When treating hyperkalemia, insulin remains efficacious in diabetics and nondiabetics and one does not need to resort to b-agonists, and diabetics do not require different doses of insulin to shift potassium
the commonly encountered “insulin-resistant” patients actually have preserved insulin-induced potassium disposal, one wonders why their high insulin levels are not causing hypokalemia
insulin independently regulates glucose and potassium uptake into cells and this independence explains why in noninsulin-dependent diabetic insulin resistance leads to impaired insulin uptake into cells but has no effect on the cell's potassium disposal
insulin suppresses glycogenolysis, gluconeogenesis, lipolysis and fatty acid release, and protein catabolism and is the principal hormone that stimulates glucose uptake into mainly skeletal muscle and to a certain extent adipocytes
Plasma [K+] is a major determinant of the resting potential of all cells
Hyperkalemia and hypokalemia are silent yet fatal disturbances because of their arrhythmogenic potentials
Basal insulin maintains fasting plasma [K+] within the normal range
When insulin levels are suppressed, plasma [K+] rises and pronounced hyperkalemia develops after a potassium load
Potassium is a well proven insulin secretagogue
Insulin is a key defender against exogenous potassium load by using intracellular buffering to minimize hyperkalemia before renal excretion
Hyperkalemia is often encountered in patients with diabetes
The insulin-deficient state in type 1 diabetes predisposes to hyperkalemia because of an impaired ability of potassium to enter cells. During hyperglycemic hypertonic states in type 1 and type 2 diabetics, potassium is carried out of cells by convective flux as the most abundant intracellular cation
good review of the potassium, glucose, insulin relationship mostly in diabetes. In diabetes, hyperkalemia is present due to the hyperglycemia and the associated exchange. Inuslin independantly regulates potassium and glucose intake into the cell. INterestingly, in IR found in diabetes, the hyperkalemia is the norm, which should cause hypokalemia--the authors were perplexed by this finding.
The avermectins are known to possess pronounced antitumor activity
Over the past few years, there have been steadily increasing reports that ivermectin may have varying uses as an anti-cancer agent, as it has been shown to exhibit both anti-cancer and anti-cancer stem cell properties
In human ovarian cancer and NF2 tumor cell lines, high-dose ivermectin inactivates protein kinase PAK1 and blocks PAK1-dependent growth
PAK1 is essential for the growth of more than 70% of all human cancers, including breast, prostate, pancreatic, colon, gastric, lung, cervical and thyroid cancers, as well as hepatoma, glioma, melanoma, multiple myeloma and for neurofibromatosis tumors
Ivermectin suppresses breast cancer by activating cytostatic autophagy, disrupting cellular signaling in the process, probably by reducing PAK1 expression
Cancer stem cells are a key factor in cancer cells developing resistance to chemotherapies and these results indicate that a combination of chemotherapy agents plus ivermectin could potentially target and kill cancer stem cells, a paramount goal in overcoming cancer
Triple-negative breast cancers, which lack estrogen, progesterone and HER2 receptors, account for 10–20% of breast cancers and are associated with poor prognosis
Ivermectin addition led to transcriptional modulation of genes associated with epithelial–mesenchymal transition and maintenance of a cancer stem cell phenotype in triple-negative breast cancers cells, resulting in impairment of clonogenic self-renewal in vitro and inhibition of tumor growth and metastasis in vivo
ivermectin synergizes with the chemotherapy agents cytarabine and daunorubicin to induce cell death in leukemia cells
Ivermectin-induced cytostatic autophagy also leads to suppression of tumor growth in breast cancer xenografts, causing researchers to believe there is scope for using ivermectin to inhibit breast cancer cell proliferation and that the drug is a potential treatment for breast cancer
Ivermectin inhibits proliferation and increases apoptosis of various human cancers
Activation of WNT-TCF signaling is implicated in multiple diseases, including cancers of the lungs and intestine,
A new screening system has found that ivermectin inhibits the expression of WNT-TCF targets
It represses the levels of C-terminal β-catenin phosphoforms and of cyclin D1 in an okadaic acid-sensitive manner, indicating its action involves protein phosphatases
In vivo, ivermectin selectively inhibits TCF-dependent, but not TCF-independent, xenograft growth without side effects
ivermectin has an exemplary safety record, it could swiftly become a useful tool as a WNT-TCF pathway response blocker to treat WNT-TCF-dependent diseases, encompassing multiple cancers.117
Accumulating evidence suggests that niclosamide targets multiple signaling pathways such as nuclear factor-kappaB (NF-kB), Wnt/β-catenin, and Notch, most of which are closely involved with cancer stem cell proliferation
The transcription factor NF-κB has been demonstrated to promote cancer growth, angiogenesis, escape from apoptosis, and tumorigenesis
NF-κB is sequestered in the cytosol of resting cells through binding the inhibitory subunit IκBα
Niclosamide blocked TNFα-induced IκBα phosphorylation, translocation of p65, and the expression of NF-κB-regulated genes
Niclosamide also inhibited the DNA binding of NF-κB to the promoter of its target genes
niclosamide has two independent effects: NF-kB activation and ROS elevation
The Wnt signaling pathway plays fundamental roles in directing tissue patterning in embryonic development, in maintaining tissue homeostasis in differentiated tissue, and in tumorigenesis
niclosamide is a potent inhibitor of the Wnt/β-catenin pathway
The Notch signaling pathway plays important roles in a variety of cellular processes such as proliferation, differentiation, apoptosis, cell fate decisions, and maintenance of stem cells
niclosamide potently suppresses the luciferase activity of a CBF-1-dependent reporter gene in both a dose-dependent and a time-dependent manners in K562 leukemia cells
niclosamide treatment abrogated the epidermal growth factor (EGF)-stimulated dimerization and nuclear translocation and transcriptional activity of Stat3, and induced cell growth inhibition and apoptosis in several types of cancer cells (e.g. Du145, Hela, A549) that exhibit relatively higher levels of Stat3 constitutive activation
niclosamide can rapidly increase autophagosome formation
niclosamide induced autophagy and inhibited mammalian target of rapamycin complex 1 (mTORC1)
Niclosamide has low toxicity in mammals (oral median lethal dose in rats >5000 mg/kg
Niclosamide is active against cancer cells such as AML and colorectal cancer cells, not only as a monotherapy but also as part of combination therapy, in which it has been found to be synergistic with frontline chemotherapeutic agents (e.g., oxaliplatin, cytarabine, etoposide, and daunorubicin)
Because niclosamide targets multiple signaling pathways (e.g., NF-κB, Wnt/β-catenin, and Notch), most of which are closely involved with cancer stem cells, it holds promise in eradicating cancer stem cells
Review article: common anti-parasitic medication, niclosamide, provides anti-proliferative effect in cancer stem cells (CSC), via inhibition of NF-kappaBeta, Wnt/B-catenin, Notch, ROS, mTORC1, and STAT2 pathways.
Ovarian cancer is the most lethal gynecologic malignancy and the fifth-most cause of overall cancer death of women in developed countries
An increasingly accepted cancer stem cell hypothesis regards tumors as caricatures of normal organs, possessing a hierarchy of cell types, at various stages of aberrant differentiation, descended from precursor tumor-initiating cells (TIC) cells that are highly resistant to conventional cytotoxics
Significant changes of gene expression in 2,928 genes were identified after niclosamide treatment for different time periods
uncoupling of mitochondrial oxidative phosphorylation is believed to be its anti-helminthic mechanism of action
we hypothesized that niclosamides antagonistic effects on OTICs could, in part, be due to its disruption of metabolism
Our results showed that genes participating in protein complexes of oxidative phosphorylation were downregulated
niclosamide treatment resulted in a more than 20% increase in reactive oxygen species (ROS) in cultured OTICs
niclosamide, which has proved to be safe and effective for the past 2 decades against numerous parasites, inhibited OTIC growth both in vitro and in vivo
niclosamide represses metabolic enzymes responsible for bioenergetics, biosynthesis, and redox regulation specifically in OTICs, presumably leading to mitochondrial intrinsic apoptosis pathways, loss of tumor stemness, and growth inhibition
Niclosamide is believed to inhibit mitochondrial oxidative phosphorylation
Niclosamide was reported to inactivate NF-κB, causing mitochondrial damage and the generation of ROS, leading to apoptosis of leukemic stem cells
niclosamide were identified in a screen for mTOR-signaling inhibitors
mTOR was reported to maintain stemness properties of HSCs by inhibiting mitochondrial biogenesis and ROS levels (39), implying that mTOR inhibitors (such as niclosamide) may interfere with mitochondria and various metabolic pathways in TICs via disruption of antioxidant responses
We observed Wnt hyperactivity in OTICs, in agreement with previous hypotheses of Wnt inhibitor effectiveness as an ovarian cancer therapy
niclosamide has now been independently identified in screens for Wnt inhibitors
downregulation of the Wnt/β-catenin target oncogenes survivin and c-Myc
ovarian carcinogenesis, the cell-to-cell signaling pathway Notch (8), were also suppressed by niclosamide (data not shown). These results agree with another recent niclosamide study in leukemia (49), and it has been widely hypothesized that disruption of Notch signaling may represent a highly effective therapy for ovarian and other solid tumors, via its essentiality to maintaining TIC stemness
Niclosamide, common anti-parasitic medication, inhibits cellular metabolism and increases ROS; both of which provide powerful anti-proliferative, anti-cancer treatment mechanism in TICs. Powerful target therapy for cancer stem cells. Also shown to inhibit Wnt stimulated oncogenes survivin and c-Myc, disrupts Notch signaling, inactivates NF-kappaBeta, and inhibits mTOR-signaling. This has been found in in vitro and in vivo studies.
use of PET in clinical research, clinical trials, and drug discovery
use of PET/CT in assessing response to therapy
In some cases, such as Hodgkins lymphoma, quantitative PET/CT imaging may not actually be needed, as success can be defined by the complete absence of tracer uptake in the PET image following a course of standardized therapy
The utilization of PET/CT to assess response to therapy is increasing in the US related, in part, to the creation and subsequent favorable results of the National Oncologic PET Registry (NOPR)
Changes in size as a result of therapy may take many months to develop and any opportunity to make early decisions about therapy success or failure is often unduly delayed or lost altogether
measures of changes in metabolic activity via FDG PET/CT can provide an alternate approach to assess response to therapy -- often very early in the course of treatment
Current recommendations are that tumor SUVs should be reported
The true tracer uptake in a patient is composed of two components: the first being the amount of tracer uptake (e.g. FDG) associated with the disease status (the signal of interest), which can be modified by the biophysiological status of the patient. One of the more important patient parameters is the blood glucose level, which has been shown to inversely-linearly affect SUVs
A prospective study by Crippa et al.30 in eight patients showed that as blood glucose levels were increased from 92.4 ±10.2 to 158 ± 13.8 mg/100 ml by glucose loading, the average SUV of 20 liver metastases decreased from 9.4 ± 5.7 to 4.3 ± 8.3
chemotherapy can result in impaired renal function, significantly reducing the clearance of plasma FDG through the kidney and thus increasing tumor SUV relative to an initial PET scan
The second component of the true tracer uptake is biological variability
The biological variability has been estimated in several test-retest studies7,32–35 at approximately 10% for scans repeated within a few days
the finding of a stem cell–like gene expression signature is of great interest because it reflects the malignant nature of a tumor with poor survival outcome
poor prognosis of HCC in patients with stemness-associated gene expression traits is assumed to reflect the abundance of liver CSCs with highly tumorigenic and/or metastatic features
CSCs have a highly invasive and metastatic capacity
The discovery and exploration of liver CSCs has expanded our knowledge of the mechanisms by which liver cancers obtain tumorigenic, metastatic, and chemotherapy- and radiation-resistant capacities
dysregulated miRNA could be involved in tumor cell proliferation and growth as well as cell cycle progression
under-expression of miR-497, 376c and 1271 in Lebanese breast cancer tissues
The upregulated miR-183 in our samples was predicted to be responsible for the decrease in expression of the BTG1 mRNA whose protein is involved in cell cycle arrest and apoptosis in breast cancer cells18.
nother molecule related to cell proliferation that was over-expressed in our data and suggested as a target of downregulated miR-376c is AURKA
the over-expression of miR-183 and miR-21 in Lebanese breast cancer tissues is consistent with downregulation of two important tumor suppressor predicted targets: AKAP12 whose protein regulates cellular adhesion dynamics by controlling cytoskeletal architecture, cell migration, and mitogenic signaling20; and LATS2 whose protein causes cell cycle arrest
dysregulation in cancer particularly in breast cancer highlights their importance in tumor development
mRNA-miRNA integration analysis of early breast cancer revealed a potential role of miRNA in increasing cellular proliferation and progression, and decreasing invasion and migration
most of the miRNA dysregulated in Lebanese breast cancer patients are similar to those dysregulated in American patients, differences in miRNA expression exist and could be attributed either to the patients’ age at diagnosis or to ethnic variation in miRNA epigenetic regulation and sequence variation of pre-miRNA
the number one cancer killer of women worldwide
microRNA (miRNA) are small non-coding 18–25 nucleotide RNA molecules currently being studied as potential diagnostic, prognostic and therapeutic biomarkers for cancer and other diseases
Extensive research on these post-transcriptional modulators has proven that they are deregulated in breast cancerous tissues and even in biological fluids from breast cancer patients
five candidate miRNAs (miR-10b, miR-148b, miR-221, miR-21, and miR-155)
Epigenetics plays a role, via disregulated miRNA, in increased cell growth, progression, and invasion in Lebanese women with breast cancer. It is not just genetics that play a role, but epigenetics.
These eight distinct cancer types included: DCIS, breast (ER(+) and ER(-)), ovarian, prostate, lung, and pancreatic carcinomas, as well as melanoma and glioblastoma. Doxycycline was also effective in halting the propagation of primary cultures of CSCs from breast cancer patients, with advanced metastatic disease (isolated from ascites fluid and/or pleural effusions)
Doxycycline behaves as a strong radio-sensitizer, successfully overcoming radio-resistance in breast CSCs
cancer cells can indeed escape the effects of Doxycycline, by reverting to a purely glycolytic phenotype. Fortunately, the metabolic inflexibility conferred by this escape mechanism allows Doxycycline-resistant (DoxyR) CSCs to be more effectively targeted with many other metabolic inhibitors, including Vitamin C, which functionally blocks aerobic glycolysis
Vitamin C inhibits GAPDH (a glycolytic enzyme) and depletes the cellular pool of glutathione, resulting in high ROS production and oxidative stress
DoxyR CSCs are between 4- to 10-fold more susceptible to the effects of Vitamin C
Doxycycline and Vitamin C may represent a new synthetic lethal drug combination for eradicating CSCs, by ultimately targeting both mitochondrial and glycolytic metabolism
inhibiting their propagation in the range of 100 to 250 µM
metabolic flexibility in cancer cells allows them to escape therapeutic eradication, leading to chemo- and radio-resistance
used doxycycline to pharmacologically induce metabolic inflexibility in CSCs, by chronically inhibiting mitochondrial biogenesis
This treatment resulted in a purely glycolytic population of surviving cancer cells
DoxyR cells are mainly glycolytic
MCF7 cells survive and develop Doxycycline-resistance, by adopting a purely glycolytic phenotype
Cancer stem cells (CSCs) are thought to be the “root cause” of tumor recurrence, distant metastasis and therapy-resistance
the conserved evolutionary similarities between aerobic bacteria and mitochondria, certain classes of antibiotics inhibit mitochondrial protein translation, as an off-target side-effect
Vitamin C was more potent than 2-DG; it inhibited DoxyR CSC propagation by > 90% at 250 µM and 100% at 500 µM
IC-50
DoxyR CSCs are between 4- to 10-fold more sensitive to Vitamin C than control MCF7 CSCs
Berberine, which is a naturally occurring antibiotic that also behaves as an OXPHOS inhibitor
treatment with Berberine effectively inhibited the propagation of the DoxyR CSCs by > 50% at 1 µM and > 80% at 10 µM.
Doxycycline, a clinically approved antibiotic, induces metabolic stress in cancer cells. This allows the remaining cancer cells to be synchronized towards a purely glycolytic phenotype, driving a form of metabolic inflexibility
Doxycycline-driven aerobic glycolysis
new synthetic lethal strategy for eradicating CSCs, by employing i) Doxycycline (to target mitochondria) and ii) Vitamin C (to target glycolysis)
Doxycycline inhibits mitochondrial biogenesis and OXPHOS,
hibits glycolytic metabolism by targeting and inhibiting the enzyme GAPDH
CSCs act as the main promoter of tumor recurrence and patient relapse
a metabolic shift from oxidative to glycolytic metabolism represents an escape mechanism for breast cancer cells chronically-treated with a mitochondrial stressor like Doxycycline, as mitochondrial dys-function leads to a stronger dependence on glucose
Vitamin C has been demonstrated to selectively kill cancer cells in vitro and to inhibit tumor growth in experimental mouse models
many of these actions have been attributed to the ability of Vitamin C to act as a glycolysis inhibitor, by targeting GAPDH and depleting the NAD pool
here we show that DoxyR CSCs are more vulnerable to the inhibitory effects of Vitamin C, at 4- to 10-fold lower concentrations, between 100 to 250 μM
concurrent use of Vitamin C, with standard chemotherapy, reduces tumor recurrence and patient mortality
after oral administration, Vitamin C plasma levels reach concentrations of ~70-220 μM
intravenous administration results in 30- to 70- fold higher plasma concentrations of Vitamin C
pro-oxidant activity results from Vitamin C’s action on metal ions, which generates free radicals and hydrogen peroxide, and is associated with cell toxicity
it has been shown that high-dose Vitamin C is more cytotoxic to cancer cells than to normal cells
This selectivity appears to be due to the higher catalase content observed in normal cells (~10-100 fold greater), as compared to tumor cells. Hence, Vitamin C may be regarded as a safe agent that selectively targets cancer cells
the concurrent use of Doxycycline and Vitamin C, in the context of this infectious disease, appeared to be highly synergistic in patients
Goc et al., 2016, showed that Doxycycline is synergistic in vitro with certain phytochemicals and micronutrients, including Vitamin C, in the in vitro killing of the vegetative spirochete form of Borrelia spp., the causative agent underlying Lyme disease
Doxycycline, an FDA-approved antibiotic, behaves as an inhibitor of mitochondrial protein translation
CSCs successfully escape from the anti-mitochondrial effects of Doxycycline, by assuming a purely glycolytic phenotype. Therefore, DoxyR CSCs are then more susceptible to other metabolic perturbations, because of their metabolic inflexibility
it appears more likely that the effectiveness relies on the properties of doxycycline as an inhibitor of tumor cell proliferation and less on its effect as a MMP inhibitor
Our results suggest that doxycycline may be useful not only for the treatment of osteolytic but also for the treatment of osteoblastic bone metastasis
A prominent feature of bone metastasis of breast cancer is the uncoupling of bone remodeling
doxycycline can improve this to some extent by increasing bone formation
the current study clearly demonstrates the benefit of doxycycline when administered from the time of the development of the tumor
In conclusion, doxycycline greatly reduced tumor burden and could also compensate for the increased bone resorption frequently associated with bone metastasis from breast cancer
The five most common adult malignancies (colorectal, breast, prostate, melanoma and lung cancer)
n breast cancer, the optimal regimen(s) for cytotoxicchemotherapy in recurrent/metastatic disease are still notdefined, despite over 30 years of ‘research’ and a plethora of RCTs since the original Cooper regimen was published in1969
The five most ‘chemo-sensitive’ cancers,namely testis, Hodgkin’s disease and non-Hodgkin’s lym- phoma, cervix and ovary
only 13 out of the 22 malignancies evaluated showed any improvement in 5-year survival, and theimprovement was greater than 10% in only three of those13 malignancies
the contribution of curative and adjuvant cytotoxic chemotherapy to 5-year survival in adults is 2.3% in Australia and 2.1% in the USA
a benefit of less than 2.5% is likely to be applicable in other developed countries
Overview
The Contribution o
the benefit of cytotoxic chemotherapy may have been overestimated for cancers of oesophagus, stomach,rectum and brain.
this reflects the presentation of results as a ‘reduction in risk’ rather than asan absolute survival benefit[89,90]and by exaggerating theresponse rates by including ‘stable disease’
recent
studies have documented impaired cognitive function inwomen receiving adjuvant treatment for breast cancer
the 5-year survival rate due solely to cytotoxicchemotherapy was 1.6%
the value of palliative chemotherapy has beenquestioned
Incredibly low impact of cytotoxic chemotherapy despite its wide spread utilization. This article referenced cost yet did not evaluate the cost of cytotoxic side effect. The question to answer: is Cytotoxic chemotherapy a valid treatment, at all, for the majority of cancers.
Treg act to prevent spontaneous autoimmunity and to limit collateral damage to healthy tissues during adaptive immunity. However, these cells also have the potential to sabotage protective antimicrobial responses
Great T cell activiation review: Il-2 stimulates NK cells primarily release from TH1 cells and T cytotoxic lymphocytes are under the control of IL-12 released primarily from dendritic cells. Inflammatory cytokines in the presence of Treg to stimulate CD4+CD25- T cell activation.
High density level of PGR in the TE was an independent prognostic factor for CF.
Our large-sized study demonstrates a wide distribution of PGR in stromal and epithelial cells of both benign and malignant prostate tissue
there seems to be a general agreement of PGR presence in the stromal cells of PCa
In line with our findings, several have also reported a high PGR expression in TE of PCa [9,10,23,25]. In contrast, others have demonstrated a total lack of PGR expression in TE
the actions of progesterone are tissue specific
In our work univariate analysis demonstrated a high PGR expression in TS to be associated with clinical failure in PCa patients. So far we have not yet demonstrated the mechanism underlying this association
Several non-genomic proliferative actions of progesterone have been proposed in tumor cells of other organs, including breast [35–37], astrocytoma [38] and osteosarcoma [39] cell lines. However, such results are contradicted by suggestions of anti-proliferative actions of progesterone in endometrial cancer
Yu et al. found PGR to be negatively regulating stromal cell proliferation in vitro
high PGR density level in TE was associated with CF in patients with Gleason score ≥ 7
Bonkhoff et al. have suggested progressive emergence of PGR during PCa progression and metastasis
Latil and co-workers found a decreased PGR expression in clinically localized tumors and increased PGR expression in hormone-refractory tumors, when compared with normal prostate tissue
Our findings provide further support to these findings, indicating that PGR plays a role in the pathogenesis of PCa
Ki67 and PGR in TE were correlated with CF (S3 Text), indicating an association between PGR and proliferative activity
The mechanism behind the PGR up-regulation in PCa has not yet been elucidated
The PGR is, like the glucocorticoid receptor, similar to androgen receptor with 88% sequence homology in the ligand-binding domain
progesterone induced expression of androgen receptor-regulated genes could be a potential mechanism contributing to the development of castrate resistant PCa
A possibility of different roles by the two PGR isoforms in normal prostate tissue and PCa, as is suggested for the estrogen receptors [13], must also be taken into account
STudy finds that increased Progesterone receptor expression on epithelial and stromal cells is associated with increased clinical failure of therapy. Several proposed mechanisms: 88% homologous with androgen receptor suggesting cross-stimulation and via progesterone induced increased androgen receptor gene stimulation i.e. epigenetics.
What is diverticulitis and what signs and symptoms occur when you have diverticulitis? What Foods You Should Eat If You Have Diverticulitis and what you should not eat when you discovered you have diverticulitis. Described many causes in this article if you are looking for causes of diverticulitis.
Diverticulitis is a very serious medical condition. It causes inflamed pouches in the lining of your intestine. It is a very serious medical condition that causes infection or inflammation of small pouches in the lining of your intestine that is known as diverticula that develop along the walls of the intestines.
To date nearly half of known human tumors show a dysregulation of the WNT signaling pathway
It should be also noted that the WNT pathway is not exclusively employed during development or overactivated in cancer. In adults many healthy tissues rely on it for renewal and homeostasis maintenance, most notably the intestine, haematopoietic system, hair, bones and skin. Therefore one might expect adverse reactions in all these organ systems, which has indeed been observed for many WNT-targeting compounds upon attempts to push them into the clinics
The intestine seems to be the most vulnerable in this regard
Ivermectin inhibits proliferation of human colon cancer and lung cancer cells both in vitro and in vivo
The anti-proliferative action, affecting both the bulk tumor cells and CSCs, was linked in this study to inhibition of WNT signaling
the anti-WNT IC50 of ivermectin is 5–10 times (~1–2 µM vs. 10 µM) lower than that of its toxic effect against chloride channels
oral bioavailability of the drug, as for other antiparasitic drugs discussed in this section, is very low
Toxicity studies in vivo have also demonstrated a wide therapeutic index for ivermectin
Its anti-proliferative activity has been demonstrated in a wide array of cancer cell lines representative of WNT-dependent cancers: non-small lung carcinoma [96], multiple myeloma [97], hepatoma [98], adrenocortical carcinoma [99], ovarian cancer [100] and glioblastoma
Niclosamide inhibits the canonical WNT pathway
In addition to inhibiting the canonical WNT pathway, niclosamide may mediate its anticancer activities through several other signaling pathways such as NOTCH [107], MTOR [108], NF-κB [97] and STAT3 [96]
review article highlights older medications that have anti-Wnt pathway effects in cancer. Roughly, 50% of cancer involve upregulated Wnt pathway activity. Other drugs of note: metformin
Pharma Sust 250mg injections treat hypogonadism disorders in men, after the illness, impotence due to lack of hormones, menstrual symptoms in men such as reducing sexual pleasure and physiological activities.
Side effects:
The drug can cause erectile dysfunction, signs of excessive sexual stimulation, reduced sperm count, decreased volume of ejaculation, water & salt retention.
In pre-puberty boys: develop sex early, increase the frequency of erectile dysfunction, enlargement of the penis, and early growth of bone heads.
This situation of "not going to the market has run out of money" is also a complaint that most doctors in Nam Khoa listen to from patients. If from the beginning, sexual and sexual attractions were simply a kind of human instinct, through which the race would be passed on to the next generation, today with development. The prosperity of human society, sex is also a joy, a happiness and sometimes also a powerful demonstration of love for couples. Because of that, the strategy of "beating fast and fast" which is an advantage in the past has become a difficult obstacle for the heart of the foolish love.
Let's find out how to overcome "the market has not run out of money" by the article Premature ejaculation cause and treatment - Men Health!
It is produced predominately by antigen-simulated CD4+ T cells, while it can also be produced by CD8+ cells, natural killer (NK) cells, and activated dendritic cells (DC)
IL-2 is an important factor for the maintenance of CD4+ regulatory T cells
plays a critical role in the differentiation of CD4+ T cells into a variety of subsets
It can promote CD8+ T-cell and NK cell cytotoxicity activity, and modulate T-cell differentiation programs in response to antigen, promoting naive CD4+ T-cell differentiation into T helper-1 (Th1) and T helper-2 (Th2) cells while inhibiting T helper-17 (Th17) differentiation
Of note, Tregs, which act to dampen the immune response, constitutively express high levels of α chain
IL-2Rα is unique to IL-2 and is expressed by a number of immune cells including T regulatory cells (Treg), activated CD4+ and CD8+T cells, B cells, mature DCs, endothelial cells
some investigators evaluated the efficacy of regimens containing low-dose IL-2
IL-2 can promote the activation and cell growth of T and NK cells
Unfortunately, not all of patients would benefit from targeted therapy and nearly all patients who initially respond to targeted inhibitors inevitably develop acquired resistance to the treatment
IL-2 also stimulates T-regulatory cells that constitutively express CTLA-4 and can suppress immune reactions. Hence, IL-2 might enhance antitumor reactivity in the presence of CTLA-4 blockade
both HD and low-dose IL-2 therapy preferentially induce the expansion of CD4+CD25+Foxp3+ Treg and the Treg level remains elevated after each cycle of HD IL-2 therapy
Due to rapid elimination and metabolism via the kidney, IL-2 has a short serum half-life of several minutes
HD IL-2-induced severe toxicities including vascular leak syndrome (VLS), pulmonary edema, hypotension, and heart toxicities