially affect mental health.


Certain embodiments of the present invention are directed to a method and system for reducing the likelihood of developing depression in an individual. Depression, also called major depression, major depressive disorder or clinical depression, is a mood disorder that causes a persistent feeling of sadness and loss of interest. It can affect how a person feels, thinks and behaves and can lead to a variety of emotional and physical problems. The methods, compositions, and systems of the present disclosure provide for treatment modalities for depression disorders and depression conditions.


The gut microbiome plays a shaping role in a variety of psychiatric disorders, including major depressive disorder (MDD). The gut microbiome has been linked to several physiological functions relevant to depression. The present inventors contend that particular populations of gut bacteria positively or negatively influence neurobehavioral outcomes.


There is an increasing trend toward evidence supporting the theory of depression as a systemic disease. A systemic illness is one that affects the whole body rather than just a single body part or organ system. This differs from a localized illness that only affects a single part of the body. Depression is a syndrome, a collection of symptoms like any disease. It happens to be a very common disorder, so that about 11% of men and about 21% of women in their lifetime will suffer with what we call major depression. There is now a growing understanding as to why depressed patients are at risk for heart disease, myocardial infarction, and stroke. A very sizable percent of depressed patients exhibit marked increases in markers of inflammation. Inflammation is involved in the pathophysiology of all of the following diseases: Diabetes, stroke, and heart attacks. Patients with inflammatory diseases have high rates of depression, and people with depression have high rates of inflammation.


Fiber is broken down by beneficial microorganisms in the gut into short-chain fatty acids (SCFA), such as butyrate. SCFA carry out many important stabilizing functions in the gut beyond. Since humans cannot break down fiber ourselves, we rely on microbes to do this for us.


Unless the vagus nerve is involved, any gut microbe—brain interaction needs to cross at least two barriers, i.e. the gut epithelium and the blood brain barrier, and permeability through both of these barriers has been shown to be affected by the microbiome. Depression and other neuropsychiatric illnesses have a pro-inflammatory phenotype and inflammatory diseases are often associated with depressive symptoms. Butyrate is believed to be active in these conditions and butyrate has anti-inflammatory features. Butyrate directly affects serotonin and gut hormone release in the enteric nervous system and thereby stimulates the vagus nerve and elicits endocrine signaling, both impacting on brain function.


There is accumulating evidence that butyrate has anti-inflammatory potential and has thus been investigated as a therapeutic agent in inflammatory bowel disorders and colitis. Many people with irritable bowel syndrome are also depressed, people on the autism spectrum tend to have digestive problems, and people with Parkinson's are prone to constipation. Researchers have also noticed an increase in depression in people taking antibiotics—but not antiviral or antifungal medications that leave gut bacteria unharmed.


SCFA affect many aspects of health, including serving as the primary energy source for colon epithelial cells, fortifying the tight junctions that keep the intestinal barrier strong, promoting the production of a healthy intestinal mucus that keeps bacteria a safe distance away from the epithelium and underlying immune system, promoting pH balance at the epithelial surface by increasing expression of intestinal transporters to generate an acidic pH favorable to other fiber-fermenting organisms, and helping the colonization of infectious organisms, which tend to prefer a slightly higher pH. SCFA directly dampen inflammatory response in dendritic cells, macrophages and activated T cells. Higher fiber intake from fruits and vegetables, which is broken down into SCFA by an individual's microbiota, has been associated with lower rates of depression. Diets rich in nutrients including fiber are linked to a reduced risk for inflammation, depression and anxiety. Both fiber and SCFA directly can also protect against the effects of a model of sickness that induces depressive behavior. The effects of fiber are mediated almost entirely by our gut microbiota, and fiber supports a population of microorganisms that survive by breaking it down. Fiber is important in a beneficial gut ecosystem to support and favor particular bacteria, especially those that generate SCFA.


Oxidative stress, caused by reactive oxygen species (ROS) or free radicals, can activate the pro-inflammatory cascade, including IL-6 and CRP, which are both associated with depression. Several antioxidants have been found to either directly dampen depressive symptoms, or are associated with a lower incidence of depression. Fresh fruits and vegetables are bursting with many antioxidants, phytonutrients and fiber content. It is recommended that an adult consume 19-38 g fiber per day.


While not bound by theory, it is believed that Coprococcus is reduced in people with depression. Conversely, it is believed that there is a positive correlation between quality of life and the ability of a gut microbiome that includes Coprococcus bacteria to synthesize a breakdown product of the neurotransmitter dopamine, called 3,4-dihydroxyphenylacetic acid, such breakdown product produced by an individual's microbiome, influencing their mental health. It is further believed that butyrate-producing bacteria, such as Faecalibacterium and Coprococcus bacteria, are associated with a higher quality of life and the absence of severe depression. Individuals who reported a lower quality of life have been noted as having a relatively low abundance of the bacterial genus Faecalibacterium. The abundance of butyrate-producing bacteria in the gut, however, has been associated with a higher quality-of-life score.


Alterations in the microbiome can lead to hyperactivation of the immune system, with production of inflammatory cytokines typically observed in depression. Depression has been associated with impairment of the microbiome's ability to produce neuroactive metabolites and with the disruption of intestinal barrier function. The health-promoting actions of gut microbiota, in particular the provision of beneficial SCFAs by many genera, such as Roseburia and Bacteroides, is relatively high in the human gut. The breakdown of cellulose by the human gut microbiota is believed to be restricted to a few species, such as Ruminococcus champanellensis, a known cellulose degrader. Bacteroides isolated from the human gut are also known to have cellulolytic activity.


Individuals suffering from autoimmune disorders show a high incidence of depressive disorders. The central nervous system (CNS) is directly connected to secondary cervical lymph nodes by a lymphatic drainage system that can evoke peripheral immune responses. Proinflammatory cytokines are believed to play a role in the onset and maintenance of depressive illness. Given the anti-inflammatory effects of many antidepressant medications, neuroimmune mechanisms are now viewed as central to the development of depressive symptoms. Indeed, a role for the adaptive immune system in the etiology of depression was initially proposed when studies revealed that depressed patients had increased numbers of circulating T helper (Th) cells (CD4+), cytotoxic T cells (CD8+), and B cells. The inflammasome is a crucial modulator of the inflammatory response, even though the mechanisms underlying this sophisticated response remain relatively unexplored.


The present inventors believe that there is bidirectional communication of the gastrointestinal microbiota with the endocrine and immune systems in a manner that mediates key brain processes including neuroinflammation, activation of the stress axes, neurotransmission and depression. Commensal bacteria shape immune response by triggering the activation of regulatory T cells through direct recognition of microbial metabolites or products, such as short-chain fatty acids (SCFAs), by T cells or dendritic cells. The microbiome has been implicated in the regulation of neuroinflammatory processes through the modulation of SCFAs, which are microbially derived by-products of fiber metabolism. The gut microbiome is undoubtedly involved in innate immune signaling pathways that impact brain morphology and function. Multiple sclerosis (MS) is a CNS autoimmune disorder, mainly linked to impairments in T cell function. Specific bacteria have been found altered in MS patients. Prior studies have found that microbiota transplantation of MS patients increased the development of spontaneous CNS autoimmunity in comparison to the animals that received a fecal transplantation from healthy twins. Antibiotic exposure has also been found to alter immune gut function while reducing ischemic brain injury. Genetic deletion of caspase-1 reduces depressive-like behavior in mice while resulting in gut microbiome alterations. Antibiotic treatment of stressed mice promoted a rebalance of the gut microbiome in a similar fashion to that found in caspase-1 knockout mice, further implying a role for the gut microbiome in the regulation of inflammasome pathways that affect brain function. Thus, a healthy and diverse gastrointestinal microbiota is deemed vital for the maintenance of a balanced immune system and appropriate brain function throughout an individual's life span.


In one embodiment, an individual's gut is provided with a population of beneficial bacteria selected from the group consisting of Lactobacillus species and such individual is further administered a fiber that maintains a therapeutically effective amount of the beneficial bacteria in the gut of the individual. In preferred embodiments, the individual is administered a therapeutically effective amount of a bacterial formulation of beneficial bacteria comprising at least one of Coprococcus, Veillonella, Roseburia, Bifidobacterium, Faecalibacterium prausnitzii, Lactobacillus rhamnosus and Prevotella. In another preferred embodiment, the individual is administered bacteria that generate short-chain fatty acids, preferably at least one of lactate, propionate and butyrate, and most preferably, butyrate. In other embodiments, the individual is administered Coprococcus bacteria, alone or in conjunction with other bacteria.


Foods particularly high in dietary fibers and polyphenols are preferably provided to the individual to maintain the beneficial bacteria. In particular, bacteria of the genera Faecalibacterium, Bifidobacterium, Lactobacillus, Coprococcus, and Methanobrevibacter are preferably established and maintained in the individual's gut. Certain embodiments of the invention include the enhancement of the production by one or more of these bacteria so as to increase the production of lactate, propionate and/or butyrate so as to inhibit biofilm formation and/or the activity of pathogens. CRISPR-Cas and/or Cpf1 may be employed to provide such characteristics to the selected bacterial species in this regard. In various embodiments of the present invention, these bacterial species are selected and administered to an individual in preferred ratios that reflect those of healthy individuals so as to attain the general balance of bacterial populations in a person's gut.


The beneficial bacteria are preferably modified to produce increased amounts of short-chain fatty acids, preferably butyrate, and may also be encapsulated in a frangible enclosure for administration. In still other embodiments, the level of Roseburia are preferably increased. In other embodiments, the levels of Akkermansia spp. in the individual's gut microbiome are increased. In still other embodiments, a therapeutically effective amount of a bacterial formulation comprising Faecalibacterium prausnitzii is administered, or a composition comprising modified L. reuteri bacteria having the ability to survive conditions in the duodenum or jejunum of the individual's small intestine.


Given the role of vagal nerve stimulation as a treatment in some cases of depression, a role for vagal stimulation in the mechanism of action of certain probiotics forms a central feature of still other embodiments of the present invention. It is believed that the vagus nerve is partially responsible for some of the effects of gut microbiota in depression. Thus, one aspect of the present invention is directed to the targeted intervention to an individual's microbiome to facilitate brain health, including administering probiotics, e.g. including Lactobacillus and Bifidobacterium, in conjunction with a prebiotic to maintain such bacteria in the individual's gut. Such an intervention is believed to promote an individual's resilience to stress and ameliorate emotional responses, including treating such individual's depression.


While not bound by theory, the inventors contend that certain antibiotics have an effect on depression. In support of this contention, it is noted that isoniazid and iproniazid, two of the first antidepressants ever developed, were originally classified and marketed as antibiotics. The selective serotonin reuptake inhibitor (SSRI) fluoxetine, specifically shapes the microbiome in a distinct manner. SSRIs are believed to have certain antimicrobial activity, reaffirming the proposition that psychotropic drugs shape the gut microbiome. The gut microbe Ruminococcus flavefaciens metabolizes fluoxetine and inhibits its mood-affecting effect. Thus, certain embodiments of the present invention include the inhibition of Ruminococcus flavefaciens so as to permit the desired effect of administered fluoxetine.


One aspect of several embodiments of the present invention is directed to the microbiome as a therapeutic target for MDD. While acknowledging that MDD is a complex disorder, and that many patients fail to respond to antidepressant treatment, others respond but do not fully remit. Neuroscience research has established the significance of gut microbiota in the development of brain systems that are essential to stress-related behaviors, including depression and anxiety. An individual's gut microbiota is deeply implicated in mood and behavioral disorders and one aspect of the present invention is that the human gut microbiota can control nervous system diseases through neuroimmune pathways. Whether it is the microbes or their metabolites that have a beneficial effect through the administration of microbes, e.g. via FMT technique, etc., embodiments of the present invention address the fact that either fecal microbiota, bacterial components, metabolites, or bacteriophages, mediate the effects of FMT.


The immune system regulates mood and the causes of the dysregulated inflammatory responses in depressed patients. Inflammation is a critical disease modifier, promoting susceptibility to depression. Controlling inflammation provides an overall therapeutic benefit and is related to microbiome alterations. Depression is associated with an imbalance of the hypothalamic-pituitary-adrenal (HPA) axis, whereby activation by cytokines (interleukins 1 and 6) trigger the release of cortisol, a potent stress hormone. Several observational studies show a bidirectional link between depression and the gut microbiome, which has been linked to dysregulation of the HPA axis. Conversely, improvements in symptoms of depression have correlated with restored stability to HPA activity. Homeostasis of the gut, determined by its unique bacterial community, is believed to be central to maintaining mood stability. Increased immunoglobulin (Ig)A- and IgM-mediated immune responses to specific bacterial lipopolysaccharides in the blood of depressed patients supports the hypothesis that increased intestinal permeability is a factor in depressive illness. Thus, one aspect of certain embodiments of the present invention are directed towards the reduction of intestinal permeability in an individual so as to address depressive illness. One such method involves the administration of beneficial bacteria as described herein to reduce an individual's intestinal permeability, including the provision and maintenance of effective populations of beneficial bacteria selected from the group consisting of Coprococcus, Veillonella, Roseburia, Bifidobacterium, Faecalibacterium prausnitzii and Prevotella.


Certain embodiments of the present invention are directed to a method for reducing the likelihood of developing depression in an individual, by providing in the gut of an individual at least two bacteria from a population of beneficial bacteria selected from the group consisting of Coprococcus, Veillonella, Roseburia, Bifidobacterium, Faecalibacterium prausnitzii and Prevotella; and administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual. Still further embodiments include increasing the levels of at least one of Roseburia, Coprococcus, Veillonella, Bifidobacterium, Lactobacillus, and Prevotella in the individual's gut microbiome.


One of skill in the art will appreciate the following haiku as it relates to the present invention as it relates to depression and its ties to an individual's microbiome:

    • I am so confused
    • if the real monster is me
    • or what's within me.


In certain embodiments, and adjustment to or modification of an individual's gut microbiome is undertaken to decrease the likelihood of depression in an individual. Thus, as it has been observed that in depressed individuals, there is a reduction in Bacteroidetes members. This same observation has been seen with individuals suffering from hypertension. Indeed, hypertensive individuals may be characterized by increased proportions of Lactobacillus and Akkermansia but also have decreased relative abundances of well-known butyrate-producing commensals, including Roseburia and Faecalibacterium within the Lachnospiraceae and Ruminococcaceae families. Thus, in some embodiments, the level of Akkermansia is reduced to counter depression.


It is believed that high blood pressure is related to depression, given the overlap of these population of individuals. ‘Depressive Hypertension’ (high blood pressure with depression), however, is believed to be a completely different disease than ‘Non-Depressive Hypertension’ (high blood pressure without depression). Thus, one aspect of the present invention is to modify the gut microbiome of an individual, via introduction of beneficial bacteria, whether from one species or several that together form a more beneficial mix of bacteria that produce beneficial metabolites, such as small chain fatty acids, such as butyrate. SCFAs are a major energy source for the epithelial cells lining the colon, which keep contents from leaking out of the gastrointestinal tract into the body. SCFAs are thought to play a role in protecting individuals from common problems like inflammation, obesity and diabetes. Other aspects involve decreasing the number of bacteria believed not to be beneficial as to the treatment of depression and/or hypertension. Still other embodiments involve the manipulation of bacteria, preferably using CRISP systems, to either increase or decrease the production of bacterial products. For example, enhancing the production of SCFA's by bacteria, whether they are bacteria that normally produce at least some SCFA or not, is done via CRISPR systems to decrease the likelihood that an individual may suffer from depression and/hypertension.


In certain embodiments, the present invention is directed to a method for reducing the likelihood of developing depression in an individual involves providing in the gut of an individual a population of beneficial bacteria selected from the group consisting of bacterial species able to make small chain fatty acids, and preferably butyrate, and administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual. The individual's gut microbiome is modified to reduce the number of undesired bacteria and to increase the number of beneficial bacteria. Bacteria are preferably modified to remove one or more virulence facts or alternatively to produce increased amounts of SCFA's, such as butyrate. Beneficial bacteria may be encapsulated in a frangible enclosure to ensure they arrive in an individual's body while still viable, e.g. such as being first released in the lower gut rather than being exposed to the harsh conditions of an individual's stomach. In other embodiments, a therapeutically effective amount of a bacterial formulation comprising Faecalibacterium prausnitzii is administered. Other embodiments include the administration of a bacterial formulation comprising at least one, and preferably at least two or more of Coprococcus, Veillonella, Roseburia, Bifidobacterium, Faecalibacterium prausnitzii and Prevotella to treat depression.


Particular embodiments of the invention are directed to a method for reducing the likelihood of depression in an individual human being by first substantially reducing the human being's resident populations of gut microbes prior to administering a therapeutically effective amount of a bacterial formulation comprising Coprococcus, followed by fiber, preferably providing fructan fiber inulin, in an amount sufficient to reduce the pH in the colon of the human being to achieve acidifying of the colon.


Preferably the bacterial formulation is encapsulated. Moreover, the Coprococcus bacteria employed are first isolated from a human being's stool and more preferably are from the human being treated. Certain embodiments include the reduction of Helicobacter pylori populations in the individual human being, whether via antibiotics or by employing a clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR associated protein (Cas) system or a CRISPR from Prevotella and Francisella 1 (Cpf1) system. In other embodiments, the bacterial formulation further includes a microbe selected from the group consisting of Streptococcus, Fusobacterium and Treponema, and in still others, includes a bacterium selected from the group consisting of Chlamydia, Shigella flexneri, Mycoplasma bacteria, Lactobacillus casei, Roseburia, Bifidobacterium, and Faecalibacterium prausnitzii; and Helicobacter pylori. Some embodiments involve increasing the levels of bacterial genera selected from the group consisting of Bifidobacterium, Lachnospira, Roseburia, Lactobacillus and Shigella. Preferrably, the bacteria reduced are selected from the group consisting of Pediococcus, Streptococcus, Enterococcus, and Leuconostoc bacteria, and again, such reduction can be achieved via the use of suitable antibiotics or the use of a CRISPR system. One objective of many embodiments is to provide the individual with a population of beneficial bacteria that have been modified to increase the level of butyrate.


Still further embodiments are directed to a method for reducing the likelihood of developing depression in an individual by providing in the gut of an individual at least two bacteria from a population of beneficial bacteria selected from the group consisting of Coprococcus, Roseburia, Bifidobacterium, and Faecalibacterium prausnitzii; and administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual. Preferably, the at least two bacteria are encapsulated. Another embodiment is directed to reducing the likelihood of developing depression in an individual by providing in the gut of an individual bacteria from a population of beneficial bacteria selected from the group consisting of Coprococcus, Roseburia, Bifidobacterium, and Faecalibacterium prausnitzii; and administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual, preferably having the number of bacteria in the human being reduced using a clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR associated protein (Cas) system or a CRISPR from Prevotella and Francisella 1 (Cpf1) system.


One objective of the present invention is to exploit knowledge of the connection between the human intestinal microbiota and major depressive and bipolar disorders by focusing on the bacteria populations and species that influence depression. While not bound by theory, it is believed that bacteria of the genus Coprococcus can be administered to depressed individuals to improve their quality of life. As the genus Coprococcus is known for its butyrate production, it is believed that increased amounts of butyrate positively impact several diseases, like inflammatory bowel disease, colorectal cancer, and preeclampsia. Provision of certain omega-3 polyunsaturated fatty acids (PUFAs) is believed to increase the production of butyrate by certain bacteria. In addition to Coprococcus, Roseburia and Faecalibacterium, and in particular, F. prausnitzii, are some of the most abundant known butyrate-producing bacteria in the human gut. An increase of bacteria belonging to these genera is believed to provide beneficial physical and mental health effects and provides useful treatments for depressive disorders. Still other bacteria found to be beneficial are from the families of Ruminococcaceae and Sutterellaceae.


It is submitted that there is a close connection between the intestinal microbiota and depression that has previously never been fully appreciated and that significant differences in the microbiota composition of depressed individuals vs. non-depressed individuals can be used to modify a person's microbiome in a fashion to reduce the risk of developing depression, which in some instances may only require a relatively minor change to the microbiota, such that with the remission of depression, a person's intestinal bacteria adopts a more normal, non-depressive composition.


Conversely, certain bacteria should preferably be reduced to combat depression, such as Veillonella and Megasphaera. While not bound by theory, it is believed that species within these genera metabolize lactate to the SCFAs propionate and acetate, and it is believed that excess amounts of propionate is associated with increased depressive-like behaviors. Similarly, it is believed that the levels of bacteria that produce hydrogen sulfide, methane and acetate should be reduced as they have been reported to be in higher abundance in those with mental disorders.


From a diagnostic respect, one aspect of various embodiments of the present invention relates to ascertaining the level of butyrate in fecal matter, as lower butyrate amounts and the presence of reduced levels of butyrate-producing bacteria is believed to be a marker for depression.


The production of butyrate and other SCFAs by host bacteria is due to the anaerobic fermentation of dietary fiber in the gut. Butyrate is the primary source of energy for colon cells and plays an important role in maintaining gut barrier integrity. Butyrate receptors are also highly expressed throughout the body, especially on immune and endocrine cells. As such, while not bound by theory, it is believed that reduced butyrate production contributes to impaired gut barrier permeability and subsequent bacterial translocation into the systemic circulation, and in addition to systemic inflammation, is a cause of depressive behaviors. The intake of fiber to feed beneficial bacteria increases the production of beneficial levels of butyrate.


In still other embodiments of the present invention, it is believed that in depressed subjects, there is a relative decrease in the bacterial genera Akkermansia, Ruminococcus, and Prevotella spp. and over-representation of Actinobacteria and Enterobacteriaceae.


Certain antidepressant drugs may alter relative abundances of different strains in gut bacteria, and thus, one objective of various embodiments is to avoid negatively affecting desired microbial compositions. It should be understood that in the employment of probiotics, such term should be understood to include biologically active therapeutics, which may comprise dead organisms, their components, and bioproducts, e.g. which may be encountered in fecal transplants. Thus, in certain embodiments, administration may comprise transferring stool from a healthy donor into the colon of a patient with an established pathology related to an altered microbiota with the aim to restore the normal microbiota and cure or ameliorate the disease. In other embodiments, however, there is an emphasis on personalizing fecal transplants such that bacteria are obtained from or derived from the subject and after incubation or modification thereof, are returned to the subject.


In various embodiments, prebiotics are employed in the form of non-digestible carbohydrates (fructans, galactooligosaccharides, starch, and others) that aid in the production of energy, metabolites, and micronutrients and that allow growth of certain groups of beneficial bacteria.


It is believed necessary to combine desired probiotics with certain prebiotics to have an impact on depressive or anxiety symptoms. Thus, in certain embodiments, a composition is provided, preferably encapsulated, that contains both a desired bacterial mixture, e.g. one that produces butyrate, and a suitable prebiotic, thus providing a more suitable and beneficial way to administer embodiments of the present invention to combat depression. In addition, however, other forms of administration can be employed, including, e.g. in form of a specific formulas, foods, beverages, and even in topical products.


In still other embodiments, certain vitamins are also employed in combination with probiotic and prebiotic compositions and combinations, For example, vitamin D has direct actions in the gut microbiota that leads to an increase in microbial diversity, with an increase of some microbial populations like Akkermansia muciniphilla and Bifidobacterium spp, which then leads to an augmented production of some microbial metabolites like SCFAs.


In addition, in certain embodiments, a combination of polyphenols and other bioactive compounds in coffee, tea, and chocolate can be combined with probiotic and prebiotic components is employed to modulate the gut microbiota to achieve a reduction of the likelihood of depression in an individual.


It is appreciated that diet remains the greatest gut microbiota shaping factor, and therefore improper dietary habits of an individual will largely determine the dysbiosis status. E.g. Hippocrates' quote “let food be thy medicine and medicine be thy food.” But the present invention in its various embodiments is important to address and stave off depressive conditions. Dietary fibers and microbiota-accessible carbohydrates, positively shape the microbial metabolism and the composition of gut microbiota.


It is known that lactate produced during exercise can be utilized by the Veillonella genus and trigger its growth. The conversion of lactate to propionate by Veillonella atypica has been also positively correlated with run time athletic performance. But excess exercise can lead to gastrointestinal problems and psychological conditions like depression. As such, moderate exercise is suggested when practicing the various embodiments of the present invention. Veilonella spp., Lactobacillus spp., Bacteroides spp., and Propionibacterium spp. are some of the most representative bacteria involved in propionate production, while acetate production is commonly spread among numerous bacterial classes. In addition to butyrate, the actions of these SCFAs extends far beyond the gut, exerting pleiotropic functions in the entire organism, with a remarkable role in host metabolism, epigenomic modifications and in gut-brain communication. SCFAs can exert influence over intestinal barrier integrity and regulate host GI immunity, resulting in peripheral immunity modulation, ultimately protecting against disease states, which involves neuroinflammation, including obesity and affective disorders. SCFAs have also shown that they can protect against neuro-toxin infiltration at the BBB through augmentation of BBB tight junction expression patterns. SCFAs have been shown to directly modulate luminal concentrations of neurotransmitters and neurotrophic factors.


Certain embodiments of the present invention are directed to a method for reducing the likelihood of developing depression in an individual human being by providing in the gut of the individual human being bacteria from a population of beneficial bacteria selected from the group consisting of Coprococcus, Roseburia, and Faecalibacterium prausnitzii, followed by administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual. Preferably, an additional step of reducing bacteria in the gut of the individual, selected from the group consisting of Pediococcus, Streptococcus, Enterococcus, and Leuconostoc bacteria, is performed. This can be accomplished by administering an antibiotic or by using a clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR associated protein (Cas) system or a CRISPR from Prevotella and Francisella 1 (Cpf1) system.


In other embodiments, the beneficial bacteria may include Akkermansia, and in still others, the beneficial bacteria are first isolated from a human being's stool. One objective is to have the population of beneficial bacteria produce a level of butyrate sufficient to inhibit biofilm formation of pathogens and/or to reduce gut inflammation. Preferably the beneficial bacteria are encapsulated.


In addition to the various microorganisms described herein, one of skill in the art will appreciate that the metabolites produced by such bacteria can also greatly influence the diagnosis and treatment of various diseases and conditions. For example, in the area of an individual's central nervous system, the modification of the number and type of microbes, specifically bacteria, in a person's gut can trigger neurotransmitter release via Toll-Like Receptor (TLR) signaling on epithelial, immune, and neuronal cells.


Neuromodulators synthesized by bacteria can be employed to stimulate particular cells of the intestines so that molecules responsible for signal transmission are produced that assist in the release of neurotransmitters. The bacteria thus act as delivery vehicles for neuroactive compounds to the receptors of epithelial cells, thus causing a depolarization in the synaptic area of neurons. The neurotransmitters that bacteria can produce include GABA, serotonin, dopamine, and acetylcholine, all of which are known to affect the emotional state of individuals. The metabolites that various bacteria can produce that similarly are believed to affect emotional states in individuals include, but are not limited to the following: L-glutamate, noradrenaline, tryptophan, phenylalanine, Kynurenic acid, quinolinic acid, indole propionic acid, acetic acid, butyric acid, folate pyroxidine, glutathione. One of skill in the art, with the guidance provided herein, will appreciate the various species of bacteria that are able to produce such metabolites and the myriad of mixtures of such bacterial species to facilitate desired production of certain metabolites. It is appreciated that certain bacteria generate metabolites that still other bacteria employ to produce still other types of metabolites that may be beneficial for an individual's maintenance of a mental state. Thus, mixtures of particular species to accomplish such objectives forms various embodiments of the present invention.


In yet another aspect of the present invention, certain diagnostics and methods are described whereby an individual's microbiome and/or microbiome profile is employed to identify individuals who possess a lower amount of certain metabolites than normal. In particular, urinary metabolites, serum metabolites and those metabolites found in stool are measured to assess their relative abundance to other individuals, thus providing a way to characterize various conditions and disease states via a comparison of such metabolites, useful not only for potential treatments to address such deficiencies or excesses, but to diagnosis conditions and diseases so that appropriate treatments can be pursued.


Other methods determine the relative abundance of microorganisms of interest in a similar fashion, thus providing a way to diagnosis the presence of a condition or disease such that a treatment regimen can then be prescribed. In certain embodiments, the method includes detecting whether the biological sample has an increased proportion of Dialister taxa and/or B. fragilis bacteria compared to a general or healthy population of subjects.


In various embodiments, a kit or system for performing a diagnostic method is provided, with such kit or system including one or more primers, probes, or antibodies specific for a protein, mRNA, bacterial species, probes that hybridize the 16 rRNA gene or any combination thereof that may be associated with particular bacteria, such as Dialister taxa and/or B. fragilis. In certain embodiments, microbiota-derived metabolites are identified and employed that are implicated in interorgan transport and the gut-brain axis. For example, in certain embodiments, indoxyl sulfate and trimethylamine-N-oxide are used to discern microbial influences on host metabolism and gut-brain communication. In various embodiments, microbiota-derived metabolites are used to probe aspects of redox homeostasis, including short-chain fatty acids (e.g., butyrate), indoles (e.g., indoxyl sulfate, indole-3-lactate), trimethylamine-N-oxide (TMAO), which are believed to be elevated in brain tissues. Thus, certain embodiments are directed to the use of commensal microbiota to mediate redox homeostasis inside the central nervous system of mammals.


Certain embodiments of the present invention are directed to a method for reducing the likelihood that an individual will experience a behavioral or psychiatric condition associated with a neurocognitive disorder by administering a therapeutically effective amount of a bacterial formulation that comprises Blautia. Other bacterial formulations include ammonia oxidizing microorganisms, such as Nitrosomonas eutropha (N. eutropha). More preferred embodiments include bacterial formulations containing Roseburia, (particularly Roseburia intestinalis, Lactobacillus paracasei, Coprococcus, F. prausnitzii, and Lactobacillus plantarum, and F. prausnitzii.


Other embodiments are directed to the reduction in the likelihood of an individual suffering cognitive or mental health issues by administering an therapeutically effective amount of a bacterial formulation that comprises Lactobacillus plantarum, with the disorders treated including not only the above listed ones, but also diseases and conditions associated with chronic stress. In preferred embodiments, such bacterial formulations are administered orally via dietary or food supplements, medicaments and/or pharmaceutical formulations.


In still other embodiments, a bacterial formulation and use thereof comprises at least one of Roseburia, Lactobacillus, Blautia, Lactobacillus paracasei, Coprococcus, F. prausnitzii, and Lactobacillus plantarum, for reducing the likelihood of and/or for preventing and/or treating mental illness, symptoms affecting mental health and/or conditions associated with chronic stress.


Mental disorders may be related to anxiety, mood, psychosis, eating behaviour, impulse control and addiction, personality, sociability, dissociation, obsessive-compulsive and post-traumatic stress. Disorders such as Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's Disease share similarities in certain respects with mental disorders due to related changes in the brain's structure, chemistry and function. As described herein, mental or neurocognitive disorders may be a disease or a condition from one of the following: Alzheimer's disease, Lewy body disease, Parkinson's disease, obsessive compulsive disorder (OCD), major depressive disorder, an anxiety disorder, chronic fatigue syndrome, post-traumatic stress disorder, a schizophrenia spectrum disorder, bipolar disorder, psychosis, and mood disorder. In further embodiments, the disorder may be associated with a dysfunction of an individual's microbiota-gut-brain axis, including diseases and conditions of the central nervous system. Various embodiments of the present invention are directed to the reduction, prevention and/or treatment of certain mental disorders, which may be selected from the group consisting of anxiety, insomnia, obsessive-compulsive disorder, post-traumatic stress disorder, depression, autism, chronic fatigue, myalgic encephalomyelitis, bipolar disorder, schizophrenia, dementia, and postpartum depression or psychosis, as well as the other conditions and diseases as referred to herein. For example, in various embodiments the mental disorder may also be associated with one or more of lyme disease, mitochondrial disorders, obesity, sleep apnea, and altitude sickness.


It is known that untreated chronic stress can result in serious health conditions such as anxiety, muscle pain, high blood pressure and a weakened immune system. Stress can contribute to the development of major illnesses, such as heart disease, depression and obesity, and often manifest in the gastrointestinal tract, e.g. inflammatory bowel disease (IBD), irritable bowel syndrome (IBS) and other functional gastrointestinal diseases, food antigen-related adverse responses, peptic ulcers and gastroesophageal reflux disease (GERD). The gut-brain axis describes the bidirectional communication that exists between the brain and the gut and the microbiota-gut-brain axis supports the role of the gut microbiome in this communication system. Because certain mental illnesses and symptoms affecting mental health are comorbid with gastrointestinal disorders whereby emotional and routine daily life stress can disrupt digestive function, certain embodiments of the present invention are directed to the use of probiotics to influence central nervous system function and regulate mood, psychological symptoms such as anxiety and depression and stress-related changes in physiology, behavior and brain function.


Significant evidence links anxiety and depression disorders to the community of microbes that live in the gastrointestinal system. Gamma-aminobutyric acid (GABA) neurotransmitters, secondary bile acids, short-chain fatty acids, and tryptophan metabolites generated from the microbiota are a few of the molecules that regulate these mechanisms.


Tryptophan is an amino acid utilized to synthesize proteins. Intestinal bacteria can directly utilize tryptophan to produce many immunologically important metabolites such as indole, indolic acid derivatives and tryptamines in the gut. Many bacterial species can convert tryptophan into indole and indole derivatives through an enzyme, tryptophanase. Indole acts as an intercellular signaling molecule within the gut microbial ecosystem and has been demonstrated to interact with the gut epithelium. Cooperation between gut microbes possessing the enzymes tryptophan monooxygenase and indole acetamide hydrolase enables the conversion of indole to indole-3-acetic acid.


One aspect of the present invention is directed to the localized production of tryptophan-derived bacterial metabolites to reduce the amount of inflammation the individual would otherwise experience. Certain preferred formulations include a combination of live bacteria, such as L. crispatus, Blautia, F. prausnitzii, and prebiotic glycogen. In certain embodiments, the bacterial formulation is combined with human milk human milk glycans in an amount sufficient to generate tryptophan metabolites.


Certain embodiments of the present invention are directed to compositions and methods of using the same that include Lactobacillus crispatus, employed to reduce inflammation through the production of tryptophan metabolites that act as AHR agonists. Tryptophan is a precursor for serotonin synthesis, a neurotransmitter which affects mood and melatonin which induces sleep. (See, eg. Simmons, et. al. US Pat. Publication No. 20230131201, incorporated herein by this reference.) Preferred metabolites comprise tryptophan metabolites, such as indole-3-aldehyde or indole-3-acetic acid, as well as those that may reduce inflammation. Thus, aspects of various embodiments of the present invention include the use of a live bacterial topical probiotic product that modulates AhR expression through the localized production of tryptophan-derived bacterial metabolites. As discussed herein, aryl hydrocarbon receptor (AHR) is a ligand-activated receptor expressed in many cell types, including intestinal epithelial cells. Such agonists include metabolites of tryptophan such as kynurenine (KYN) and/or kynurenic acid (KYNA). Moreover, bacterial metabolites such as SCFAs modulate AHR activity, sometimes not directly as ligands, but nevertheless in a manner to stabilize and facilitate AHR actions. AHR is believed to act as a mediator in the communication between the host and a microbiome of the host, e.g., its gut microbiota. AHR is believed to be activated by microbial-specific metabolites of dietary tryptophan. Thus, in certain embodiments, one objective is to generate tryptophan metabolites to enhance the intestinal epithelial barrier function. Bacterial formulations are selected to generate such metabolites due to the bacterial decomposition of tryptophan, preferably AHR ligands that activate AHR. In addition, SCFAs can also enhance the gene induction of AHR, and thus, the generation of acetate, propionate, and butyrate improve the response induced by an AHR ligand.


In certain embodiments, removing or selectively killing certain bacteria in an individual's microbiome, followed by purposeful administration with a pre-selected bacterial composition, is one aspect of the present invention. As described herein, and via the numerous references incorporated herein by reference, the use of CRISPR systems to achieve desired modifications of seceted bacteria can be employed to achieve the selective reduction of certain undesired bacterial populations, while other bacteria can be transformed to eliminate certain virulence factors thereof, enabling a person of skill in the art to tailor make bacterial formulations and to practice the methods as set forth herein.


As described herein, butyrate is a beneficial short chain fatty acid primarily produced by gut microbiota and is a major energy source for colonic cells. Butyrate is released when tributyrin is orally consumed. Various bacterial formulations described herein generate butyrate, such as Roseburia and F. prausnitzii, and as such, such a combination forms various aspects of the claimed invention. In certain embodiments, Akkermansia muciniphila and Faecalibacterium prausnitzii are enriched in an individual's gut microbiome by administering tributyrins to the individual. Because Akkermansia muciniphila is an acetate and propionate-producing, mucin-degrading bacteria, its presence in the gut is associated with health benefits. Certain aspects of various embodiments of the present invention are predicated on the inverse relationship-between colonization of Akkermansia muciniphila and obesity. Moreover, certain embodiments are directed to a method of improving an individual's mood or ability to cope with stress by administering an amount of tributyrin to the individual to generate a release of butyrate to the individual's gut. Thus, a preferred method includes administering described bacterial formulations in addition to tributyrin to achieve reduction in mental disorders, including stress.


One will appreciate that this Summary of the Invention is not intended to be all encompassing one of skill in the art will appreciate that the entire disclosure, as well as the incorporated references, provides a basis for the scope of the present invention as it may be claimed now and in future applications.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is an illustration depicting the connection between an individual's gut microbiome and the individual's mental state of depression.





DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS

As referenced above, various embodiments of the invention are directed to a method for reducing the likelihood of depression in an individual human being by first substantially reducing the human being's resident populations of gut microbes prior to administering a therapeutically effective amount of a bacterial formulation comprising Coprococcus, followed by fiber, preferably fructan fiber inulin, so as to increase the levels of butyrate in the individual and to reduce the person's likelihood of depression.


In various embodiments, particular bacterial species are targeted for modification and use to address the treatment of depression. For example, L. reuteri is well-established as one of the most ubiquitous members of the naturally-occurring gut bacteria. Host-specific strains of L. reuteri have been documented to confer broad-spectrum protection from an assortment of microbial and chemical associated disease in humans and animals. However, traditional probiotic therapy involves administration of bacteria with the hope that some bacteria will survive the harsh gastric conditions and colonize the colon where the bacteria will reproduce and live indefinitely. Far fewer bacteria survive in the duodenum, jejunum or ileum because of factors such as acidity, immune response and bile concentration. In certain embodiments, it is believed that bacteria must be present in the duodenum or jejunum of the small intestine for lowering cholesterol and in particular bile acid. Thus, certain aspects of the present invention are directed to the modification of particular bacteria using CRISPR-Cas and/or Cpf1 systems to provide bacteria having the ability to survive the conditions in the duodenum or jejunum of the small intestine. Thus, in one embodiment, CRISPR systems are employed to render certain bacteria adaptive to harsh acid conditions and that are otherwise considered to be beneficial to a person in avoiding fatty liver disease. Highly bile salt hydrolase active bacteria provide an improved agent for reducing serum cholesterol, serum lipids, body fat, and atherogenic index and for prophylaxis and treatment of atherosclerosis, cardiovascular and cerebrovascular diseases. Modification of an individual's gut microbes to render a significant population thereof to have enhanced degrees of BSH characteristics is one objective of various embodiments of the present invention.


Oral administration of probiotics has been shown to significantly reduce cholesterol levels, such cholesterol-lowering effects ascribed to BSH activity. Deconjugated bile salts are less efficiently reabsorbed than their conjugated counterparts, which results in the excretion of larger amounts of free bile acids in feces. Also, free bile salts are less efficient in the solubilization and absorption of lipids in the gut. Therefore, deconjugation of bile salts is believed to lead to a reduction in serum cholesterol either by increasing the demand for cholesterol for de novo synthesis of bile acids to replace those lost in feces or by reducing cholesterol solubility and thereby absorption of cholesterol through the intestinal lumen. Microbial BSHs function in the detoxification of bile salts and in doing so increase the intestinal survival and persistence of producing strains. Thus, one embodiment of the present invention is directed to enhancing the BSH activity by a probiotic bacterium to maximize its prospects of survival in the hostile environment of the gastrointestinal tract. Increased intestinal survival increases the overall beneficial effects associated with strains possessing such BSH enhanced activities. Enhanced BSH activity benefits probiotic bacterium that are able to survive and perform in the intestinal milieu. BSH significantly contributes to bile tolerance and survival and persistence of strains in the intestinal tract. Thus, certain embodiments are directed to the manipulation of bacterial strains to enhance the BSH activity of probiotic strains (either to over express a native BSH or to express or over express a heterologous BSH) to improve their survivability in the intestinal tract. Extraction of fecal bacteria from a person and employing the techniques as described herein on such native populations to enhance various aspects thereof, including for example BSH activity, and then returning such modified gut bacteria to the individual, is one method that may be used to address depression in a positive manner.


This is accomplished in various embodiments by the employment of CRISPR-Cas and Cpf1 systems to insert BSH genes in select bacteria. Certain embodiments include the administration of bile-hydrolyzing strains (especially those modified by CRISPR-Cas and/or Cpf1 systems) to control serum cholesterol. The ingestion of probiotics as described herein is believed to be deemed preferable to statins as a way to achieve a cholesterol-lowering therapy. Manipulation of BSH activity as described herein provides for more robust probiotics (whether delivered orally or via the fecal transplantations as described herein) with improved competitiveness and performance. Statin drugs target many of the underlying inflammatory pathways involved in metabolic syndrome (MetS). Thus, certain embodiments relate to the use of CRISPR-Cas systems to modify bacteria of an individual's microbiome so that they produce effective levels of statin drugs. The metabolic syndrome (MetS) is comprised of a cluster of closely related risk factors, including visceral adiposity, insulin resistance, hypertension, high triglyceride, and low high-density lipoprotein cholesterol; all of which increase the risk for the development of type 2 diabetes and cardiovascular disease. A chronic state of inflammation appears to be a central mechanism underlying the pathophysiology of insulin resistance and MetS. Thus, in various embodiments of the present invention, use of probiotics and prebiotics in combination, as described herein, is employed to address the cause of depression, but that is also believed to address related conditions, such as MetS.


In one embodiment, the bacteria employed and that are modified via CRISPR-Cas and Cpf1 to enhance expression of BSH include Lactobacillus, Bifidobacteria, Pediococcus, Streptococcus, Enterococcus, or Leuconostoc. In another embodiment, the Lactobacillus is Lactobacillus reuteri, optionally, Lactobacillus reuteri (NCIMB 701359), Lactobacillus reuteri (NCIMB 701089), Lactobacillus reuteri (ATCC 55148), Lactobacillus reuteri (ATCC 23272), Lactobacillus reuteri (NCIMB 702655), Lactobacillus reuteri (LMG 18238), Lactobacillus reuteri (CCUG 32271), Lactobacillus reuteri (CCUG 32305), Lactobacillus reuteri (CCUG 37470), Lactobacillus reuteri (CCUG 44001) or Lactobacillus reuteri (CCUG 44144). In another embodiment, the Lactobacillus reuteri adheres to the gastrointestinal epithelial cells, competes for adhesion, or inhibits the binding of other bacteria due to cell surface proteins.


The human gut is a rich habitat populated by numerous microorganisms, each having a CRISPR system. In certain embodiments, the CRISPR-Cas system may be employed to render certain bacteria sensitized to certain antibiotics such that specific chemical agents can selectively choose those bacteria more susceptible to antibiotics, see, e.g. US Pat. Publication No. 2013/0315869 to Qimron, which is incorporated in its entirety by this reference. Another aspect of certain embodiments includes making synthetic CRISPR-containing RNAs that target genes of interest and using them with Cas enzymes.


In various embodiments, the CRISPR-Cas and or Cpf1 system is employed to control the composition of the gut flora, such as by circumventing commonly transmitted modes of antibiotic resistance and distinguishing between beneficial and pathogenic bacteria. For applications that require the removal of more than one strain, multiple spacers that target shared or unique sequences may be encoded in a single CRISPR array and/or such arrays may be combined with a complete set of cas genes to instigate removal of strains lacking functional CRISPR-Cas/Cpf1 systems. Because of the sequence specificity of targeting, CRISPR-Cas/CPF1 systems may be used to distinguish strains separated by only a few base pairs.


There are ongoing ethical concerns arising with respect to the use of CRISPR-Cas systems—especially as it relates to modification of the human genome. In preferred embodiments of the present invention, however, such issues are much less prevalent for various reasons. First, because preferred embodiments relate to the modification of microbes—rather than to the human genome—and especially those microbes that show tropism for humans, the unintended consequences of employing Crispr-Cas on organisms is lessened, if not eliminated. Moreover, use of CRISPR-Cas to also insert genes that have controllable elements such that the cells are killed by triggering the expression of such genes, is another way to reduce if not eliminate concerns about an unintended release of a modified organism. These types of controls are well known to those of skill in the art and have been long employed, for example, by those involved in creating genetically engineered organisms, such as by inserting genes so that organisms become susceptible to various conditions, such as temperature, antibiotic exposure, etc., such that microbes that may somehow escape desired conditions will not be viable. Modifying the human genome, made possible by the CRISPR technique, has its upsides but also equally daunting downsides. Permanent deletion of genes from the human genome is much more controversial than deletion or modification of bacterial genes. Thus, one desirable aspect of the present invention is directed to the far less controversial modification of gut microbes resident in the human being to promote health and to trigger the desired immune responses as described herein.


In various embodiments of the present invention, the present inventors submit that bacterial expression of RNA molecules can be employed to generate miRNA molecules that interact with the human host mRNA during bacterial infection. Thus, such microRNAs derived from bacterial RNAs are used to regulate gene expression of the human host cell involved in different human diseases, including depression (which is believed to be categorized as a legitimate human disease that can be treated.) Bacterially derived microRNA sequences can significantly regulate the expression of various human genes and thus, enhancing an individual's gut bacteria by employing CRISPR systems to regulate microRNA sequences forms various embodiments of the present invention. In addition to depression, microRNAs are believed to be involved in many human diseases, such as cancer, diabetes, rheumatoid arthritis, and others that respond to a particular bacterial environment, and thus, while the present description is focused on depression, it will be understood that other diseases can similarly be addressed by employment of the systems and methods as described herein.


MicroRNAs (miRNA) are small important regulators of gene expression and are currently believed to regulate approximately 70% of human genes. More than a thousand different miRNA have been characterized in the human genome and they all are assumed to function by a similar mechanism: The miRNAs base-pair with target messenger RNA (mRNA) and recruit nucleases that degrade the targeted RNA from the termini and/or inhibit translation. In cancer and many other diseases, deregulation of gene-expression is observed and in many cases miRNAs have been shown to play an integral part or even the causative role in disease development. According to various embodiments, the present invention concerns a method for the treatment, amelioration or prevention of a disease or medical disorder associated with the presence or over-expression of microRNA. Therefore, in certain aspects of the invention, inhibiting miRNA activity is a strategy to treat disease, especially depression.


miRNAs are a class of highly conserved non-coding regulatory factors that negatively regulate more than half of the protein-coding genes in mammals, are essential to most biological processes, including proliferation, differentiation and apoptosis, and their transcription is tightly controlled. In certain embodiments, a CRISPR system and/or a modified CRISPR interference system (CRISPRi) employing inactive Cas9, may be used to reversibly prevent the expression of both monocistronic miRNAs and polycistronic miRNA clusters. Such CRISPR-based systems are reversible and thus provide advantages over more conventional knockdown techniques. The CRISPR/CRISPRi system may be adapted to target a particular miRNA sequence by employing a single repression vector, often entailing using a 20-bp sequence and thus, such a CRISPR/CRISPRi method is useful in the generation of vectors that target multiple miRNAs and with reduced toxicity and can silence miRNAs with no off-target effects. Using such CRISPR systems to silence miRNAs involved in the progression of depression is therefore one focus of particular embodiments of the present invention.


In other embodiments, the use of CRISPR-Cas systems is employed to increase butyrate production of select bacteria. For example, F. prausnitzii, one of the most abundant species in the colon, is an important producer of butyrate, a major product of carbohydrate fermentation which is implicated in providing protection against colorectal cancer and ulcerative colitis. CRISPR systems are used to enhance the production of butyrate by insertion of genes into select F. prausnitzii bacteria to protect against colorectal cancer and other diseases—including reducing the likelihood of depression in an individual.


Because CRISPR-Cas/Cpf1 acts before transcription occurs, it is able to be employed to target regulatory and other elements on the DNA of microbes that make up a person's gut microbiome. In certain embodiments, CRISPR-Cas may be employed to deliver fluorescent markers to certain DNA sequences, thus permitting one to determine whether any particular sample has been treated in accordance with the present invention, thus ensuring, for example, identity of various materials, compliance with safety issues, effectiveness of gene expression or excision, etc. permitting labeling of living cells with a desired color to discern particular attributes and states.


Other embodiments are focused on diet as it relates to the use of probiotics. The gut microbiota plays a critical role in transforming dietary polyphenols into absorbable biologically active species, acting on the estimated 95% of dietary polyphenols that reach the colon. Certain embodiments rely upon the ability to deliver agents via mucosal adhesive strips, such as described, for example, in U.S. Pat. No. 8,701,671, which is fully incorporated herein by this reference. Thus, in various embodiments of the present invention, the engineering of communal bacteria with improved properties using a CRISPR/Cas system is employed to provide for the enhancement of health, especially as it relates to an individual's microbiome. In certain embodiments the present invention is directed to delivering to microbial cells in vivo a delivery vehicle with at least one nucleic acid encoding a gene or nucleotide sequence of interest, such method employing an RNA-guided nuclease. The microbial cells may be either or both pathogenic microbial cells or non-pathogenic bacterial cells and the gene or nucleotide sequence of interest may be a virulence factor gene, a toxin gene, an antibiotic resistance gene, or a modulatory gene, and most preferably the nucleotide sequence of interest comprises 16S ribosomal DNA (rDNA). In various embodiments, the delivery vehicle is a bacteriophage. Thus, various embodiments of the present invention include the use of CRISPR-Cas, with the recognition that this system can be employed to benefit human health by modifying the bacterial and other microbe communities that humans have long been exposed to in a fashion such that the beneficial aspects of such microbes can be preserved, while the disadvantageous aspects can be “cut out” of the microbe DNA—rather than attempting to change or modify the DNA of a human.


The present invention is one way in which human health concerns can be benefited directly by the use of a DNA deletion system without affecting the long term and permanent deletion of human genes. It is not believed to be obvious, let alone intuitive, that human health can be benefited by such a DNA deletion system used in a fashion that affects only gut microbes in a human's system.


Another aspect of the present invention includes the ability to load or impregnate mucosal strips with any number of active agents to achieve other desirable aspects, such as administration of particular vitamins, medicinal components, and certain CRISPR-Cas modified bacteria. In some embodiments the microbes are encapsulated within encapsulation structures selected to provide the desired degree of adhesion to the mucous membranes of the throat, gut, etc., and adapted to release the active ingredients slowly over time in situ. These encapsulation structures may be distributed within the base material in the strip composition. In one embodiment, the encapsulation structures comprise multilamellar microparticles. The multilamellar microparticles are selected to exhibit good adhesion to the mucous membranes of the throat, and are small enough to be effectively distributed in the strip. The strips of the present invention provide the requisite pliability and tensile strength necessary to securely adhere to a person's mucosal tissues for at least one hour, more preferably at least two hours, and preferably a bioadhesive polymer is selected from the group consisting of polycarbophil, carbomer, one or more acrylic polymers, one or more polyacrylic acids, copolymers of these polymers, a water soluble salt of a co-polymer of methyl vinyl ether and maleic acid or anhydride, a combination thereof and their salts. In certain embodiments, a mucosal adhesive strip has a coated surface for resisting bioadhesion that includes at least one patterned polymer including coating layer having a plurality of features attached to or projected into a base surface. The features each have at least one microscale (<1 mm) dimension and have at least one neighboring feature having a substantially different geometry. The patterned coating layer preferably provides an average roughness factor (R) of from 4 to 50. The coating layer resists or enhances bioadhesion as compared to the base surface. An article having a surface coating with topography for controlling bioadhesion comprises a base surface, at least one patterned polymer comprising coating layer including a plurality of spaced apart features attached to or projected into the base surface which provide at least a first feature spacing distance. The features each have at least one microscale dimension and at least one neighboring feature having a substantially different geometry. The coating layer provides an average roughness factor (R) of from 2 to 50, preferably being from 4 to 50. The coating layer resists or enhances bioadhesion as compared to the base surface.


Still other embodiments include the use of bacteria that have been modified to remove or disable one or more virulence factors of the particular bacteria. In this regard, one aspect of the present invention is directed to the modification of certain human-specific pathogens by targeting one or more virulence factors thereof, preferably by using CRISPR-Cas or CRISPR-Cpf1 systems, to excise virulence factors genes, or at least portions thereof or transcriptional or translational controls therefore, such that such pathogenic pathogens are deprived of their undesired pathogenic characteristics. One of skill in the art can readily assess the number and identity of human-specific pathogens, as well as the particular virulence factors associated therewith, and can then, employing the CRISPR systems as referenced herein, remove, render incapable or otherwise disable the virulence facts of such microorganisms such that they no long pose a pathogenic threat to humans. Certain embodiments provide for the delivery, via the strips as described herein, of one or more of the following microorganisms selected from the group comprising Lactobacillus lactis, Lactobacillus helveticus, Lactobacillus jensenii, Lactobacillus acidophilus, Lactobacillus bulgaricus, Lactobacillus amylovorus, Lactobacillus delbrueckii, Lactobacillus casei, Lactobacillus crispatus, Lactobacillus gasseri, Lactobacillus johnsonii, Lactobacillus paracasei, Lactobacillus pentosus, Lactobacillus rhamnosus, Lactobacillus curvatus, Lactobacillus plantarum, Lactobacillus brevis, Lactobacillus buchneri, Lactobacillus fructivorans, Lactobacillus hilgardii, Lactobacillus fermentum, Lactobacillus reuteri, Lactobacillus viridescens, Bifidobacterium bifidum, and Lactobacillus ingluviei. The CRISPR-Cas system is preferably employed to excise the virulence factors of one or more of the following bacteria: Lactobacillus lactis, Lactobacillus helveticus, Lactobacillus jensenii, Lactobacillus acidophilus, Lactobacillus bulgaricus, Lactobacillus amylovorus, Lactobacillus delbrueckii, Lactobacillus casei, Lactobacillus crispatus, Lactobacillus gasseri, Lactobacillus johnsonii, Lactobacillus paracasei, Lactobacillus pentosus, Lactobacillus rhamnosus, Lactobacillus curvatus, Lactobacillus plantarum, Lactobacillus brevis, Lactobacillus buchneri, Lactobacillus fructivorans, Lactobacillus hilgardii, Lactobacillus fermen turn, Lactobacillus reuteri, Lactobacillus viridescens, Bifidobacterium bifidum, Lactobacillus ingluviei and preferably selected from the group comprising the following microorganisms deposited with the German Collection for Microorganisms and Cell Cultures where they are numbered as DSM 25972, DSM 25987, DSM 25988, DSM 25989, DSM 25973 and have been in accordance with the Budapest Treaty regarding International Recognition of the Deposition of Microorganisms for the purpose of patent deposition. In a preferred embodiment of the invention, strips containing effective amounts of these bacteria are provided that are attached to the soft palate region of a person's mouth or on other mucosal surfaces. Other LAB that may be employed in various embodiments include the following: lactobacillus slaivarius CICC 23174; Lactobacillus plantarum CGMCC 1.557, Lactobacillus rhamnosus ATCC 53103, and Lactobacillus acidophilus ATCC 4356.


Moreover, in preferred embodiments, the microbes modified are limited to those demonstrating human tropism such that undesired and unintended changes to other animals and organisms are not affected and that the only implications of such genomic alterations of human specific pathogens are restricted to such species in a manner that is not capable of affecting other than the particular human disease at issue. This can include, for example, modifications and/or employment of integrons, which are a two-component genetic recombination system present in the chromosome of many bacterial species. The integron incorporates mobile genes termed gene cassettes into a reserved genetic site via site-specific recombination, named the Integron/gene cassette system. The integron consists of three basic elements: an integrase gene, an attachment site and a promoter. These elements can be manipulated to, for example, decrease the ability of a particular bacteria in a person's gut from being able to effectively attach to epithelial tissue; or alternatively, to coaggregate with other bacteria.


To provide necessary and sufficient written disclosure and enablement of the various embodiments of the present invention, the following references are incorporated by reference in their entireties: U.S. Pat. No. 9,017,718 to Tan; 20140065218 to Lang et. al.; U.S. Pat. Nos. 6,599,883; 8,383,201; 5,158,789; 20070218114 to Sorousch; 20040136923 to Davidson; U.S. Pat. No. 8,999,372 to Davidson; 20090196907 to Bunick; 20090196908 to Lee; 20030124178 to Haley; 20070293587 to Haley; 20100285098 to Haley; 2006-0204591 to Burrell; U.S. Pat. No. 7,087,249 to Burrelll; U.S. Pat. No. 6,210,699 to Acharya; U.S. Pat. No. 8,865,211 to Tzannis; 20140199266 to Park; U.S. Pat. No. 6,599,883 to Romeo; PCT/US2008/080362 to Dussia; 2007-0218114 to Duggan; 20040136923 to Davidson; 20110142942 to Schobel; 20040120991 to Gardner et al.; Fuchs et al. U.S. Pat. No. 4,136,162; 20040136923 to Davidson; U.S. Pat. No. 4,163,777 to Mitra; U.S. Pat. No. 5,002,970 to Eby, III; 20040096569 to Barkalow et al.; 20060035008 to Virgallito et al.; 20030031737 to Rosenbloom; U.S. Pat. No. 6,919,373 to Lam et al.; 20050196358 to Georglades et al.; U.S. Pat. No. 3,832,460 to Kosti; 2002002057 to Battey et al.; 20040228804 to Jones, et al.; U.S. Pat. No. 6,054,143 to Jones; U.S. Pat. No. 5,719,196 to Uhari; 20150150792 to Klingman; 20140333003 to Allen; 20140271867 to Myers; 20140356460 to Lutin; 20150038594 to Borges; U.S. Pat. No. 6,139,861 to Friedman; 20150216917 to Jones; 20150361436 to Hitchcock; 20150353901 to Liu; U.S. Pat. No. 9,131,884 to Holmes; 20150064138 to Lu; 20150093473 to Barrangou; 20120027786 to Gupta; 20150166641 to Goodman; 20150352023 to Berg; 20150064138 to Lu; 20150329875 to Gregory; 20150329555 to Liras; 20140199281 to Henn; US20050100559 (proctor and Gamble); 20120142548 to Corsi et al.; U.S. Pat. Nos. 6,287,610, 6,569,474, US20020009520, US20030206995, US20070054008; and U.S. Pat. No. 8,349,313 to Smith; and U.S. Pat. No. 9,011,834 to McKenzie; 20080267933 to Ohlson et. al.; 20120058094 to Blasser et. al.; U.S. Pat. No. 8,716,327 to Zhao; 20110217368 to Prakash et. al.; 20140044734 to Sverdlov et al.; 20140349405 to Sontheimer; 20140377278 to Elinav; 20140045744 to Gordon; 20130259834 to Klaenhammer; 20130157876 to Lynch; 20120276143 to O'Mahony; 20150064138 to Lu; 20090205083 to Gupta et al.; 20150132263 to Liu; and 20140068797 to Doudna; 20140255351 to Berstad et al.; 20150086581 to Li; PCT/US2014/036849 and WO 2013026000 to Bryan; 20190070225 to Strandwitz, et. al.; and 202/00132270 to Bolte.


Another aspect of certain embodiments of the present invention is directed to a thin film mucosal layered strip wherein modified bacteria (e.g. via the CRISPR-Cas system) is encapsulated in a frangible enclosure and is present in an amount of at least about 0.5 ml. Other treatment agents may be encapsulated in such strips, such that antibiotics or co-aggregation agents or LAB, etc. can be encapsulated in a manner that they can be released at a time when the person so desires and/or when the strip dissolves to a certain extent, e.g. when the walls of the encapsulating shell is worn thin enough to fracture to release the agent(s), such as when a particular bacterial species that would prosper in the gut but may not survive through the oral cavity or passing through the stomach. The manner in which a capsule can be fractured in order to release its solvent contents is variable and will be understood by those of skill in the art. Preferably, the capsule is constructed in a manner that it is sufficiently robust such that mere transport and packaging of the strips containing such capsules does not cause any leakage or breakage of such capsules. Instead, the design of capsules is such that they are frangible with a considerable amount of force being directly applied thereto once the strips are placed on a particular mucosal surface, such as on the soft palette of a human, such that the person's tongue, when pressing against such capsule, can cause it to fracture to release the contents of the capsule. In other embodiments, two or more different materials may be released.


Short-chain fatty acid production by commensal bacteria is important in regulating the immune system in the gut. Butyrate plays a direct role in inducing the differentiation of regulatory T cells and suppressing immune responses associated with inflammation. Butyrate is normally produced by microbial fermentation of dietary fiber and plays a central role in maintaining colonic epithelial cell homeostasis and barrier function. Various embodiments described herein promote the production of butyrate via modified microbes administered to an individual, alone or in concert with the various other agents as described herein.


Preferably, the modified bacteria employed in certain embodiments of the present invention are administered orally to a patient in order to deliver the therapeutic directly to the site of inflammation in the gut. The advantage of this approach is that it avoids systemic administration of immunosuppressive drugs and delivers the therapeutic directly to the gastrointestinal tract. The viability and stability of such modified bacteria is preferably enhanced to support the production of such microbes of desired agents or therapeutic molecules, e.g. butyrate, SOFA, tomatidine, p53 protein, etc. and by doing so, a method is provided that reduce gut inflammation, enhance gut barrier function, and/or treat or reduce the incidence of depression, autoimmune disorders, inflammatory related diseases, etc. Preferably, such modified bacteria are capable of producing therapeutic anti-inflammation and/or gut barrier enhancer molecules, particularly in the presence of reactive nitrogen species, and more preferably the bacteria are functionally silent until they reach an environment containing local RNS, wherein expression of the therapeutic molecule is induced. In certain embodiments, the genetically or CRISPR engineered bacteria are non-pathogenic and may be introduced into the gut in order to reduce gut inflammation and/or enhance gut barrier function. For example, in some embodiments, the bacteria are under the control of a RNS-responsive regulatory region and a corresponding RNS-sensing transcription factor such that a desired product, e.g. butyrate, is produced, which induces the differentiation of regulatory T cells in the gut and/or promotes the barrier function of colonic epithelial cells. Use of such modified bacteria, especially those modified via CRISPR-Cas systems, provides a way to generate a desired therapeutic effect in a manner that lowers the safety issues associated with systemic exposure.


Various embodiments of the present invention are directed to the field of oncology, and in particular, embodiments directed to a method of ameliorating, treating, or preventing a malignancy in a human subject wherein the steps of the method assist or boost the immune system in eradicating cancerous cells. Cancer victims are very often subject to feelings of enhanced depression, and thus, one aspect of the present invention is to address not only the cause of a person's cancer so as to treat the same, but also the attendant feelings of depression. In certain embodiments, the administration of beneficial bacteria to an individual's microbiome is achieved, with such bacteria being modified so as to produce effective amounts of desired compositions, compounds, agents, etc., e.g. tomatidine, p53 protein, etc., to address cancerous conditions. In several embodiments, the administration of such beneficial bacteria and microbes to an individual's microbiome invokes either an active (or a passive) immune response to destroy, weaken or render less invasive certain cancerous cells. Various other embodiments are drawn to the co-administration of biological adjuvants (e.g., interleukins, cytokines, Bacillus Comette-Guerin, monophosphoryl lipid A, etc.) in combination with conventional therapies for treating cancer. In particular, the co-administration of various pre-biotic compositions to enhance and sustain the desired effects of the beneficial modified bacteria forms another aspect of the present invention. In this regard, incorporation by reference of U.S. Patent Publication No. 2016/0213702 to Maltzahn et al. is included as part of the written description of various aspects of the present invention. For example, in view of the fact that the microbiota of humans is complex and varies by individual depending on genetics, age, sex, stress, nutrition and diet, modifying the numbers and species of gut, oral, vaginal and skin microbiota can alter community function and interaction with the host. A number of probiotic bacteria known in the art, as well as some foods considered to be ‘prebiotic’ that contain substances that promote the growth of certain bacteria and that stimulate beneficial microbiota shifts to improve human health, can be employed in concert with the modified bacteria as described herein to effect desired cancer treatment regimens. For example, the administration of glycans in an amount effective to modulate the abundance of the bacterial taxa can be used to achieve better outcomes for cancer patients.


One application of the present invention is to provide a CRISPR-Cas modified bacteria, such as a lactobacteria or BCG, to a person diagnosed with depression, so as to facilitate the production of SOFA, preferably butyrate, or other beneficial compounds, e.g. tomatidine, in a manner that is effective to preserve muscle mass and function in such individual. Other embodiments include CRISPR-Cas, CasX, CasY, etc. modified bacteria that express levels of tumor suppressor factors, such as p53, in a manner that provides an effective, therapeutic amount to an individual via the production of such factors by one or more of the individual's microbiome (e.g. gut, oral, skin, vaginal, etc.) By having the individual's microbiome responsible for administration of such factors, instead of attempting to administer such factors via more traditional routes, such as injection, pills, etc., it is believed that a better result can be attained in a much more natural fashion. Moreover, in view of the ability to further modify bacteria in various ways to provide desired factors at particular times, or in conjunction with particular agents, it is possible to fine tune the administration of desired factors, such as p53, butyrate, etc. so as to reduce any under or over production thereof. For example, rendering particular modified bacteria sensitive to a predetermined antibiotic can thus provide a way to reduce the numbers of any given modified bacteria in a manner to control the populations of such bacteria in an individual's microbiome, and hence, control the level of production of factors produced by such bacteria. To comply with written description and enablement requirements, incorporated herein by the following references are the following patent publications: U.S. Patent Publication Nos. 2014/0349405 to Sontheimer; 2014/0377278 to Elinav; 2014/0045744 to Gordon; 2013/0259834 to Klaenhammer; 2013/0157876 to Lynch; 2012/276143 to O'Mahony; 2015/0064138 to Lu; 2009/0205083 to Gupta et al.; 2015/0132263 to Liu; U.S. Pat. No. 8,945,839 to Zhang; 2014/0255351 to Berstad et al.; 2015/0086581 to Li; PCT/US2014/036849 and WO 2013026000 to Bryan; 2016/0199424 to Berry et al.; 2013/0326645 to Cost et al.; 2018/0312851 to Falb et al.; 2018/0296582 to von Maltzahn et al.; 2018/0207165 to Harmsen et al., 2018/0000878 to Goodman et al. and 2018/0326008 to Schreiber et al.; 20190029871 to Kovarik; 20170106025 to Kovarik; 20190262298 to Kanthasamy, et. al.; 20190390284 to Kim; 20040170617 to Finegold; 20200188454 to Slykerman; 20180140698 to Clube; and 20170312232 to Vitetta, et. al.


CRISPR-based genetic editing tools offer an efficient way to manipulate expression levels of multiple genes and to provide a solution towards the multivariate modular metabolic engineering, to optimize the drug synthesis pathways with modular, multiplex regulation using only a few core proteins (e.g., dCas9) that are guided to specific sequences by guide RNAs.


In still other embodiments of the present invention, modifying bacteria so as to administer them to a person's microbiome is performed in a manner so that particular agents, factors or proteins derived from fungi and mushrooms, are rendered possible, with desired mushroom derived components believed to have mood altering as well as anti-cancer characteristics, either alone or when used in conjunction with other agents. In particular, combining the referenced ability to have bacteria within a person produce desired amounts of SOFA, alone or in combination with being administered tomatidine, as well as having the same bacteria (or in other embodiments, another bacteria) produce a separate cancer-fighting agent, is one novel aspect of the present invention. In particular, by assessing initially the particular bacterial constituents of an individual's microbiome and then administering to such individual a similar species of microbe, but one which has been modified, preferably via employment of a CRISPR-Cas system, one is able to effectively administer to such individual various desired anti-depressive treatments in a way that is believed to be far less disruptive, efficient and dependable as compared to other routes of administration. The modification of specially designed bacteria that reside in a person's body is believed to alleviate the concerns regarding genetic alteration of the human genome, as what is being modified is a microbiome that is present in a person's body—but is not directly involved in the human genome itself. There are a myriad of ways to combine various triggering factors to turn on or off particular productions of agents, factors or proteins that may be included in such modified microbiome species. The present invention in various embodiments is directed to at least those embodiments where therapeutic agents can be administered by the microbiome of the individual that has cancer so as to effectively treat the depression and/or remedy the symptoms resulting from the disease.


One aspect of the present invention is directed to the employment and modification of an individual's microbiome to address muscle mass retention and as a corollary thereof, to address the counterpart of obesity by lessening the amount of fat storage by such individual. The ties between obesity, inflammation and depression are largely acknowledged, as discussed herein. In certain embodiments, the provision of effective amounts of tomatidine is rendered available to an individual via the inoculation of the individual's microbiome (e.g. oral or gut) by particular bacteria that have been modified to express amounts of tomatidine. Still other embodiments also involve the reduction in the amount of acetate levels in an individual's body, which in turn lowers the amount of insulin the individual will produce, which has the effect of keeping fat cells from storing more energy in the form of fat. The reductions in the amount of acetate available in an individual's body further reduces the amount of the hormone ghrelin, thus reducing the hunger drive of the individual. Thus, the modification of an individual's microbiome influences various aspects of their metabolism in a manner that not only retains and maintains the ability to nurture muscle tissue, but to also reduce obesity by affecting the amount of fat that the body stores. While not bound by theory, it is believed that the gut bacteria of an individual is a substantial source of acetate production. The production of acetate by gut microbes is believed to send signals to the brain of the individual to initiate the production of insulin, conveyed via the vagus nerve. Fine tuning of the amount and type of gut microbes (e.g. via the use of antibiotics to initially reduce the kind and numbers of undesired bacteria, followed by purposeful inoculation of an individual's gut microbiome with modified microbes, e.g. via CRISPR-Cas insertion of particular factors, proteins, etc., such as tomatidine) is an effective way to address not only muscle wasting issues, but also hypertension and obesity issues of individuals.


While there are many gut bacteria that produce acetate and butyrate, particular bacteria are preferably selected and even more preferably are modified using CRISPR-Cas systems to address the levels of acetate and/or butyrate production once such bacteria are introduced (or enhanced) to an individuals' microbiome. Preferably the gut microbiota are members of two bacterial divisions: the Bacteroidetes and the Firmicutes, and most preferably include F. prausnitzii. The modification of an individual's gut microbiome is directed in a manner such that the typical increase seen in the relative abundance of the Firmicutes and a corresponding division-wide decrease in the relative abundance of the Bacteroidetes in obese individuals, is addressed. Obese people have more Firmicutes and almost 90% less Bacteroidetes than the lean people. Preferably, the administration of modified Bacteroidetes is achieved to more substantially reflect gut populations in more lean individuals, and by doing so, reducing the amount of acetate produced by the overall gut microbiome. Such a shift in the population of gut microbes to favor Bacteroidetes over Firmicutes, whether or not coupled with the administration of tomatidine, is one aspect of the present invention's objective of achieving a greater proportion of muscle mass than fat that would otherwise occur in any given individual. In still other embodiments, addressing the acetate production by especially Firmicutes, which has an increased capacity for fermenting polysaccharides relative to the lean-associated microbiome, is another way to achieve this objective, and addresses the significant obesity issues especially prevalent in Western societies.


In yet another embodiment, encapsulated structures, preferably microencapsulated structures, are employed that are filled with desired agents, including but not limited to tomatidine, butyrate, etc. and/or microbes, especially bacteria that are found in an individual's gut microbiome, such as F. prausnitzii, such that effective amounts of the agents can be administered to treat particular diseases. Other agents may include those effective in combatting cancer, such as but not limited to tomatidine, p53 protein, statins, PTEN, rapamycin, and other agents able to treat cancer symptoms. Preferably, the bacteria comprise bacteria that are found in the communities of healthy humans, including, for example, F. prausnitzii, Streptococcus, Actinomyces, Veillonella, Fusobacterium, Porphromonas, Prevotella, Treponema, Neisseria, Haemophilus, Eubacteria, Lactobacterium, Capnocytophaga, Eikenella, Leptotrichia, Peptostreptococcus, Staphylococcus, and Propionibacterium. Such encapsulated structures may be provided as strips that may be manufactured to have desired dissolvable aspects thereto and that further have encapsulated portions that house the desired agents.


Similarly, it is desired to increase the presence in samples provided to urban dwelling expectant mothers of other bacteria, and in particular, Bacteroides-Prevotella, bifidobacteria, Desulfovibrio spp., Clostridium clostridiforme, and Faecalibacterium prausnitzii. Avoidance of antibiotics by the expectant mother during the period to which she is exposed to the various Amish soil constituents is desired if not critical in certain embodiments due to the profound changes due to such during antibiotic treatment. In other embodiments, the use of CRISPR-Cas systems is employed to increase butyrate production of these bacteria. For example, F. prausnitzii, one of the most abundant species in the colon, is an important producer of butyrate, a major product of carbohydrate fermentation which is implicated in providing protection against colorectal cancer and ulcerative colitis.


An individual's microbiome includes the collective genomes of all the microorganisms that are part of the body's ecosystem. As stated herein, various autoimmune diseases are capable of being ameliorated by the practice of the present invention, including Crohn's disease. Prior researchers have found that several specific microbes were more prevalent in patients with Crohn's than in their healthy counterparts, while other bugs were less common in Crohn's cases. Addressing this dysbiosis, or imbalance, in the microbial ecosystem is one aspect of the present invention. In certain embodiments, certain microbiota that were known to disappear in the guts of Crohn's cases, are reintroduced, including Faecalibacterium prausnitzii, and at the same time, several bacteria that are known to proliferate in Crohn's cases, including those linked to IBD and colorectal cancer, are targeted to remove pathogenic abilities. In particular embodiments, providing a collection of microbes, preferably including, for example a higher than normal (e.g. that is found in any random sampling of Amish soil) amount of Faecali, more preferably Enterococcus faecalis, is achieved to expose expectant mothers and infants thereto in order to trigger desired immune system responses. Enhancing the growth and viability of this particular bacterium in the gut—and then use of such modified bacterium to treat individuals with various diseases, such as Crohn's disease and other autoimmune diseases. Similarly, Faecalibacterium prausnitzii, which represent more than 5% of the bacteria in the intestine, is encouraged to populate the guts of patients. Such enhanced growth of this bacterium may also be employed to combat certain forms of inflammatory bowel disease. In various embodiments of the present invention, Enterococcus faecalis are subjected to CRISPR-Cas procedures to remove undesired virulence and pathogenicity factors, such as several genes isolated from resistant enterococci (agg, gelE, ace, cyl LLS, esp, cpd, fsrB) which encode virulence factors such as the production of gelatinase and hemolysin, adherence to caco-2 and hep-2 cells, and capacity for biofilm formation. Deletion and removal of certain antibiotic resistance, for example the acquisition of vancomycin resistance by enterococci, is desired also so as to properly and safely employ this bacteria in the present invention. In a particular embodiment, the addition of E. faecalis LAB3 1 is employed to trigger desired immune system responses.


In certain embodiments, it may be advantageous to genetically modify a gut mucosal-associated bacteria with polynucleotides and as taught herein to express or overexpress the polynucleotides as taught herein or to produce or overproduce the polypeptides, such as butyrate and acetate, directly into the vicinity of, or within the gut mucosal barrier of a human. In a preferred embodiment, the gut mucosal-associated bacteria may by any bacteria from the species F. prausinitzii, Prevotella intermedia, and/or Akkermansia muciniphilla. Such overproduction may be realized by genetic modification tools involving recombinant DNA technologies, genome editing such as by using tools based on CRISPR/cas-like systems, or by classical mutation selection systems.


In an embodiment, the genetically modified host cell may be any bacteria, particularly one which is not from a species of bacteria that naturally occurs or lives in the vicinity of or within the gut mucosal barrier of a mammal. Non-limiting examples of such bacteria include any beneficial isolated intestinal bacterial strains, e.g. probiotic bacteria, particularly strains selected from the genera Lactococcus, Lactobacillus, or Bifidobacterium may be used. In addition, strict anaerobic intestinal bacteria may be used such as those belonging to the genera known to occur in the human intestinal tract. As described herein, in various embodiments, strictly anaerobic bacteria are encapsulated or microencapsulated to avoid contact with oxygen, and are delivered to a human such that the encapsulation is dissolved or fractured to release such bacteria in a portion of the body, e.g. gut, where it can thrive.


Certain embodiments employ the bacterium Flavobacterium akiainvivens, which was discovered in 2012 on the plant Wikstroemia oahuensis, or “akia,” which is a flowering shrub endemic to Hawaii. That bacterium has been found on that plant and no other. The bacterium forms 2- to 3-millimeter diameter colonies that range from cream to off-white in color and wet to mucoid in viscosity, and (it) was isolated from decaying Wikstroemia oahuensis collected on the island of Oahu.


Certain embodiments are directed to the targeted manipulation of the gut microbiome for therapeutic applications, such as the manipulation of the gut microbiome achieved by altering the microbiota population and composition, or by modifying the functional metabolic activity of the microbiome to promote health and restore the microbiome balance. There has been recent progress in the engineering of gut commensals, which also presents great potential for bio-medical applications. Specifically, in Bacteroides thetaiotaomicron, components for tunable gene expression were developed and characterized and expected functional outputs were observed in mice after administration of these engineered B. thetaiotaomicron. Thus, one aspect of various embodiments is to harness such engineered commensals, especially F. prausnitzii for the overproduction of butyrate, for therapeutic purposes.



F. prausnitzii was first isolated in 1922 by C. Prausnitz. Morphologically, F. prausnitzii is a Gram-negative, non-motile and non-sporeforming rod with a diameter of 0.5 to 0.9.times.2.4 to 14.0 .mu.m. F. prausnitzii is a strictly anaerobic bacterium that produces butyrate, formate, D-lactate and CO2 but no hydrogen as fermentation products and F. prausnitzii growth is inhibited by acidic pH and bile salts. The amount of F. prausnitzii in the healthy human gut is linked to diet. Inulin-derived prebiotics have been shown to significantly increase F. prausnitzii concentration in the gut. F. prausnitzii is statistically linked to eight urinary metabolites: dimethylamine, taurine, lactate, glycine, 2-hydroxyisobutyrate, glycolate, 3,5-hydroxylbenzoate and 3-aminoisobutyrate. It is believed that F. prausnitzii has pronounced anti-inflammatory effects. While not bound by theory, F. prausnitzii may induce an increased secretion of an anti-inflammatory cytokine interleukin 10, and a decreased secretion of pro-inflammatory cytokines like interleukin 12 and tumor necrosis factor-.alpha. production. It is further believed that F. prausnitzii has the ability to suppress inflammation, and it is hypothesized that this is due to metabolite(s) secreted by F. prausnitzii, including but not limited to butyrate. The number of F. prausnitzii is significantly higher in the gut of healthy subjects as compared to IBD and it is believed that F. prausnitzii is crucial to gut homeostasis and disease protection.


With the guidance provided herein, as well as the numerous references incorporated by reference herein, one of skill in the art will understand the feasibility of using engineered bacteria to directly manipulate the functional output of the microbiota without major modulation of the microbiota population and composition. Components in the normal diet and/or employing prebiotics and engineered probiotics are therefore harnessed to render a targeted effect on the host through modulating the functional output of the microbiome.



F. prausnitzii is a multi-skilled commensal organism and a chief member of human microbiota. It is broadly distributed in the digestive tract of mammals and also in some insects. It is rich in the hind gut rather than in the stomach, as well as jejunum. The consumption of a higher quantity of animal meat, animal fat, sugar, processed foods, and low fiber diet (the typical westernized diet) reduces the count of F. prausnitzii, while a high-fiber (vegetables and fruits) and low meat diet enhance the count of F. prausnitzii. It is known to consume a variety of diet containing polysaccharides, such as the prebiotic inulin, arabinoxylans, apple pectin, oligofructose, resistant starch, fructan supplement, pectins and some host-derived carbon sources (including d-glucosamine and N-Acetyl-d-glucosamine). Meta-analyses also show that the increased consumption of fiber significantly reduces the risk of mortality.


The discovery of the clustered regularly interspaced short palindromic repeats (CRISPR) and the CRISPR-associated nuclease 9 (Cas9) system, has led to an array of strategies to manipulate the gut microbiome with precision. Engineered phage (with the CRISPR-Cas9 system) can be employed to target pathogenic bacteria, or remove a population of bacteria that aids pathogenic bacterial growth, thereby fine-tuning and restoring the balance of the gut microbiome. CRISPR/Cas9 can also be used to manipulate and differentiate genetically heterogeneous bacteria, even of the same species. Unlike conventional drugs, the CRISPR/Cas9 system targets specific bacteria at the gene level to selectively remove pathogens, virulence factors, genes of undesired expressed proteins, etc. and can further be used as an antimicrobial adjuvant to improve antibiotic treatment. Citorik et. al. demonstrated how CRISPR/Cas9 can be delivered using bacteriophages, targeting the ndm-1 gene, which codes for the broad-spectrum carbapenemase, New-Delhi metallo-.beta.-lactamase. Ndm-1 targeting CRISPR/Cas9 specifically eliminated E. coli harboring the gene without affecting wild-type, or other, E. coli strains present in a synthetic consortium of microbes. Other examples include the re-sensitization of bacteria to antibiotics and immunization of bacteria to incoming plasmids conferring antibiotic resistance using temperate phages. Yosef et al. used CRISPR/Cas9 to target ndm-1 and ctx-M-15, which expresses a broad-spectrum beta-lactamase, and effectively selected the transduced bacteria that were antibiotic-sensitive. Thus, CRISPR/Cas9 may be employed to manipulate the gut microbiome by discriminating at the gene level to change the characteristics and functional output of the gut microbiome for therapeutic applications.


Higher consumption of fruit, vegetables, fibre and red wine has been linked to higher abundances of beneficial bacteria, including butyrate-producing Faecalibacterium prausnitzii. In certain embodiments, desired bacteria, such as SOFA-producers, can also be promoted by metformin. In certain embodiments, the so-called cross feeding of bacteria is encouraged to achieve desired butyrate production. For example, excess acetate produced by certain bacteria is subsequently utilized by butyrate-producing bacteria, such as Faecalibacterium prausnitzii, Roseburia, and Eubacterium, to produce butyrate. This ‘cross-feeding’ effect between Bifidobacterium and butyrate-producing bacteria ultimately leads to an increased butyrate production and augments beneficial effects, such as improvement of the gut barrier integrity and pathogen inhibition. The health-promoting attributes of butyrate-producing bacteria are supported in numerous diseased conditions, such as IBD, Crohn's disease, and ulcerative colitis, where a significant reduction of butyrate-producing bacteria is reported. Though these butyrate-producing bacteria are not directly affected by the supplementation of oligosaccharides, their butyrate production is elevated due to the increased availability of fermentative end products generated by Bifidobacterium. Thus, in various embodiments, prebiotics play an important role in mediating complex interactions among populations in the gut microbiota, thus presenting opportunities to achieve therapeutic approaches. Thus, in certain embodiments, a mixture of bacteria is provided in a probiotic composition to encourage such cross-feeding effect between Bifidobacterium and butyrate-producing bacteria, such as F. prausnitzii Bifidobacterium utilizes supplemented prebiotics, which stimulates their growth. Acetate produced by Bifidobacterium becomes a carbon source for the butyrate-producing microbes, stimulating their growth and butyrate-producing activities and, in turn, modulating the microbiome function and improving gut health. Similarly, provision of dietary fiber that can be metabolized by colonic bacteria into butyrate, achieves the objective of enhancing production of this desired short-chain fatty acid (SOFA), which also acts as a histone deacetylase (HDAC) inhibitor that epigenetically upregulates tumor-suppressor genes in CRC cells and anti-inflammatory genes in immune cells.



F. prausnitzii is not detectable in the fecal samples of babies under 6 months of age (Hopkins et al., 2005.). After that, the number starts to increase gradually, and children of 1 to 2 years of age already have a significant amount of F. prausnitzii in their GI tract. As it is believed that babies are born essentially sterile, and receive bacteria from the environment immediately upon birth, vaginally delivered infants receive their first bacteria as they pass through the birth canal and thus have microbial communities resembling those found in the vaginal microbiota of their mothers, dominated by Lactobacillus, Prevotella or Sneathia spp. Babies born by Caesarean section (C-section) don't receive vaginal microbes but instead get their first bacteria usually from the skin microbiota of their mother, dominated by taxa such as Staphylococcus, Corynebacterium and Propionibacterium spp. There is increasing evidence that the early colonization of bacteria affects the health of the infants and also influences the host health later in life. It is therefore one aspect of the present invention to provide to babies, especially those not born vaginally, with a bacterial composition that mimics what the baby would naturally experience if it were born vaginally.



F. prausnitzii is one of the most abundant bacteria in a healthy human gut and is believed to have a positive effect on the human gut health. F. prausnitzii belongs to the Clostridium leptum group (Clostridium cluster IV), belonging to phylum Firmicutes (Lineage: Bacteria; Firmicutes; Clostridia; Clostridiales; Ruminococcaceae; Faecalibacterium; Faecalibacterium prausnitzii). F. prausnitzii has been previously called Fusobacterium prausnitzii (also cited as F. prausnitzii), with it only distantly being related to Fusobacteria and more closely related to members of Clostridium cluster IV.


Moderate butyrate levels can prevent high-fat-diet-induced insulin insensitivity through epigenetic regulation, and mitochondrial beta-oxidation. F. prausnitzii is one of the unique organisms that reduce various autoimmune diseases, especially type-1 diabetes via the modulation of gut epithelium homeostasis and immune system. Studies associated with gut microbiota and type-1 diabetes have a lower proportion of butyrate-producing organisms, such as Firmicutes and Clostridium, which protects against autoimmune diabetes. While not bound by theory, F. prausnitzii is believed to regulate the development of autoimmune diabetes via butyrate dependent complementary pathways. An abundant quantity of butyrate can lower the gut barrier function and enhance cell apoptosis, with high levels of butyrate stimulating GLP-1 secretion and enhancing insulin sensitivity through cAMP signals, such as PKA and Epac, which inhibit gastric emptying. Due to the inhibition of gastric emptying, butyrate can be excreted slowly and accumulates, influencing the anti-inflammatory potential, pH, and oxidative stress.


Butyrate is the major product of carbohydrate fermentation in the colon. Butyrate modulates several processes and is a known anti-proliferative agent. In cultured cell lines, butyrate inhibits DNA synthesis and cell growth, mainly by inhibiting histone deacetylase. Butyrate is also suggested to regulate the citric acid cycle, fatty acid oxidation, electron transport and TNF-.alpha. signaling. Animal studies have indicated that butyric acid may have antineoplastic properties, which means that it may protect against colon cancer. As dietary fiber is protective against colon cancer because carbohydrates entering the large bowel stimulate the production of butyrate. Butyrate has also been suggested to provide protection against ulcerative. F. prausnitzii is an important producer of butyrate, and the decrease of F. prausnitzii has been correlated to lower concentrations of fecal butyrate in healthy human subjects and it is believed that F. prausnitzii plays an important role in the protection of the colon. While not bound by theory, the benefits of butyrate are thought to depend on several aspects, such as time of exposure and butyrate amount. Increased butyrate production by F. prausnitzii is therefore a desired outcome and employment of CRISPR systems to achieve the same, employing the known gens involved in butyrate by F. prausnitzii is one important embodiment of the present invention.


Studies have shown that there was a statistically significant reduction in the F. prausnitzii abundance during both fiber-free and fiber-supplemented diets, but it is postulated that the reduction during the fiber-supplemented diet was due to the use of pea fiber, which is not believed to support the growth of F. prausnitzii, and thus, with the proper fiber being employed, the increase in butyrate production is achieved. In situations where there is insufficient fiber for the beneficial bacteria to consume, the bacteria end up eroding the mucus of the gut and leads to epithelial access by mucosal pathogens.


The relative abundance of Bacteroidetes and Firmicutes has been linked to obesity, with the Firmicutes ratio being significantly higher in obese individuals. It is believed that a high number of F. prausnitzii leads to higher energy intake, because F. prausnitzii is responsible for a significant proportion of fermentation of unabsorbed carbohydrates in the gut.



F. prausnitzii cultivation has proven difficult because the bacterium is a strictly obligatory anaerobe that does not tolerate any oxygen. As described herein, encapsulation of F. prausnitzii is achieved such that it can be effectively delivered such that the encapsulated structure can degrade or be fractured at an appropriate time and place to release such bacteria to a human to derive beneficial results, e.g. the increased production of butyrate. For example, microencapsulation, in a xanthan and gellan gum matrix, and a subsequent freeze-drying protocol can be employed to achieve this result.


Proton pump inhibitors (PPIs) are among the top 10 most widely used drugs in the world. PPI use has been associated with an increased risk of enteric infections, most notably Clostridium difficile. The gut microbiome plays an important role in enteric infections, by resisting or promoting colonization by pathogens. The differences between PPI users and non-users are consistently associated with changes towards a less healthy gut microbiome. These differences are in line with known changes that predispose to C. difficile infections and can potentially explain the increased risk of enteric infections in PPI users. On a population level, the effects of PPI are more prominent than the effects of antibiotics or other commonly used drugs. PPIs change the gut microbiome through their direct effect on stomach acid. This acidity forms one of the main defenses against the bacterial influx that accompanies ingesting food and oral mucus. PPIs reduce the acidity of the stomach, allowing more bacteria to survive this barrier. Species in the oral microbiome are more abundant in the gut microbiome of PPI users. Gastric bypass surgery compromises the stomach acid barrier and leads to gut microbiome changes similar to the PP I-associate.


Antibiotics can lead to severe changes in the gut microbiota. Antibiotics are also commonly used in treatment of IBD, even though little is known about the effects of antibiotics on gut microbiota. The fecal number of F. prausnitzii is lowered in long treatment periods with antibiotics but it is not presently known how antibiotic resistance of F. prausnitzii may affect human health. It is believed, however, that F. prausnitzii has a notable impact on gut homeostasis and thus, the susceptibility of F. prausnitzii to different antibiotics is believed to be important in the treatment of various ailments. Provision of additional F. prausnitzii after a regimen of antibiotics is therefore one aspect of various methods of the present invention. Antibiotic-induced changes in the gut microbiota are usually temporary, but long-term microbial population fluctuations have also been reported. It is believed that antibiotics may even move the gut microbiota to a new, alternative stable state. Antibiotic-induced alterations in the gut microbiota raise the disease risk by increasing the susceptibility to pathogen colonization; for example, diarrhea caused by Clostridium difficile is a well-known consequence of antibiotic courses. The use of live F. prausnitzii is preferred due to the greater immuno-stimulatory effects of live F. prausnitzii, via TLR2 activation. It is believed that this effect is potentially linked to its barrier maintaining properties. It is butyrate, instead of other substances produced by F. prausnitzii, that exerts significant anti-inflammatory effects observed, and it is believed that the target of butyrate is histone deacetylase 1 (HDAC1).


In other embodiments, the bacterial composition employed includes both F. prausnitzii and Akkermansia muciniphila, another abundant member of the human gut microbiota. It is further believed that Faecalibacterium prausnitzii plays a vital role in diabetes and can be used as an intervention strategy to treat dysbiosis of the gut's microbial community that is linked to the inflammation, which precedes autoimmune disease and diabetes.


The microbiota in adults is relatively stable until the persons get 60 years old. Gut alterations lead to elevated gut permeability and reduced gut mucosal immunity, contributing to the development of various cancers, autoimmune disorders, inflammatory bowel diseases, metabolic syndrome and neurodegenerative diseases. The resultant elevated intestinal permeability is a consequence of reduced expression of tight junction proteins that favors the uncontrolled passage of antigens and enables the translocation of bacterial lipopolysaccharide to the gut connective tissues and to the blood circulation, causing insulin resistance and metabolic endotoxemia.


The gastrointestinal tract pH normally ranges between 5 and 5.5 in the ileum and the colon has a range from 6.6 to 7.0, which is one of the main factors in constructing the shape of the microbial communities in the colon. Diet compositions containing fermentable polysaccharides are regulators of the intestinal pH, which facilitates a more acidic environment through the end-products of SCFAs in the gut.


Stool pH becomes more alkaline with the increase in age and differs significantly between genders with higher consumption of animal protein being one possible mechanism for higher pH. Such alkalinity is generally caused due to its alkaline metabolites produced by proteolytic putrefactive bacteria, such as Bacteroides, Propionibacterium, Streptococcus, Clostridium, Bacillus, and Staphylococcus.


An individual generally represents a unique collection of genera and sub-species and it may be different based on the diet (vegetarian or Western with high protein or fat), the age of the host organism, genetic and environmental factors. Diet greatly influences the diversity of the microbiota in the gut and the microbiota is genetically well equipped to utilize various nutritional substrates to maintain a normal gut microbiota pattern. An adequate SOFA (butyrate) production level is essential for gut integrity and butyrate-producing bacteria, such as Eubacterium, Fusobacterium, Anaerostipes, Roseburia, Subdoligranulum, and Faecalibacterium, but especially, F. prausnitzii, have the potential of anti-inflammatory effect and help to reduce bacterial translocation, improve the organization of tight junctions and stimulate the secretion of mucin to maintain the integrity of the gut, with beneficial effects against inflammation in the gut.


Inflammation is one of the major pathophysiological factors leading to insulin resistance and progressively causes type-2 diabetes. F. prausnitzii counts significantly decreased in diabetic individuals with negative correlation to glycated hemoglobin HbA1c values. Along with Akkermansia muciniphila, F. prausnitzii is abundantly found in individuals with normal glucose tolerance compared to the pre-diabetic subjects. F. prausnitzii can convert acetate into butyrate using butyryl-CoA: Acetate CoA-transferase (BUT) pathways, thereby providing a balanced pH in the gut.


While specific embodiments and applications of the present invention have been described, it is to be understood that the invention is not limited to the precise configuration and components disclosed herein. Various modifications, changes, and variations which will be apparent to those skilled in the art may be made in the arrangement, operation, and details of the methods and systems of the present invention disclosed herein without departing from the spirit and scope of the invention. Those skilled in the art will appreciate that the conception upon which this disclosure is based, may readily be utilized as a basis for designing of other methods and systems for carrying out the several purposes of the present invention to instruct and encourage the prevention and treatment of various human diseases. It is important, therefore, that the claims be regarded as including any such equivalent construction insofar as they do not depart from the spirit and scope of the present invention.

Claims
  • 1. A method for modifying an individual's gut-brain axis to provide neurocognitive protection, comprising, providing an individual human being with a population of beneficial bacteria selected from the group consisting of at least two bacteria selected from the group consisting of Blautia, Coprococcus, Faecalibacterium prausnitzii; Nitrosomonas eutropha, Roseburia intestinalis, Lactobacillus paracasei, and Lactobacillus plantarum; administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual; andreducing a number of bacteria in the gut of the individual selected from the group consisting of Pediococcus, Streptococcus, Enterococcus, and Leuconostoc bacteria.
  • 2. The method of claim 1, wherein said beneficial bacteria further comprises Akkermansia.
  • 3. The method as set forth in claim 1, wherein the beneficial bacteria are selected from the group consisting of bacterial species able to produce small chain fatty acids.
  • 4. The method as set forth in claim 1, wherein the beneficial bacteria produce butyrate.
  • 5. The method as set forth in claim 1, further comprising increasing the number of beneficial bacteria.
  • 6. The method as set forth in claim 1, wherein said beneficial bacteria generate tryptophan metabolites that act as aryl hydrocarbon receptor (AHR) agonists.
  • 7. The method as set forth in claim 1, wherein said beneficial bacteria produce tryptophan metabolites selected from the group consisting of indole-3-aldehyde, indole-3-ethanol, indole-3-pyruvate, and indole-3-acetic acid.
  • 8. The method as set forth in claim 1, wherein said beneficial bacteria further comprise Lactobacillus crispatus.
  • 9. The method as set forth in claim 1, wherein at least some bacteria in the bacterial formulation have been modified by using a using a clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR associated protein (Cas) system or a CRISPR from Prevotella and Francisella 1 (Cpf1) system to enhance the generation of a tryptophan metabolites.
  • 10. A method for modifying an individual's gut-brain axis to provide neurocognitive protection, comprising, providing an individual human being with a population of beneficial bacteria selected from the group consisting of at least two bacteria selected from the group consisting of Blautia, Coprococcus, Faecalibacterium prausnitzii; Nitrosomonas eutropha, Roseburia intestinalis, Lactobacillus paracasei, and Lactobacillus plantarum; administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual; andwherein the bacterial formulation generates an amount of tryptophan metabolites sufficient to act as aryl hydrocarbon receptor (AHR) agonists and thereby reduce inflammation in the individual's gut.
  • 11. The method as set forth in claim 10, wherein at least some bacteria in the bacterial formulation have been modified by using a using a clustered regularly interspaced short palindromic repeats (CRISPR) CRISPR associated protein (Cas) system or a CRISPR from Prevotella and Francisella 1 (Cpf1) system to enhance the generation of a tryptophan metabolite.
  • 12. The method as set forth in claim 10, wherein the beneficial bacteria are encapsulated in a frangible enclosure.
  • 13. The method as set forth in claim 10, further comprising increasing the levels of Roseburia in the individual's gut microbiome.
  • 14. The method as set forth in claim 10, further comprising increasing the populations of at least two of the following in the individual's gut microbiome: Akkermansia muciniphila, Faecalibacterium prausnitzii, Bifidobacterium longum, Roseburia intestinalis, Coprococcus spp. and Veillonella.
  • 15. The method as set forth in claim 10, further comprising inhibiting monoacylglycerolacyltransferase-3 (MGAT3) synthesis in the individual to lower triacylglycerol (TAG) production.
  • 16. The method as set forth in claim 10, further comprising reducing bacteria in the gut of the individual selected from the group consisting of Pediococcus, Streptococcus, Enterococcus, and Leuconostoc bacteria.
  • 17. The method as set forth in claim 10, wherein a secretion of glucagon-like peptide-1 (GLP-1) is stimulated in the individual due to the production of butyrate by the beneficial bacteria.
  • 18. The method as set forth in claim 10, further comprising administering Bifidobacterium breve and B. longum in an amount sufficient to achieve at least one of the following: decrease the severity of diarrhea; and reduce the symptoms of celiac disease.
  • 19. A method for modifying an individual's gut-brain axis to provide neurocognitive protection, comprising, providing an individual human being with a population of beneficial bacteria selected from the group consisting of at least two bacteria selected from the group consisting of Coprococcus, Faecalibacterium prausnitzii; Roseburia intestinalis, Lactobacillus paracasei, and Lactobacillus plantarum; administering fiber to the individual to maintain a therapeutically effective amount of the beneficial bacteria in the gut of the individual; andwherein the bacterial formulation generates an amount of tryptophan metabolites sufficient to act as aryl hydrocarbon receptor (AHR) agonists and thereby reduce inflammation in the individual's gut.
  • 20. The method as set forth in claim 19, wherein the beneficial bacteria are encapsulated in a frangible enclosure.
RELATED APPLICATIONS

This application is a continuation-in-part of U.S. patent application Ser. No. 18/232,980, filed Aug. 11, 2023, which is a continuation of U.S. patent application Ser. No. 18/232,433, filed Aug. 10, 2023, which is a continuation-in-part of U.S. patent application Ser. No. 18/143,399, filed May 4, 2023, which is a continuation-in-part application of U.S. patent application Ser. No. 17/893,384, filed Aug. 23, 2022, which is a continuation-in-part application of U.S. patent application Ser. No. 17/694,775, filed Mar. 15, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 17/023,736, filed Sep. 17, 2020 (now U.S. Pat. No. 11,419,903, issued Aug. 23, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 17/011,175, filed Sep. 3, 2020 (now U.S. Pat. No. 11,273,187, issued Mar. 15, 2022), which is a continuation-in-part of U.S. patent application Ser. No. 16/722,117, filed Dec. 20, 2019 (now U.S. Pat. No. 10,842,834, issued Nov. 24, 2020), which is a continuation-in-part of U.S. patent application Ser. No. 16/229,252, filed Dec. 21, 2018 (now U.S. Pat. No. 10,512,661, issued Dec. 24, 2019), which is a continuation-in-part of U.S. patent application Ser. No. 15/392,173, filed Dec. 28, 2016 (now U.S. Pat. No. 10,245,288, issued Apr. 2, 2019), which is a non-provisional of U.S. Provisional Patent Application Ser. No. 62/275,341, filed on Jan. 6, 2016. This application is a continuation-in-part of U.S. patent application Ser. No. 18/130,946, filed Apr. 5, 2023, which is continuation-in-part of U.S. patent application Ser. No. 18/178,847, filed Mar. 28, 2023, which is a continuation-in-part of U.S. patent application Ser. No. 18/087,545, filed Dec. 22, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 17/854,422, filed Jun. 30, 2022 (now U.S. Pat. No. 11,672,835, issued Jun. 13, 2023), which is a continuation-in-part of U.S. patent application Ser. No. 17/848,759, filed Jun. 24, 2022 (now U.S. Pat. No. 11,642,382, issued May 9, 2023), which is a continuation-in-part of U.S. patent application Ser. No. 17/835,204 filed Jun. 8, 2022 (now U.S. Pat. No. 11,529,379, issued Dec. 20, 2022), which is a continuation-in-part of U.S. patent application Ser. No. 17/567,295 filed Jan. 3, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 17/337,600, filed Jun. 3, 2021 (now U.S. Pat. No. 11,213,552, issued Jan. 4, 2022), which is a continuation-in-part of Ser. No. 17/027,953, filed on Sep. 22, 2020 (now U.S. Pat. No. 11,026,982, issued Jun. 8, 2021), which is a continuation-in-part of U.S. patent application Ser. No. 16/917,096, filed Jun. 30, 2020 (now U.S. Pat. No. 10,940,169, issued Mar. 9, 2021), which is a continuation-in-part of U.S. patent application Ser. No. 16/782,364, filed Feb. 5, 2020 (now U.S. Pat. No. 10,835,560, issued Nov. 17, 2020), which is a continuation-in-part of U.S. patent application Ser. No. 16/423,375, filed May 28, 2019 (now U.S. Pat. No. 10,555,976, issued Feb. 11, 2020), which is a continuation of U.S. patent application Ser. No. 16/160,336, filed Oct. 15, 2018 (now U.S. Pat. No. 10,314,866, issued Jun. 11, 2019), which is a continuation of U.S. patent application Ser. No. 15/403,823, filed Jan. 11, 2017 (now U.S. Pat. No. 10,111,913, issued Oct. 30, 2018), which is a non-provisional of U.S. Provisional Patent Application Ser. No. 62/296,186, filed on Feb. 17, 2016. This application is a continuation-in-part of U.S. patent application Ser. No. 18/103,768, filed Jan. 31, 2023, which is a continuation-in-part of U.S. patent application Ser. No. 17/738,771, filed May 6, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 16/904,056, filed Jun. 17, 2020 (now U.S. Pat. No. 11,523,934, issued Dec. 13, 2022), which is a continuation-in-part of U.S. patent application Ser. No. 15/983,250 filed on May 18, 2018 (now U.S. Pat. No. 10,687,975, issued Jun. 23, 2020), which is a continuation-in-part of U.S. patent application Ser. No. 15/384,716 filed on Dec. 20, 2016 (now issued U.S. Pat. No. 9,987,224, issued Jun. 5, 2018), which claims priority of U.S. Provisional Patent Application Ser. No. 62/387,405, filed on Dec. 24, 2015. This application is also a continuation-in-part of U.S. patent application Ser. No. 17/836,079, filed Jun. 9, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 16/884,772 filed on May 27, 2020 (now U.S. Pat. No. 11,357,722, issued Jun. 14, 2022, which is a continuation-in-part of U.S. patent application Ser. No. 16/136,950, filed Sep. 20, 2018 (now U.S. Pat. No. 10,668,014, issued Jun. 2, 2020), which is a continuation of U.S. patent application Ser. No. 15/385,278, filed Dec. 20, 2016 (now U.S. Pat. No. 10,085,938, issued Dec. 2, 2018), which claims the benefit of U.S. Provisional Application Ser. No. 62/387,404, filed Dec. 24, 2015. The present application is a continuation-in-part of U.S. patent application Ser. No. 17/543,992, filed Dec. 7, 2021, which is a continuation-in-part of U.S. patent application Ser. No. 16/804,361, filed Feb. 28, 2020 (now U.S. Pat. No. 11,191,665, issued Dec. 7, 2021), which is a continuation-in-part of U.S. patent application Ser. No. 16/020,433, filed Jun. 27, 2018 (now U.S. Pat. No. 10,583,033, issued Mar. 10, 2020), which is a continuation-in-part application of U.S. Ser. No. 15/342,643, filed Nov. 3, 2016 (now U.S. Pat. No. 10,010,568, issued Jul. 3, 2018), which seeks priority from U.S. Provisional Patent Application Ser. No. 62/260,906, filed Nov. 30, 2015. This application is a continuation-in-part application of U.S. patent application Ser. No. 16/776,861, filed Jan. 30, 2020 (now U.S. Pat. No. 10,864,109, issued Dec. 15, 2020), which is a continuation of U.S. patent application Ser. No. 16/142,171, filed Sep. 26, 2018 (now U.S. Pat. No. 10,548,761, issued Feb. 4, 2020), which is a continuation-in-part of U.S. patent application Ser. No. 15/395,419, filed Dec. 30, 2016 (now U.S. Pat. No. 10,086,018, issued Oct. 2, 2018), which is a non-provisional of U.S. Provisional Patent Application Ser. No. 62/274,550, filed on Jan. 4, 2016. This application is a continuation-in-part of U.S. patent application Ser. No. 16/426,346, filed May 30, 2019 (now U.S. Pat. No. 10,716,815, issued Jul. 21, 2020), which is a continuation of U.S. patent application Ser. No. 15/639,767, filed Jun. 30, 2017 (now issued U.S. Pat. No. 10,314,865, issuing Jun. 11, 2019), which is a continuation-in-part of U.S. patent application Ser. No. 15/437,976, filed Feb. 21, 2017 (now U.S. Pat. No. 9,730,967, issued Aug. 15, 2017), which is a continuation-in-part application of U.S. patent application Ser. No. 15/228,454, filed Aug. 4, 2016 (now U.S. Pat. No. 9,585,920, issued Mar. 7, 2017), which is a continuation-in-part application of U.S. patent application Ser. No. 14/954,074, filed on Nov. 30, 2015 (now issued U.S. Pat. No. 9,457,077, issued Oct. 4, 2016). The present application is a continuation-in-part of U.S. patent application Ser. No. 17/543,992, filed Dec. 7, 2021, which is a continuation-in-part of U.S. patent application Ser. No. 16/804,361, filed Feb. 28, 2020 (now U.S. Pat. No. 11,191,665, issued Dec. 7, 2021), which is a continuation-in-part of U.S. patent application Ser. No. 16/020,433, filed Jun. 27, 2018 (now U.S. Pat. No. 10,583,033, issued Mar. 10, 2020), which is a continuation-in-part application of U.S. Ser. No. 15/342,642, filed Nov. 3, 2016 (now U.S. Pat. No. 10,010,568, issued Jul. 3, 2018), which seeks priority from U.S. Provisional Patent Application Ser. No. 62/260,906, filed Nov. 30, 2015. This application is a continuation-in-part application of U.S. patent application Ser. No. 16/037,053, filed Jul. 17, 2018 (abandoned). This application is also a continuation-in-part of U.S. patent application Ser. No. 14/752,192 filed on Jun. 26, 2015 (now U.S. Pat. No. 9,549,842, issued Jan. 24, 2017). The entire disclosure of the prior applications are considered to be part of the disclosure of the accompanying application and are hereby incorporated by reference.

US Referenced Citations (523)
Number Name Date Kind
1492600 Laskey May 1924 A
3178341 Hamill et al. Apr 1965 A
3640741 Etes Feb 1972 A
3832460 Kosti Aug 1974 A
3996934 Zaffaroni Dec 1976 A
4136145 Fuchs et al. Jan 1979 A
4136162 Fuchs et al. Jan 1979 A
4163777 Mitra Aug 1979 A
4226848 Nagai et al. Oct 1980 A
4250163 Nagai et al. Feb 1981 A
4285934 Tinnell Aug 1981 A
4286592 Chandrasekaran Sep 1981 A
4292299 Suzuki et al. Sep 1981 A
4381296 Tinnell Apr 1983 A
4517173 Kizawa et al. May 1985 A
4518721 Dhabhar et al. May 1985 A
4568639 Lew Feb 1986 A
4572832 Kigasawa et al. Feb 1986 A
4668232 Cordes et al. May 1987 A
4687841 Spilburg et al. Aug 1987 A
4713243 Schiraldi et al. Dec 1987 A
4715369 Susuki et al. Dec 1987 A
4720486 Spilburg et al. Jan 1988 A
4740365 Yukimatsu et al. Apr 1988 A
4765983 Takayanagi et al. Aug 1988 A
4830008 Meer May 1989 A
4849246 Schmidt Jul 1989 A
4867970 Newsham et al. Sep 1989 A
4889720 Konishi Dec 1989 A
4894232 Reul et al. Jan 1990 A
4900554 Yanagibashi et al. Feb 1990 A
4906463 Cleary et al. Mar 1990 A
4915948 Gallopo et al. Apr 1990 A
4995555 Woodruff Feb 1991 A
5002970 Eby, III Mar 1991 A
5059189 Cilento et al. Oct 1991 A
5064654 Berner et al. Nov 1991 A
5081157 Pomerantz Jan 1992 A
5081158 Pomerantz Jan 1992 A
5116621 Oji et al. May 1992 A
5137729 Kuroya et al. Aug 1992 A
5158789 DuRoss Oct 1992 A
5166233 Kuroya et al. Nov 1992 A
5190053 Meer Mar 1993 A
5192802 Rencher Mar 1993 A
5196202 Konishi Mar 1993 A
5277877 Jeffrey et al. Jan 1994 A
5284161 Karell Feb 1994 A
5298258 Akemi et al. Mar 1994 A
5314915 Rencher May 1994 A
5332576 Mantelle Jul 1994 A
5462749 Rencher Oct 1995 A
5465734 Alvarez et al. Nov 1995 A
5466465 Royds et al. Nov 1995 A
5505956 Kim et al. Apr 1996 A
5518733 Lamothe et al. May 1996 A
5578315 Chien et al. Nov 1996 A
5585098 Coleman Dec 1996 A
5614501 Richards Mar 1997 A
5629003 Horstmann et al. May 1997 A
5643603 Bottenberg et al. Jul 1997 A
5700478 Biegajski et al. Dec 1997 A
5713852 Anthony et al. Feb 1998 A
5718702 Edwards Feb 1998 A
5719196 Uhari et al. Feb 1998 A
5792067 Karell Aug 1998 A
5800832 Tapolsky et al. Sep 1998 A
5804211 Robertson et al. Sep 1998 A
5843021 Edwards et al. Dec 1998 A
5855872 Libin Jan 1999 A
5876995 Bryan Mar 1999 A
5895804 Lee et al. Apr 1999 A
5900245 Sawhney et al. May 1999 A
5948430 Zerbe et al. Sep 1999 A
5988171 Sohn et al. Nov 1999 A
6054143 Jones Apr 2000 A
6072100 Mooney et al. Jun 2000 A
6139861 Friedman Oct 2000 A
6161541 Woodson Dec 2000 A
6174546 Therriault et al. Jan 2001 B1
6210699 Acharya et al. Apr 2001 B1
6250307 Conrad et al. Jun 2001 B1
6284235 Foreman et al. Sep 2001 B1
6287610 Bowling et al. Sep 2001 B1
6352711 Campbell Mar 2002 B1
6375963 Repka et al. Apr 2002 B1
6390096 Conrad et al. May 2002 B1
6431174 Knudson et al. Aug 2002 B1
6432986 Levin Aug 2002 B2
6439238 Brenzel et al. Aug 2002 B1
6453905 Conrad et al. Sep 2002 B1
6458380 Leaderman Oct 2002 B1
6458777 Sonis et al. Oct 2002 B1
6467485 Schmidt Oct 2002 B1
6502574 Stevens et al. Jan 2003 B2
6509028 Williams et al. Jan 2003 B2
6523542 Knudson et al. Feb 2003 B2
6552024 Chen et al. Apr 2003 B1
6555125 Campbell Apr 2003 B2
6569474 Clayton et al. May 2003 B2
6578580 Conrad et al. Jun 2003 B2
6585997 Moro et al. Jul 2003 B2
6599883 Romeo et al. Jul 2003 B1
6620846 Jonn et al. Sep 2003 B1
6713463 Sonis et al. Mar 2004 B2
6722577 Dobyns, III Apr 2004 B2
6726920 Theeuwes et al. Apr 2004 B1
6734157 Radwanski et al. May 2004 B2
6748951 Schmidt Jun 2004 B1
6794318 Anderson et al. Sep 2004 B2
6803420 Cleary et al. Oct 2004 B2
6916480 Anderson et al. Jul 2005 B2
6919373 Lam et al. Jul 2005 B1
6923981 Leung et al. Aug 2005 B2
7001609 Matson et al. Feb 2006 B1
7067116 Bess et al. Jun 2006 B1
7087249 Burrell et al. Aug 2006 B2
7097853 Garbe et al. Aug 2006 B1
7122198 Singh et al. Oct 2006 B1
7138135 Chen et al. Nov 2006 B2
7143709 Brennan et al. Dec 2006 B2
7146981 Knudson et al. Dec 2006 B2
7267975 Strobel et al. Sep 2007 B2
7276246 Zhang Oct 2007 B2
7287646 Gierskcky Oct 2007 B2
7306812 Zhang Dec 2007 B2
7332230 Krumme Feb 2008 B1
7353194 Kerker et al. Apr 2008 B1
7357891 Yang et al. Apr 2008 B2
7425292 Yang et al. Sep 2008 B2
7441559 Nelson et al. Oct 2008 B2
7470397 Meathrel et al. Dec 2008 B2
7500484 Nelson et al. Mar 2009 B2
7540432 Majerowski et al. Jun 2009 B2
7566310 Badr et al. Jul 2009 B2
7575739 Tamarkin et al. Aug 2009 B2
7579019 Tapolsky et al. Aug 2009 B2
7579078 Hartmann et al. Aug 2009 B2
7615235 Rademacher et al. Nov 2009 B2
7632525 Dodds et al. Dec 2009 B2
7645803 Tamarkin et al. Jan 2010 B2
7648712 Bess et al. Jan 2010 B2
7650848 Brennan et al. Jan 2010 B2
7666502 Magill et al. Feb 2010 B2
7669603 Knudson et al. Mar 2010 B2
7686021 Knudson et al. Mar 2010 B2
7704518 Tamarkin et al. Apr 2010 B2
7727466 Meathrel et al. Jun 2010 B2
7799337 Levin Sep 2010 B2
7820145 Tamarkin et al. Oct 2010 B2
7820420 Whitlock Oct 2010 B2
7824588 Yang et al. Nov 2010 B2
7824704 Anderson et al. Nov 2010 B2
7845356 Paraschac et al. Dec 2010 B2
7862808 Isolauri et al. Jan 2011 B2
7901925 Bojrab Mar 2011 B2
7906140 Bromley et al. Mar 2011 B2
7937159 Lima et al. May 2011 B2
7984714 Hausmann et al. Jul 2011 B2
7992566 Pflueger et al. Aug 2011 B2
7997266 Frazier et al. Aug 2011 B2
8030457 Jackson et al. Oct 2011 B2
8034601 Boileau et al. Oct 2011 B2
8034606 Park et al. Oct 2011 B2
8104478 Pflueger et al. Jan 2012 B2
8110215 Koenig et al. Feb 2012 B2
8197872 Mills et al. Jun 2012 B2
8349313 Smith et al. Jan 2013 B2
8357368 Dudek et al. Jan 2013 B2
8362206 Wallach et al. Jan 2013 B2
8383201 Berry et al. Feb 2013 B2
8420074 Rehberger et al. Apr 2013 B2
8454729 Mittelmark et al. Jun 2013 B2
8481299 Gueniche et al. Jul 2013 B2
8496914 Bonfiglio Jul 2013 B2
8584685 Kovarik et al. Nov 2013 B2
8585588 Kovarik et al. Nov 2013 B2
8591412 Kovarik et al. Nov 2013 B2
8657879 Shalon et al. Feb 2014 B2
8685389 Baur et al. Apr 2014 B2
8701671 Kovarik Apr 2014 B2
8716327 Zhao et al. May 2014 B2
8757173 Kovarik et al. Jun 2014 B2
8758764 Masignani et al. Jun 2014 B2
8815538 Lanzalaco et al. Aug 2014 B2
8829165 Jackson et al. Sep 2014 B2
8859741 Jackson et al. Oct 2014 B2
8865211 Tzannis et al. Oct 2014 B2
8936030 Kovarik et al. Jan 2015 B2
8945839 Zhang et al. Feb 2015 B2
8951775 Castiel et al. Feb 2015 B2
8999372 Davidson et al. Apr 2015 B2
9010340 Kovarik et al. Apr 2015 B2
9011834 McKenzie et al. Apr 2015 B1
9016221 Brennan et al. Apr 2015 B2
9017718 Tan et al. Apr 2015 B2
9028841 Henn et al. May 2015 B2
9044420 Dubensky, Jr. Jun 2015 B2
9045547 Jackson et al. Jun 2015 B2
9056912 Grandi et al. Jun 2015 B2
9095704 McGuire et al. Aug 2015 B2
9131884 Holmes et al. Sep 2015 B2
9149429 Kovacs et al. Oct 2015 B2
9234204 Qvit-Raz et al. Jan 2016 B2
9254295 Adams et al. Feb 2016 B2
9288981 Gandhi et al. Mar 2016 B2
9295682 Nunes et al. Mar 2016 B2
9314489 Kelly et al. Apr 2016 B2
9408880 Kovarik et al. Aug 2016 B2
9445936 Kovarik Sep 2016 B2
9457077 Kovarik et al. Oct 2016 B2
9549842 Kovarik Jan 2017 B2
9585920 Kovarik et al. Mar 2017 B2
9730967 Kovarik et al. Aug 2017 B2
9750802 Kovarik et al. Sep 2017 B2
9795641 Nardelli et al. Oct 2017 B2
9987224 Kovarik et al. Jun 2018 B2
10085938 Kovarik et al. Oct 2018 B2
10086018 Kovarik Oct 2018 B2
10111913 Kovarik Oct 2018 B2
10195273 Clube Feb 2019 B2
10245288 Kovarik Apr 2019 B2
10314865 Kovarik Jun 2019 B2
10314866 Kovarik Jun 2019 B2
10512661 Kovarik Dec 2019 B2
10548761 Kovarik Feb 2020 B2
10555976 Kovarik Feb 2020 B2
10668014 Kovarik et al. Jun 2020 B2
10683323 Prakash et al. Jun 2020 B2
10687975 Kovarik et al. Jun 2020 B2
10716815 Kovarik et al. Jul 2020 B2
10730827 Wortmann et al. Aug 2020 B2
10760075 Sommer et al. Sep 2020 B2
10835560 Kovarik Nov 2020 B2
10842834 Kovarik Nov 2020 B2
10864109 Kovarik Dec 2020 B2
10940169 Kovarik et al. Mar 2021 B2
11026982 Kovarik Jun 2021 B2
11083760 Han Aug 2021 B2
11213552 Kovarik Jan 2022 B2
11273187 Kovarik Mar 2022 B2
11357722 Kovarik et al. Jun 2022 B2
11419903 Kovarik Aug 2022 B2
11523934 Kovarik et al. Dec 2022 B2
11529379 Kovarik Dec 2022 B2
11642382 Kovarik May 2023 B2
11672835 Kovarik Jun 2023 B2
20020009520 Clayton et al. Jan 2002 A1
20020022057 Battey et al. Feb 2002 A1
20020037310 Jonn et al. Mar 2002 A1
20020044988 Fuchs et al. Apr 2002 A1
20030031737 Rosenbloom Feb 2003 A1
20030062050 Schmidt Apr 2003 A1
20030083287 Burgess et al. May 2003 A1
20030104041 Hsu et al. Jun 2003 A1
20030106243 Tucker Jun 2003 A1
20030124178 Haley Jul 2003 A1
20030140930 Knudson et al. Jul 2003 A1
20030149387 Barakat et al. Aug 2003 A1
20030149445 Knudson et al. Aug 2003 A1
20030206995 Bowling et al. Nov 2003 A1
20040053352 Ouyang et al. Mar 2004 A1
20040057962 Timmerman Mar 2004 A1
20040096569 Barkalow et al. May 2004 A1
20040110111 Wasylucha Jun 2004 A1
20040115223 Follansbee Jun 2004 A1
20040120991 Gardner et al. Jun 2004 A1
20040136923 Davidson et al. Jul 2004 A1
20040142463 Walker et al. Jul 2004 A1
20040166501 Azimzai et al. Aug 2004 A1
20040170617 Finegold Sep 2004 A1
20040180080 Furasawa et al. Sep 2004 A1
20040224007 Zhang Nov 2004 A1
20040228804 Jones et al. Nov 2004 A1
20050004417 Nelson et al. Jan 2005 A1
20050159637 Nelson et al. Jan 2005 A9
20050118655 Weinstock et al. Jun 2005 A1
20050137109 Quan et al. Jun 2005 A1
20050196358 Georgiades et al. Sep 2005 A1
20050260544 Jones et al. Nov 2005 A1
20060018843 Fine Jan 2006 A1
20060035008 Virgallito et al. Feb 2006 A1
20060064903 Tucker Mar 2006 A1
20060127330 Tsuchida et al. Jun 2006 A1
20060188813 Shimada Aug 2006 A1
20060204591 Burrel et al. Sep 2006 A1
20060207721 Slominski et al. Sep 2006 A1
20060252087 Tang et al. Nov 2006 A1
20070054008 Clayton et al. Mar 2007 A1
20070057086 Van Kippersluis et al. Mar 2007 A1
20070059718 Toner et al. Mar 2007 A1
20070059774 Grisham et al. Mar 2007 A1
20070063026 Mamaropolos et al. Mar 2007 A1
20070087020 O'Connor Apr 2007 A1
20070093420 Yeomans et al. Apr 2007 A1
20070098744 Knorr et al. May 2007 A1
20070102010 Lemperle et al. May 2007 A1
20070122455 Myers et al. May 2007 A1
20070123448 Kaplan et al. May 2007 A1
20070148136 Whitlock Jun 2007 A1
20070202057 Fankhauser et al. Aug 2007 A1
20070207955 Tanihara et al. Sep 2007 A1
20070218114 Duggan Sep 2007 A1
20070227545 Conrad et al. Oct 2007 A1
20070231923 Cumberland et al. Oct 2007 A1
20070246052 Hegde et al. Oct 2007 A1
20070261701 Sanders Nov 2007 A1
20070280964 Knorr et al. Dec 2007 A1
20070293587 Haley Dec 2007 A1
20070295340 Buscemi Dec 2007 A1
20080032253 Montgomery et al. Feb 2008 A1
20080075825 Fuisz et al. Mar 2008 A1
20080112983 Bufe et al. May 2008 A1
20080242543 Banerjee et al. Oct 2008 A1
20080267933 Ohlson et al. Oct 2008 A1
20080286210 He et al. Nov 2008 A1
20080305089 Bufe et al. Dec 2008 A1
20090004275 Martyn et al. Jan 2009 A1
20090098192 Fuisz Apr 2009 A1
20090130199 Kovacs et al. May 2009 A1
20090148482 Peters Jun 2009 A1
20090196907 Bunick et al. Aug 2009 A1
20090196908 Lee et al. Aug 2009 A1
20090205083 Gupta et al. Aug 2009 A1
20100029832 Pinnavaia et al. Feb 2010 A1
20100040593 Hedman et al. Feb 2010 A1
20100040712 Fisher Feb 2010 A1
20100081681 Blagosklonny Apr 2010 A1
20100092406 Perez-Davidi et al. Apr 2010 A1
20100143447 Hansen et al. Jun 2010 A1
20100229876 Knudson et al. Sep 2010 A1
20100247644 Domb et al. Sep 2010 A1
20100260720 Sprenger Oct 2010 A1
20100285098 Haley Nov 2010 A1
20110009834 Asmussen et al. Jan 2011 A1
20110033542 Myers et al. Feb 2011 A1
20110088701 Thornton Apr 2011 A1
20110100378 Rousseau May 2011 A1
20110142942 Schobel et al. Jun 2011 A1
20110217368 Prakash et al. Sep 2011 A1
20110230587 MacInnis et al. Sep 2011 A1
20110230727 Sanders et al. Sep 2011 A1
20110250626 Williams et al. Oct 2011 A1
20110274795 Bogue et al. Nov 2011 A1
20110290694 Fuisz et al. Dec 2011 A1
20120027786 Gupta et al. Feb 2012 A1
20120029832 Dodgson Feb 2012 A1
20120039806 Lahoud et al. Feb 2012 A1
20120058094 Blaser et al. Mar 2012 A1
20120128597 Peters et al. May 2012 A1
20120142548 Corsi et al. Jun 2012 A1
20120148629 Holvoet et al. Jun 2012 A1
20120276143 O'Mahony et al. Nov 2012 A1
20120276149 Littman et al. Nov 2012 A1
20120276525 Kovarik et al. Nov 2012 A1
20120283269 Blagosklonny et al. Nov 2012 A1
20120294822 Russo et al. Nov 2012 A1
20120301452 Gueniche et al. Nov 2012 A1
20130059815 Fournell et al. Mar 2013 A1
20130087155 Hedman et al. Apr 2013 A1
20130157876 Lynch et al. Jun 2013 A1
20130225440 Friedman et al. Aug 2013 A1
20130236488 Dashper et al. Sep 2013 A1
20130252983 Cerione et al. Sep 2013 A1
20130259834 Klaenhammer et al. Oct 2013 A1
20130310416 Blagosklonny Nov 2013 A1
20130315869 Qimron et al. Nov 2013 A1
20130323025 Crawford et al. Dec 2013 A1
20130323100 Poulton et al. Dec 2013 A1
20130326645 Cost et al. Dec 2013 A1
20130330215 Li Dec 2013 A1
20140030332 Baron et al. Jan 2014 A1
20140044677 Qvit-Raz et al. Feb 2014 A1
20140045744 Gordon et al. Feb 2014 A1
20140065209 Putaala et al. Mar 2014 A1
20140065218 Lang et al. Mar 2014 A1
20140066817 Kovarik et al. Mar 2014 A1
20140068797 Doudna et al. Mar 2014 A1
20140125550 Kaneko et al. May 2014 A1
20140154290 Peters et al. Jun 2014 A1
20140199266 Park et al. Jul 2014 A1
20140255351 Berstad et al. Sep 2014 A1
20140271867 Myers et al. Sep 2014 A1
20140294915 Barreca et al. Oct 2014 A1
20140296139 Cohen et al. Oct 2014 A1
20140333003 Allen et al. Nov 2014 A1
20140349405 Sontheimer et al. Nov 2014 A1
20140356460 Lutin Dec 2014 A1
20140363441 Grandea, III et al. Dec 2014 A1
20140364460 Freed-Pastor et al. Dec 2014 A1
20140377278 Elinav et al. Dec 2014 A1
20150004130 Faber et al. Jan 2015 A1
20150017143 Holvoet et al. Jan 2015 A1
20150017227 Kim et al. Jan 2015 A1
20150038594 Borges et al. Feb 2015 A1
20150045546 Siksnys et al. Feb 2015 A1
20150064138 Lu et al. Mar 2015 A1
20150071957 Kelly et al. Mar 2015 A1
20150086581 Li et al. Mar 2015 A1
20150093473 Barrangou et al. Apr 2015 A1
20150132263 Liu et al. May 2015 A1
20150147371 Kovarik et al. May 2015 A1
20150150792 Klingman Jun 2015 A1
20150166641 Goodman et al. Jun 2015 A1
20150190435 Henn et al. Jul 2015 A1
20150202136 Lanzalaco et al. Jul 2015 A1
20150216917 Jones et al. Aug 2015 A1
20150224072 Pellikaan Aug 2015 A1
20150252358 Maeder et al. Sep 2015 A1
20150259728 Cutliffe et al. Sep 2015 A1
20150329555 Liras et al. Nov 2015 A1
20150329875 Gregory et al. Nov 2015 A1
20150352023 Berg et al. Dec 2015 A1
20150353901 Liu et al. Dec 2015 A1
20150361436 Hitchcock et al. Dec 2015 A1
20150374607 Lanzalaco et al. Dec 2015 A1
20160000754 Stamets Jan 2016 A1
20160000841 Yamamoto et al. Jan 2016 A1
20160008412 Putaala et al. Jan 2016 A1
20160024510 Bikard et al. Jan 2016 A1
20160040216 Akins et al. Feb 2016 A1
20160069921 Holmes et al. Mar 2016 A1
20160089315 Kleinberg et al. Mar 2016 A1
20160089405 Lue Mar 2016 A1
20160095316 Goodman et al. Apr 2016 A1
20160120915 Blaser et al. May 2016 A1
20160122806 Amini et al. May 2016 A1
20160151427 Whitlock et al. Jun 2016 A1
20160151428 Bryan Jun 2016 A1
20160158294 Von Maltzahn et al. Jun 2016 A1
20160168594 Zhang et al. Jun 2016 A1
20160175327 Adams et al. Jun 2016 A1
20160199424 Berry et al. Jul 2016 A1
20160206564 Trachtman Jul 2016 A1
20160206666 Falb et al. Jul 2016 A1
20160206668 Kort et al. Jul 2016 A1
20160213702 Von Maltzahn et al. Jul 2016 A1
20160243132 Adams et al. Aug 2016 A1
20160271106 Shi et al. Sep 2016 A1
20160271189 Cutcliffe et al. Sep 2016 A1
20160311913 Sun et al. Oct 2016 A1
20160314281 Apte et al. Oct 2016 A1
20160354416 Gajewski et al. Dec 2016 A1
20160374941 Barreca et al. Dec 2016 A1
20170014341 Armer et al. Jan 2017 A1
20170020932 Cutcliffe et al. Jan 2017 A1
20170027914 Qi Feb 2017 A1
20170042860 Kashyap et al. Feb 2017 A1
20170042924 Otsuka et al. Feb 2017 A1
20170071986 Kovarik et al. Mar 2017 A1
20170079947 Richards Mar 2017 A1
20170100328 Kovarik et al. Apr 2017 A1
20170232043 Falb et al. Aug 2017 A1
20170240625 Zeller et al. Aug 2017 A1
20170246269 Hajishengallis et al. Aug 2017 A1
20170298115 Loomis et al. Oct 2017 A1
20170312232 Vitetta et al. Nov 2017 A1
20170342141 Russo et al. Nov 2017 A1
20170348303 Bosse et al. Dec 2017 A1
20180000878 Goodman et al. Jan 2018 A1
20180015131 Gajewski et al. Jan 2018 A1
20180016647 Van Sinderen et al. Jan 2018 A1
20180092899 Liu et al. Apr 2018 A1
20180100169 Soucaille et al. Apr 2018 A1
20180110795 Frias-Lopez Apr 2018 A1
20180111984 Bigal et al. May 2018 A1
20180127490 Bigal et al. May 2018 A1
20180134772 Sharma et al. May 2018 A1
20180140698 Clube et al. May 2018 A1
20180207165 Harmsen et al. Jul 2018 A1
20180235987 Von Maltzahn et al. Aug 2018 A1
20180258100 Gregory et al. Sep 2018 A1
20180296582 von Maltzahn et al. Oct 2018 A1
20180303658 Kovarik et al. Oct 2018 A1
20180312851 Falb et al. Nov 2018 A1
20180326008 Schreiber et al. Nov 2018 A1
20180371405 Barrangou et al. Dec 2018 A1
20190000815 Melin Jan 2019 A1
20190018012 Kovarik Jan 2019 A1
20190059314 Aharoni et al. Feb 2019 A1
20190290605 Rasochova et al. Jun 2019 A1
20190120960 Konradi et al. Jul 2019 A1
20190262298 Kanthasamy et al. Aug 2019 A1
20190315642 Parsley et al. Oct 2019 A1
20190388471 June et al. Dec 2019 A1
20190390284 Kim Dec 2019 A1
20200009185 Shin et al. Jan 2020 A1
20200009268 Scholz Jan 2020 A1
20200032224 Schaefer et al. Jan 2020 A1
20200148642 Konradi et al. May 2020 A1
20200155447 Edwards May 2020 A1
20200188454 Slykerman Jun 2020 A1
20200190494 Hou et al. Jun 2020 A1
20200197215 Kovarik et al. Jun 2020 A1
20200199555 Zhang Jun 2020 A1
20210169954 Balani et al. Jun 2021 A1
20210198665 Sommer et al. Jul 2021 A1
20210308028 Yang et al. Oct 2021 A1
20210321756 McLaughlin et al. Oct 2021 A1
20210361560 Krueger et al. Nov 2021 A1
20210386659 Kim Dec 2021 A1
20220000760 Rasochova Jan 2022 A1
20220023259 Davidson et al. Jan 2022 A1
20220031590 Pesaro et al. Feb 2022 A1
20220031767 Duportet et al. Feb 2022 A1
20220071877 Zenobia et al. Mar 2022 A1
20220088001 Kovarik et al. Mar 2022 A1
20220088090 Lobacki et al. Mar 2022 A1
20220118031 Kovarik Apr 2022 A1
20220135987 Leveau et al. May 2022 A1
20220193150 Kovarik Jun 2022 A1
20220193157 Zimmerman et al. Jun 2022 A1
20220257410 Kovarik Aug 2022 A1
20220296500 Kovarik Sep 2022 A1
20220331374 Richter et al. Oct 2022 A1
20220339208 Abel et al. Oct 2022 A1
20220387402 Aspnes et al. Dec 2022 A1
20230040879 Kovarik Feb 2023 A1
20230106721 Catania et al. Apr 2023 A1
20230131201 Kovarik Apr 2023 A1
20230165706 Tye et al. Jun 2023 A1
20230218682 Tye et al. Jul 2023 A1
20230241129 Simmons et al. Aug 2023 A1
Foreign Referenced Citations (25)
Number Date Country
4412190 Oct 1995 DE
410696 Jan 1991 EP
S56-100714 Aug 1981 JP
WO 9822097 May 1998 WO
WO 2006007922 Jan 2006 WO
WO 2006015445 Feb 2006 WO
WO 2006133879 Dec 2006 WO
WO 2008088426 Jul 2008 WO
WO 2008097890 Aug 2008 WO
WO 200905242 Apr 2009 WO
WO 2010041143 Apr 2010 WO
WO 2011020780 Feb 2011 WO
WO 2011029701 Mar 2011 WO
WO 2013026000 Feb 2013 WO
WO 2013107750 Jul 2013 WO
WO 2013182038 Dec 2013 WO
WO 2014103488 Jul 2014 WO
WO 2014152338 Sep 2014 WO
WO 2014182632 Nov 2014 WO
WO 2014196913 Dec 2014 WO
WO 2015069682 May 2015 WO
WO 2016070151 May 2016 WO
WO 2017211753 Dec 2017 WO
WO 2019067621 Apr 2019 WO
WO 2022187274 Sep 2022 WO
Non-Patent Literature Citations (193)
Entry
U.S. Appl. No. 17/854,389, filed Jun. 30, 2022, Kovarik.
U.S. Appl. No. 18/143,399, filed May 4, 2023, Kovarik.
U.S. Appl. No. 18/232,433, filed Aug. 10, 2023, Simmons et al.
U.S. Appl. No. 18/232,980, filed Aug. 11, 2023, Kovarik.
“Oral Cavity,” University of Michigan Medical School, Date Unknown, retrieved Nov. 20, 2019 from https://histology.medicine.umich.edu/resources/oral-cavity, 5 pages.
“The structure behind the simplicity of CRISPR/Cas9,” The Scinder at Medium.com, Dec. 23, 2015, retrieved from https://medium.com/the-scinder/the-structure-behind-the-simplicity-of-crispr-cas9-6f8cb60695c4, 8 pages.
Abruzzo et al., “Influence of Lactobacillus Biosurfactants on Skin Permeation of Hydrocortisone,” Pharmaceutics, vol. 13, No. 6, May 2021, 14 pages.
Agrawal et al., “Technique to Control pH in Vicinity of Biodegrading PLA-PGA Implants,” Journal of Biomedical Materials Research, vol. 38, No. 2, 1997, pp. 105-114.
Aguilar-Toala et al., “Potential role of natural bioactive peptides for development of cosmeceutical skin products,” Peptides, vol. 122, No. 170170, Dec. 2019, 8 pages. Abstract only.
Athanasiou et al., “In Vitro Degradation and Release Characteristics of Biodegradable Implants Containing Trypsin Inhibitor,” Clinical Orthopaedics and Related Research, vol. 315, Jun. 1995, pp. 272-281. Abstract only.
Auerbach et al., “Angiogenesis assays: Problems and pitfalls,” Cancer and Metastasis Reviews, vol. 19, 2000, pp. 167-172.
Basseri et al., “Antibiotics for the Treatment of Irritable Bowel Syndrome,” Gastroenterology & Hepatology, vol. 7, No. 7, Jul. 2011, pp. 455-493.
Baud et al., “Microbial diversity in the vaginal microbiota and its link to pregnancy outcomes,” Scientific Reports, vol. 13, No. 9061, 2023, 12 pages.
Blumen et al., “Radiofrequency Ablation for the Treatment of Mild to Moderate Obstructive Sleep Apnea,” The Laryngoscope, vol. 112, No. 11, Nov. 2002, pp. 2086-2092.
Bocheva et al., “Protective Role of Melatonin and Its Metabolites in Skin Aging,” International Journal of Molecular Sciences, vol. 23, No. 1238, Jan. 2022, 23 pages.
Brietzke et al., “Injection Snoreplasty: Extended Follow-Up and New Objective Data,” Otolaryngology-Head and Neck Surgery, vol. 128, No. 5, May 2003, pp. 605-615. Abstract only.
Brietzke et al., “Injection Snoreplasty: How to Treat Snoring without All the Pain and Expense,” Otolaryngology—Head and Neck Surgery, vol. 124, No. 5, May 2001, pp. 503-510. Abstract only.
Brietzke et al., “Injection Snoreplasty: Investigation of Alternative Sclerotherapy Agents,” Otolaryngology—Head and Neck Surgery, vol. 130, No. 1, Jan. 2004, pp. 47-57. Abstract only.
Brown et al., “Improving the Diagnosis of Vulvovaginitis: Perspectives to Align Practice, Guidelines, and Awareness,” Population Health Management, vol. 23, Suppl. 1, 2020, pp. S3-S12.
Catalano et al., “Additional palatal implants for refractory snoring,” Otolaryngology—Head and Neck Surgery, vol. 137, No. 1, Jul. 2007, pp. 105-109. Abstract only.
Charulatha et al., “Influence of different crosslinking treatments on the physical properties of collagen membranes,” Biomaterials, vol. 24, No. 5, 2003, pp. 759-767.
Chen et al., “Targeting Aldehyde Dehydrogenase 2: New Therapeutic Opportunities,” Physiological Reviews, vol. 94, No. 1, 2014, 65 pages.
Choi et al., “Therapeutic Effects of Cold-Pressed Perilla Oil Mainly Consisting of Linolenic acid, Oleic Acid and Linoleic Acid on UV-Induced Photoaging in NHDF Cells and SKH-1 Hairless Mice,” Molecules, vol. 25, Feb. 2020, 19 pages.
Chuang et al., “Effects of exogenous crosslinking on in vitro tensile and compressive moduli of lumbar intervertebral discs,” Clinical Biomechanics, vol. 22, No. 1, Jan. 2007, pp. 14-20. Abstract only.
Courage, “Fiber-Famished Gut Microbes Linked to Poor Health,” Scientific American, Mar. 23, 2015, retrieved fromhttps://www.scientificamerican.com/article/fiber-famished-gut-microbes-linked-to-poor-health, 10 pages.
De Seta et al., “The Vaginal Community State Types Microbiome-Immune Network as Key Factor for Bacterial Vaginosis and Aerobic Vaginitis,” Frontiers in Microbiology, vol. 10, No. 2451, Oct. 30, 2019, 8 pages.
Ding et al., “Resveratrol accelerates wound healing by inducing M2 macrophage polarisation in diabetic mice,” Pharmaceutical Biology, vol. 60, No. 1, 2022, pp. 2328-2337.
Douam et al., “Genetic Dissection of the Host Tropism of Human-Tropic Pathogens,” Annual Review of Genetics, vol. 49, 2015, pp. 21-45.
Dunkley et al., “A role for CD4+ T cells from orally immunized rats in enhanced clearance of Pseudomonas aeruginosa from the lung,” Immunology, vol. 83, 1994, pp. 362-369.
Earlia et al., “GC/MS Analysis of Fatty Acids on Pliek U Oil and Its Pharmacological Study by Molecular Docking to Filaggrin as a Drug Candidate in Atopic Dermatitis Treatment,” Scientific World Journal, Nov. 2019, 7 pages.
Enomoto et al., “Koji amazake Maintains Water Content in the Left Cheek Skin of Healthy Adults: A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Comparative Trial,” Clinical, Cosmetic and Investigational Dermatology, vol. 15, Jul. 2022, pp. 1283-1291.
Farhadihosseinabadi et al., “The in vivo effect of Lacto-N-neotetraose (LNnT) on the expression of type 2 immune response involved genes in the wound healing process,” Scientific Reports, vol. 10, No. 997, Jan. 2020, 11 pages.
Fischer et al., “[Radiofrequency ablation of the soft palate (somnoplasty). A new method in the treatment of habitual and obstructive snoring].,” HNO, vol. 48, No. 1, Jan. 2000, pp. 33-40. Abstract only.
Friedman et al., “Patient Selection and Efficacy of Pillar Implant Technique for Treatment of Snoring and Obstructive Sleep Apnea/Hypopnea Syndrome,” Otolaryngology—Head and Neck Surgery, vol. 134, No. 2, Feb. 2006, pp. 187-196. Abstract only.
Gajer et al., “Temporal Dynamics of the Human Vaginal Microbiota,” Science Translational Medicine, vol. 4, No. 132, May 2, 2012, 21 pages.
Gratzer et al., “Control of pH Alters the Type of Cross-linking Produced by 1-Ethyl-3-(3-Dimethylaminopropyl)-Carbodiimide (EDC) Treatment of Acellular Matrix Vascular Grafts,” Journal of Biomedical Materials Research, vol. 58, No. 2, 2001, pp. 172-179.
Guilleminault et al., “Snoring (I). Daytime sleepiness in regular heavy snorers,” Chest, vol. 99, 1991, pp. 40-48.
Guilleminault et al., “The sleep apnea syndromes,” Annual Review of Medicine, vol. 27, Feb. 1976, pp. 465-484. First Page Only.
Gura, “Systems for Identifying New Drugs Are Often Faulty,” Science, vol. 278, No. 5340, Nov. 7, 1997, pp. 1041-1042.
Han et al., “Proanthocyanidin: A natural crosslinking reagent for stabilizing collagen matrices,” Journal of Biomedical Materials Research, vol. 65A, No. 1, Apr. 2003, pp. 118-124. Abstract only.
Hedman et al., “Exogenous Cross-Linking Increases the Stability of Spinal Motion Segments,” Spine, vol. 31, No. 15, Jul. 2006, pp. E480-E485. Abstract only.
Hennessy et al., “Statins as next generation anti-microbials: Is there potential for repurposing?,” Antimicrob. Agents Chemother., Jun. 20, 2016, 46 pages.
Hildebrand et al., “Vaginitis,” NCBI Bookshelf, Updated Nov. 14, 2022, 12 pages.
Hoffmann et al., “Glutaraldehyde and oxidised dextran as crosslinker reagents for chitosan-based scaffolds for cartilage tissue engineering,” Journal of Materials Science: Materials in Medicine, vol. 20, Mar. 2009, pp. 1495-1503.
Hunter et al., “Meniscal material properties are minimally affected by matrix stabilization using glutaraldehyde and glycation with ribose,” Journal of Orthopaedic Research, vol. 23, 2005, pp. 555-561.
Jain, “Barriers to Drug Delivery in Solid Tumors,” Scientific American, Jul. 1994, pp. 58-65.
Kilkkinen et al., “Use of antimicrobials and risk of type 1 diabetes in a population-based mother-child cohort,” Diabetologia, vol. 49, 2006, pp. 66-70.
Kim et al., “Kaempferol tetrasaccharides restore skin atrophy via PDK1 inhibition in human skin cells and tissues: Bench and clinical studies,” Biomedicine & Pharmacotherapy, vol. 156, No. 113864, Dec. 2022, 13 pages.
Kim et al., “Spermidine-induced recovery of human dermal structure and barrier function by skin microbiome,” Communications Biology, vol. 4, No. 231, 2021, 11 pages.
Kim et al., “β-Glucogallin isolated from Fusidium coccineum and its enhancement of skin barrier effects,” Applied Biological Chemistry, vol. 63, No. 77, Nov. 2020, 7 pages.
Kimoto et al., “New Lactococcus Strain with Immunnomodulatory Activity: Enhancement of Th1-Type Immune Response,” Microbiol. Immunol., vol. 48, No. 2, 2004, pp. 75-82.
Klapperich et al., “A novel biocompatible adhesive incorporating plant-derived monomers,” Journal of Biomedical Materials Research Part A, vol. 91, No. 2, pp. 378-374.
Klingspor et al., “Research Article: Enterococcus faecium NCIMB 10415 Modulates Epithelial Integrity, Heat Shock Protein, and Proinflammatory Cytokine Response in Intestinal Cells,” Mediators of Inflammation, vol. 2015, No. 304149 ,2015, 12 pages.
Ko, “Effects of Glycogen on Ceramide Production in Cultured Human Keratinocytes via Acid Sphingomyelinase Activation,” Master's Thesis Submitted to the Graduate School of Public Health (Korea), 2018, 53 pages.
Komuro et al., “Sphingomyelin maintains the cutaneous barrier via regulation of the STAT3 pathway,” The FASEB Journal, vol. 36, No. 4, Apr. 2022, 17 pages.
Kurek-Gorecka et al., “Bee Products in Dermatology and Skin Care,” Molecules, vol. 25, No. 3, Jan. 2020, 17 pages.
Kyriakopoulos et al., “Taurine and N-Bromotaurine in Topical Treatment of Psoriasis,” Advances in Experimental Medicine and Biology, vol. 1370, 2022, pp. 99-111. Abstract only.
Laneri et al., “Plant cell culture extract of Cirsium eriophorum with skin pore refiner activity by modulating sebum production and inflammatory response,” Phytotherapy Research, vol. 35, No. 1, Jan. 2021, pp. 530-540.
Lebeer et al., “Selective targeting of skin pathobionts and inflammation with topically applied lactobacilli,” Cell Reports Medicine, vol. 3, No. 2, Feb. 2022, 22 pages.
Lenger et al., “D-mannose vs other agents for recurrent urinary tract infection prevention in adult women: a systematic review and meta-analysis,” American Journal of Obstetrics and Gynecology, vol. 223, No. 2, Aug. 2020, pp. 265.e1-265.e13.
Lew et al., “Bioactives from probiotics for dermal health: functions and benefits,” Journal of Applied Microbiology, vol. 114, No. 5, May 2013, pp. 1241-1253.
Lewis et al., “Vaginal Microbiome and Its Relationship to Behavior, Sexual Health, and Sexually Transmitted Diseases,” Obstetrics & Gynecology, vol. 129, No. 4, Apr. 2017, pp. 643-654.
Liu et al., “Activation of aryl hydrocarbon receptor in Langerhans cells by a microbial metabolite of tryptophan negatively regulates skin inflammation,” Journal of Dermatological Science, vol. 100, No. 3, Dec. 2020, pp. 192-200. Abstract only.
Liu et al., “The potential of Streptococcus thermophiles (TCI633) in the anti-aging,” Journal of Cosmetic Dermatology, vol. 21, No. 6, Jun. 2022, pp. 2635-2647.
Ma et al., “The vaginal microbiome: rethinking health and diseases,” Annual Review of Microbiology, vol. 66, 2012, pp. 371-389.
Mach et al., “Endurance exercise and gut microbiota: A review,” Journal of Sport and Health Science, vol. 6, No. 2, Jun. 2017, pp. 179-197.
Mahdiani et al., “Protective effect of luteolin against chemical and natural toxicants by targeting NF-κB pathway,” Biofactors, vol. 48, No. 4, Jul. 2022, pp. 744-762. Abstract only.
Malaguarnera et al., “Bifidobacterium longum with Fructo-Oligosaccharides in Patients with Non Alcoholic Steatohepatitis,” Digestive Diseases and Sciences, vol. 57, 2012, pp. 545-553.
Matsui et al., “Biological Rhythms in the Skin,” International Journal of Molecular Sciences, vol. 17, No. 801, May 2016, 15 pages.
Mayrovitz et al., “Assessing Potential Circadian, Diurnal, and Ultradian Variations in Skin Biophysical Properties,” Cureus, vol. 13, No. 9, Sep. 2021, 18 pages.
McFadzean, “Exercise can help modulate human gut microbiota,” Honors Thesis Submitted to the University of Colorado Department of Evolutionary Biology, Apr. 7, 2014, 34 pages.
Nakai et al., “Effects of Topical N-Acetylcysteine on Skin Hydration/Transepidermal Water Loss in Healthy Volunteers and Atopic Dermatitis Patients,” Annals of Dermatology, vol. 27, No. 4, Aug. 2015, pp. 450-451.
Neves et al., “Efficacy of a topical serum containing L-ascorbic acid, neohesperidin, pycnogenol, tocopherol, and hyaluronic acid in relation to skin aging signs,” Journal of Cosmetic Dermatology, vol. 21, No. 10, Oct. 2022, pp. 4462-4469. Abstract only.
Nisbet et al., “Clinical and in vitro evaluation of new anti-redness cosmetic products in subjects with winter xerosis and sensitive skin,” International Journal of Cosmetic Science, vol. 41, No. 6, Dec. 2019, pp. 534-547.
Norton et al., “The immune response to Lactococcus lactis: Implications for its use as a vaccine delivery vehicle,” FEMS Microbiology Letters, vol. 120, No. 3, Jul. 15, 1994, pp. 249-256. Abstract only.
O'Hanlon et al., “In vaginal fluid, bacteria associated with bacterial vaginosis can be suppressed with lactic acid but not hydrogen peroxide,” BMC Infectious Diseases, vol. 11, No. 200, 2011, 8 pages.
Paladine et al., “Vaginitis: Diagnosis and Treatment,” American Family Physician, vol. 97, No. 5, Mar. 1, 2018, pp. 321-329.
Park et al., “Fermented black rice and blueberry with Lactobacillus plantarum MG4221 improve UVB-induced skin injury,” Food and Agricultural Immunology, vol. 32, No. 1, 2021, pp. 499-515.
Pinto et al., “Plantaricin A synthesized by Lactobacillus plantarum induces in vitro proliferation and migration of human keratinocytes and increases the expression of TGF-β1, FGF7, VEGF-A and IL-8 genes,” Peptides, vol. 32, No. 9, Sep. 2011, pp. 1815-1824. Abstract only.
Ragusa et al., “Spirulina for Skin Care: A Bright Blue Future,” Cosmetics, vol. 8, No. 1, Jan. 2021, 19 pages.
Ravel et al., “Vaginal microbiome of reproductive-age women,” PNAS, vol. 108, Suppl. 1, Mar. 15, 2011, pp. 4680-4687.
Repa et al., “Mucosal co-application of lactic acid bacteria and allergen induces counter-regulatory immune responses in a murine model of birch pollen allergy,” Vaccine, vol. 22, No. 1, 2003, pp. 87-95. Abstract only.
Scaglione et al., “Considerations on D-mannose Mechanism of Action and Consequent Classification of Marketed Healthcare Products,” Frontiers In Pharmacology, vol. 12, No. 636377, Mar. 2, 2021, 7 pages.
Schaeffer et al., “Effect of Carbohydrates on Adherence of Escherichia coli to Human Urinary Tract Epithelial Cells,” Infection and Immunity, vol. 30, No. 2, Nov. 1980, pp. 531-537.
Sevilla et al., “Revisiting the role of melatonin in human melanocyte physiology: A skin context perspective,” Journal of Pineal Research, vol. 72, No. 3, Apr. 2022, 23 pages.
Sheikh, “Is Crispr the Next Antibiotic?,” The New York Times, Oct. 29, 2019, retrieved from https://www.nytimes.com/2019/28/health/crispr-genetics-antibiotic-resistance.html, 2 pages.
Shen et al., “Propionibacterium acnes related anti-inflammation and skin hydration activities of madecassoside, a pentacyclic triterpene saponin from Centella asiatica,” Bioscience, Biotechnology, and Biochemistry, vol. 83, No. 3, 2019, pp. 561-568.
Sheweita et al., “Preclinical studies on melanogenesis proteins using a resveratrol-nanoformula as a skin whitener,” International Journal of Biological Macromolecules, vol. 223, Part A, Dec. 2022, pp. 870-881. Abstract only.
Simmering et al., “The Increase in Hospitalizations for Urinary Tract Infections and the Associated Costs in the United States, 1998-2011,” Open Forum Infectious Diseases, vol. 4, No. 1, Feb. 24, 2017, 7 pages.
Sivieri et al., “Lactobacillus acidophilus CRL 1014 improved “gut health” in the SHIME reactor,” BMC Gastroenterology, vol. 13, No. 100, 2013, 9 pages.
Spinler et al., “Human-derived probiotic Lactobacillus reuteri demonstrate antimicrobial activities targeting diverse enteric bacterial pathogens,” Anaerobe, vol. 14, Feb. 29, 2008, pp. 166-171.
Sporn et al., “Chemoprevention of cancer,” Carcinogenesis, vol. 21, No. 3, 2000, pp. 525-530.
Thongaram et al., “Human milk oligosaccharide consumption by probiotic and human-associated bifidobacteria and lactobacilli,” Journal of Dairy Science, vol. 100, No. 10, Oct. 2017, pp. 7825-7833.
Traisaeng et al., “A Derivative of Butyric Acid, the Fermentation Metabolite of Staphylococcus epidermidis, Inhibits the Growth of a Staphylococcus aureus Strain Isolated from Atopic Dermatitis Patients,” Toxins, vol. 11, No. 6, May 2019, 12 pages.
Van Der Veer et al., “Comparative genomics of human Lactobacillus crispatus isolates reveals genes for glycosylation and glycogen degradation: implications for in vivo dominance of the vaginal microbiota,” Microbiome, vol. 7, No. 49, 2019, 14 pages.
Van Hemert et al., “Migraine associated with gastrointestinal disorders: review of the literature and clinical implications,” Frontiers in Neurology, vol. 5, No. 241, Nov. 2014, 4 pages.
Wan et al., “Luteolin-7-glucoside Promotes Human Epidermal Stem Cell Proliferation by Upregulating β-Catenin, c-Myc, and Cyclin Expression,” Stem Cells International, vol. 2019, No. 1575480, Jun. 2019, 10 pages.
Wilbie et al., “Delivery Aspects of CRISPR/Cas for in Vivo Genome Editing,” Accounts of Chemical Research, vol. 52, 2019, pp. 1555-1564.
Yamamura et al., “Oral mucosal adhesive Film containing local anesthetics: in vitro and clinical evaluation.” Journal of Biomedical Materials Research, Fall 1998, vol. 43, No. 3, pp. 313-317. Abstract only.
Yatsuhashi et al., “Effects of Glycogen on Ceramide Production in Cultured Human Keratinocytes via Acid Sphingomyelinase Activation,” Journal of Applied Glycoscience, vol. 68, 2021, pp. 41-46.
Yosipovitch et al., “Time-Dependent Variations of the Skin Barrier Function in Humans: Transepidermal Water Loss, Stratum Corneum Hydration, Skin Surface pH, and Skin Temperature,” Journal of INvestigative Dermatology, vol. 110, No. 1, Jan. 1998, pp. 20-23.
Zahedi et al., “Development of plasma functionalized polypropylene wound dressing for betaine hydrochloride controlled drug delivery on diabetic wounds,” Scientific Reports, vol. 11, No. 9641, 2021, 18 pages.
Zhao et al., “Microbiome-generated amyloid and potential impact on amyloidogenesis in Alzheimer's disease (AD),” Journal of Nature and Science, vol. 1, No. 7, 2015, pp. 1-5.
Zhou et al., “Nicotinamide Mononucleotide Combined With Lactobacillus fermentum TKSN041 Reduces the Photoaging Damage in Murine Skin by Activating AMPK Signaling Pathway,” Frontiers in Pharmacology, vol. 12, No. 643089, Mar. 2021, 17 pages.
Official Action for U.S. Appl. No. 14/574,517 dated Jan. 6, 2016, 13 pages.
Notice of Allowance for U.S. Appl. No. 14/574,517, dated Apr. 15, 2016, 8 pages.
Corrected Notice of Allowance for U.S. Appl. No. 14/574,517, dated Jul. 7, 2016, 2 pages.
Official Action for U.S. Appl. No. 14/954,074, dated Jun. 30, 2016, 4 pages.
Notice of Allowance for U.S. Appl. No. 14/954,074, dated Jul. 20, 2016, 7 pages.
Official Action for U.S. Appl. No. 15/270,034, dated Apr. 6, 2017, 5 pages.
Notice of Allowance for U.S. Appl. No. 15/270,034, dated May 5, 2017, 7 pages.
Official Action for U.S. Appl. No. 15/392,173, dated Jan. 22, 2018, 8 pages.
Official Action for U.S. Appl. No. 15/392,173, dated Jul. 6, 2018, 13 pages.
Notice of Allowance for U.S. Appl. No. 15/392,173, dated Dec. 5, 2018, 8 pages.
Official Action for U.S. Appl. No. 16/229,252, dated Feb. 28, 2019, 5 pages.
Notice of Allowance for U.S. Appl. No. 16/229,252, dated Aug. 21, 2019, 7 pages.
Official Action for U.S. Appl. No. 16/722,117, dated Feb. 20, 2020, 6 pages.
Notice of Allowance for U.S. Appl. No. 16/722,117, dated Jul. 30, 2020, 8 pages.
Official Action for U.S. Appl. No. 17/011,175, dated Jun. 17, 2021, 9 pages.
Notice of Allowance for U.S. Appl. No. 17/011,175, dated Nov. 5, 2021, 8 pages.
Official Action for U.S. Appl. No. 17/023,736, dated Nov. 10, 2021, 7 pages.
Notice of Allowance for U.S. Appl. No. 17/023,736, dated Apr. 14, 2022, 8 pages.
Official Action for U.S. Appl. No. 17/893,384, dated May 9, 2023, 8 pages.
Notice of Allowance for U.S. Appl. No. 17/893,384, dated Aug. 23, 2023, 7 pages.
Official Action for U.S. Appl. No. 15/403,823, dated Oct. 30, 2017, 7 pages.
Official Action for U.S. Appl. No. 15/403,823, dated May 25, 2018, 9 pages.
Notice of Allowance for U.S. Appl. No. 15/403,823, dated Jun. 28, 2018, 9 pages.
Official Action for U.S. Appl. No. 16/160,336, dated Nov. 27, 2018, 6 pages.
Notice of Allowance for U.S. Appl. No. 16/160,336, dated Feb. 15, 2019, 7 pages.
Official Action for U.S. Appl. No. 16/423,375, dated Jul. 3, 2019, 6 pages.
Notice of Allowance for U.S. Appl. No. 16/423,375, dated Oct. 16, 2019, 8 pages.
Official Action for U.S. Appl. No. 16/782,364, dated Apr. 9, 2020, 5 pages.
Notice of Allowance for U.S. Appl. No. 16/782,364, dated Jul. 27, 2020, 7 pages.
Official Action for U.S. Appl. No. 16/917,096, dated Jul. 31, 2020, 5 pages.
Official Action for U.S. Appl. No. 16/617,096, dated Oct. 19, 2020, 8 pages.
Official Action for U.S. Appl. No. 17/027,953, dated Jan. 29, 2021, 5 pages.
Notice of Allowance for U.S. Appl. No. 17/027,953, dated Apr. 19, 2021, 8 pages.
Official Action for U.S. Appl. No. 17/337,600, dated Jul. 6, 2021, 5 pages.
Notice of Allowance for U.S. Appl. No. 17/337,600, dated Sep. 9, 2021, 7 pages.
Official Action for U.S. Appl. No. 17/835,204, dated Jul. 28, 2022, 6 pages.
Notice of Allowance for U.S. Appl. No. 17/835,204, dated Aug. 24, 2022, 7 pages.
Official Action for U.S. Appl. No. 17/848,759, dated Sep. 14, 2022, 6 pages.
Notice of Allowance for U.S. Appl. No. 17/848,759, dated Dec. 29, 2022, 7 pages.
Corrected Notice of Allowance for U.S. Appl. No. 17/848,759, dated Jan. 12, 2023, 4 pages.
Official Action for U.S. Appl. No. 17/854,422, dated Sep. 28, 2022, 7 pages.
Official Action for U.S. Appl. No. 17/854,422, dated Jan. 10, 2023, 6 pages.
Notice of Allowance for U.S. Appl. No. 17/854,422, dated Feb. 17, 2023, 7 pages.
Official Action for U.S. Appl. No. 18/087,545, dated May 24, 2023, 5 pages.
Notice of Allowance for U.S. Appl. No. 18/087,545, dated Jul. 26, 2023, 7 pages.
Official Action for U.S. Appl. No. 18/178,847, dated Jul. 13, 2023, 8 pages.
Notice of Allowance for U.S. Appl. No. 18/178,847, dated Aug. 8, 2023, 8 pages.
Official Action for U.S. Appl. No. 18/130,946, dated Jun. 30, 2023, 6 pages.
Notice of Allowance for U.S. Appl. No. 18/130,946, dated Aug. 1, 2023, 8 pages.
Official Action for U.S. Appl. No. 15/228,454, dated Sep. 23, 2016, 11 pages.
Notice of Allowance for U.S. Appl. No. 15/228,454, dated Jan. 23, 2017, 7 pages.
Official Action for U.S. Appl. No. 15/437,976, dated Mar. 29, 2017, 8 pages.
Notice of Allowance for U.S. Appl. No. 15/437,976, dated Jul. 12, 2017, 7 pages.
Official Action for U.S. Appl. No. 15/639,767, dated Aug. 14, 2017, 11 pages.
Official Action for U.S. Appl. No. 15/639,767, dated Sep. 27, 2018, 13 pages.
Notice of Allowance for U.S. Appl. No. 15/369,767, dated Feb. 15, 2019, 8 pages.
Official Action for U.S. Appl. No. 16/426,346, dated Aug. 2, 2019, 10 pages.
Official Action for U.S. Appl. No. 16/426,346, dated Jan. 13, 2020, 7 pages.
Notice of Allowance for U.S. Appl. No. 16/426,346, dated Mar. 25, 2020, 7 pages.
Official Action for U.S. Appl. No. 13/367,052, dated Jan. 16, 2014, 8 pages.
Notice of Allowance for U.S. Appl. No. 13/367,052, dated Feb. 24, 2014, 5 pages.
Official Action for U.S. Appl. No. 14/225,503, dated May 4, 2016, 6 pages.
Notice of Allowance for U.S. Appl. No. 14/225,503, dated Jul. 20, 2016, 5 pages.
Official Action for U.S. Appl. No. 14/752,192, dated Jul. 8, 2016, 8 pages.
Notice of Allowance for U.S. Appl. No. 14/752,192, dated Sep. 16, 2016, 5 pages.
Official Action for U.S. Appl. No. 15/378,425, dated May 15, 2019, 82 pages.
Official Action for U.S. Appl. No. 15/378,425, dated Oct. 2, 2019, 41 pages.
Official Action for U.S. Appl. No. 15/378,425, dated Jul. 15, 2020, 21 pages.
Official Action for U.S. Appl. No. 15/378,425, dated Nov. 10, 2020, 29 pages.
Official Action for U.S. Appl. No. 15/385,278, dated Oct. 30, 2017, 23 pages.
Official Action for U.S. Appl. No. 15/385,278, dated Apr. 13, 2018, 18 pages.
Notice of Allowance for U.S. Appl. No. 15/385,278, dated May 31, 2018, 10 pages.
Official Action for U.S. Appl. No. 16/136,950, dated Nov. 25, 2019, 11 pages.
Official Action for U.S. Appl. No. 16/136,950, dated Jan. 31, 2020, 8 pages.
Official Action for U.S. Appl. No. 16/884,772, dated Sep. 30, 2021, 8 pages.
Notice of Allowance for U.S. Appl. No. 16/884,772, dated Feb. 22, 2022, 7 pages.
Official Action for U.S. Appl. No. 15/384,716, dated Nov. 1, 2017, 31 pages.
Notice of Allowance for U.S. Appl. No. 15/384,716, dated Apr. 2, 2018, 9 pages.
Official Action for U.S. Appl. No. 15/983,250, dated Mar. 5, 2019, 23 pages.
Official Action for U.S. Appl. No. 15/983,250, dated May 24, 2019, 21 pages.
Official Action for U.S. Appl. No. 15/983,250, dated Jan. 14, 2020, 8 pages.
Notice of Allowance for U.S. Appl. No. 15/983,250, dated Feb. 14, 2020, 8 pages.
Official Action for U.S. Appl. No. 16/904,056, dated Dec. 6, 2021, 12 pages.
Official Action for U.S. Appl. No. 16/904,056, dated May 17, 2022, 11 pages.
Notice of Allowance for U.S. Appl. No. 16/904,056, dated Aug. 11, 2022, 8 pages.
Corrected Notice of Allowance for U.S. Appl. No. 16/904,056, dated Aug. 24, 2022, 6 pages.
Official Action for U.S. Appl. No. 18/103,768, dated Apr. 25, 2023, 5 pages.
Notice of Allowance for U.S. Appl. No. 18/103,768, dated Aug. 1, 2023, 7 pages.
Official Action for U.S. Appl. No. 18/143,399, dated Sep. 7, 2023, 8 pages.
Related Publications (1)
Number Date Country
20240024382 A1 Jan 2024 US
Provisional Applications (10)
Number Date Country
62072476 Oct 2014 US
62053926 Sep 2014 US
62014855 Jun 2014 US
61919297 Dec 2013 US
62275341 Jan 2016 US
62296186 Feb 2016 US
62387405 Dec 2015 US
62387404 Dec 2015 US
62260906 Nov 2015 US
62274550 Jan 2016 US
Continuations (7)
Number Date Country
Parent 17893384 Aug 2022 US
Child 18143399 US
Parent 16160336 Oct 2018 US
Child 16423375 US
Parent 15403823 Jan 2017 US
Child 16160336 US
Parent 17836079 Jun 2022 US
Child 18234132 US
Parent 15385278 Dec 2016 US
Child 16136950 US
Parent 16142171 Sep 2018 US
Child 16776861 US
Parent 15639767 Jun 2017 US
Child 16426346 US
Continuation in Parts (43)
Number Date Country
Parent 18232980 Aug 2023 US
Child 18234132 US
Parent 18232433 Aug 2023 US
Child 18232980 US
Parent 18143399 May 2023 US
Child 18232433 US
Parent 17694775 Mar 2022 US
Child 17893384 US
Parent 17023736 Sep 2020 US
Child 17694775 US
Parent 17011175 Sep 2020 US
Child 17023736 US
Parent 16722117 Dec 2019 US
Child 17011175 US
Parent 16229252 Dec 2018 US
Child 16722117 US
Parent 15392173 Dec 2016 US
Child 16229252 US
Parent 18130946 Apr 2023 US
Child 18234132 US
Parent 18178847 Mar 2023 US
Child 18130946 US
Parent 18087545 Dec 2022 US
Child 18178847 US
Parent 17854422 Jun 2022 US
Child 18087545 US
Parent 17848759 Jun 2022 US
Child 17854422 US
Parent 17835204 Jun 2022 US
Child 17848759 US
Parent 17567295 Jan 2022 US
Child 17835204 US
Parent 17337600 Jun 2021 US
Child 17567295 US
Parent 17027953 Sep 2020 US
Child 17337600 US
Parent 16917096 Jun 2020 US
Child 17027953 US
Parent 16782364 Feb 2020 US
Child 16917096 US
Parent 16423375 May 2019 US
Child 16782364 US
Parent 18103768 Jan 2023 US
Child 18234132 US
Parent 17738771 May 2022 US
Child 18103768 US
Parent 16904056 Jun 2020 US
Child 17738771 US
Parent 15983250 May 2018 US
Child 16904056 US
Parent 15384716 Dec 2016 US
Child 15983250 US
Parent 16884772 May 2020 US
Child 17836079 US
Parent 16136950 Sep 2018 US
Child 16884772 US
Parent 17543992 Dec 2021 US
Child 18234132 US
Parent 16804361 Feb 2020 US
Child 17543992 US
Parent 16020433 Jun 2018 US
Child 16804361 US
Parent 15342642 Nov 2016 US
Child 16020433 US
Parent 16776861 Jan 2020 US
Child 18234132 US
Parent 15395419 Dec 2016 US
Child 16142171 US
Parent 16426346 May 2019 US
Child 18234132 US
Parent 15437976 Feb 2017 US
Child 15639767 US
Parent 15228454 Aug 2016 US
Child 15437976 US
Parent 14954074 Nov 2015 US
Child 15228454 US
Parent 15270034 Sep 2016 US
Child 18234132 US
Parent 14954074 Nov 2015 US
Child 15270034 US
Parent 14574517 Dec 2014 US
Child 14954074 US
Parent 16037053 Jul 2018 US
Child 18234132 US
Parent 14752192 Jun 2015 US
Child 16037053 US