Literatura magnezijum askorbat



Literatura:

Preuzeto sa: http://mineralsinc.com/WriteUp/MagnesiumAscorbate_w.htm

Magnesium Ascorbate being a natural neutral salt has significantly higher gastrointestinal tolerance and hence is more safe and effective in Magnesium therapy. Knowing the widespread requirements the body has for magnesium, magnesium is being looked to more and more as adjunct to several medical regimens. The more magnesium is investigated, the more far reaching are its apparent benefits.
This organically bound form of magnesium is recognized as the natural and more bio-available delivery system for the vital anti-stress mineral.
Structure
Requirements
The Food and Nutrition Board of the Institute of Medicine of the United States National Academy of Sciences has recommended the following Adequate Intake (AI) and Recommended Dietary Allowance (RDA) values for magnesium

Age (Year)
Infants
AI (milligrams/day)
0 – 6 months 30
7 – 12 months 75
Children RDA (milligrams/day)
1 – 3 years 80
4 – 8 years 130
Boys
9 – 13 years 240
4 – 18 years 410
Girls
9 – 13 years 240
4 – 18 years 360
Men
19 – 30 years 400
31 – 70 + years 420
Women

19 – 30 years 310
31 – 70 + years 320
Pregnancy
14 – 18 years 400
19 – 30 years 350
31 – 50 years 380
Lactation
14 – 18 years 360
19 – 30 years 310
31 – 50 years 320
Nutritional significance of Magnesium in cardiovascular treatment
Magnesium’s importance to the health of a large number of biological systems is receiving ever-increasing support. Magnesium has an essential part in hundreds of biochemical roles, and depending upon the biological systems in the body that is involved, it may perform its function via different mechanisms.
Magnesium’s place in the area of platelet aggregation appears to be crucial to the control or palliation of a number of important health problems. The hyperactivity of platelets that results in their aggregation and the subsequent release of potent vasoconstrictors may be at the base of problems, such as migraine headaches and vascular diseases associated with diabetes, strokes and various cardiovascular diseases.
Recent research indicates magnesium deficiency may be a factor in the development of atherosclerosis and coronary artery disease- the major cause of heart attacks. Several clinical studies indicate that maintenance of adequate red blood cell magnesium levels may be key in fighting these health problems.
Role of Ascorbic acid in cardiovascular therapy
In the early 1990s, several large population studies showed a reduction in cardiovascular disorder in those who consumed vitamin C (Ascorbic acid). A study, which evaluated 11,348 participants over a ten-year period of time, showed that high vitamin C intake extended average life span and reduced mortality due to cardiovascular disease by 42%. This was again confirmed by a study involving 11,178 participants who took vitamin C and vitamin E supplements.
Several studies suggest different mechanism of action by which ascorbic acid protects against cardiovascular disease.
a.        Atherosclerosis is a natural protective mechanism against the arterial deteriorating effects by a vitamin C deficiency. Thus vitamin C deficiency leads to atherosclerosis which in turn causes cardiovascular disease.
b.       Vitamin C enables the arterial system to expand and contract with youthful elasticity.
c.        Co-administration of nitrate drugs with vitamin C to coronary artery disease patients prolongs the vasodilating effects of the nitrate drugs and the energy producing capacity of the cells is maintained. Otherwise, nitrate drugs alone cause progressive weakening of the heart muscle’s ability to produce energy and also the vascular system stops responding to the dilating effects of the drug. Vitamin C thus potentially helps to prevent the development of nitrate tolerance.
d.       An especially damaging effect of nitrate drugs is that they cause a decrease in the intracellular product of cGMP. This energy substrate is required to maintain cellular energy levels. Vitamin C has been shown to protect against nitrate-induced depletion of cGMP. The use of vitamin C during pre-treatment with nitrate drugs increased the availability of nitric oxide, an important precursor to cGMP.
Thus published research findings suggest that ascorbic acid may reduce mortality in coronary artery disease patients, increase life span and possibly eliminate the effects of nitrate tolerance in those taking nitrate drugs.
Magnesium Ascorbate, which provides both Magnesium and Ascorbic acid (Vitamin C) appears to be unique and promising product, in that both the components have the potential to reduce the risk of cardiovascular disease.
Other nutritionally important roles of Magnesium and
L- Ascorbic acid
Magnesium is the fourth most abundant mineral in the body. It is essential to build the bones and teeth and numerous metabolic functions.
Magnesium is also an ingredient in some antacids and laxatives and is used to prevent premature birth and treat certain types of convulsions and rapid heartbeats (tachycardia). Magnesium works with calcium and phosphorous to build strong bones and teeth.
It also plays important roles in:
·         Normal metabolism
·         Proper nerve and muscle function
·         Stimulating calcium function
·         Preventing dental cavities
·         Promoting immunity and
·         Boosting the actions of potassium and some of B-vitamins
·         It may also aid in the treatment of asthma, cardiac arrhythmia, high blood pressure, fibromyalgia and diabetes.
In addition to its use for the treatment of hypomagnesemia, magnesium is used for the treatment of certain cardiac arrhythmia’s, in particular torsasde de points and eclampsia. Magnesium may also have value for the prevention of osteoporosis and for the management of migraine headaches in some. There is preliminary evidence that magnesium may help some with premenstrual syndrome, type 2 diabetes mellitus and hypertension.
Ascorbic acid is involved in modulating iron absorption, transport and storage. It aids in the intestinal absorption of iron by reducing ferric iron to ferrous iron and may stimulate ferritin synthesis to promote iron storage in cells. It is involved in the biosynthesis of corticosteroids, aldosterone, the conversion of cholesterol to bile acids and functions as a reducing agent for mixed-function oxidases.
For all of this, ascorbic acid is best known for its antioxidant properties and its possible role in the prevention of certain chronic degenerative disorders, such as coronary heart disease and cancer. In fact, ascorbic acid may be the most important water-soluble antioxidant in the body.
Ascorbic acid has antioxidant activity. It may also have antiatherogenic, anticarcinogenic, antihypertensive, antiviral, antihistaminic, immunomodulatory, opthalmoprotective and airway-protective actions. It may aid in the detoxification of some heavy metals, such as lead and other toxic chemicals.
Ascorbic acid or, more specifically, ascorbate is an excellent reducing agent, and it acts as cofactor in various biochemical reactions to reduce the transition metals, iron and copper. It can be oxidized by most reactive oxygen and nitrogen species thought to play roles in tissue injury associated with various diseases. By virtue of this scavenging activity, ascorbate inhibits lipid peroxidation, oxidative DNA damage and oxidative protein damage.
The antioxidant activity of ascorbate is well established and that activity may be helpful in the prevention of some cancers and cardiovascular diseases. It may also be helpful in protecting against some of the lipid oxidation caused by smoking.
Ascorbic acid may also be helpful as immune stimulator and modulator in some circumstances. It may help prevent cataract.



Efekti tretmana Kancera visokim koncentracijama Askorbata

Izvor: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3798917/

The Effects of High Concentrations of Vitamin C on Cancer Cells

This article has been cited by other articles in PMC.

Abstract

The effect of high doses of vitamin C for the treatment of cancer has been controversial. Our previous studies, and studies by others, have reported that vitamin C at concentrations of 0.25–1.0 mM induced a dose- and time-dependent inhibition of proliferation in acute myeloid leukemia (AML) cell lines and in leukemic cells from peripheral blood specimens obtained from patients with AML. Treatment of cells with high doses of vitamin C resulted in an immediate increase in intracellular total glutathione content and glutathione-S transferase activity that was accompanied by the uptake of cysteine. These results suggest a new role for high concentrations of vitamin C in modulation of intracellular sulfur containing compounds, such as glutathione and cysteine. This review, discussing biochemical pharmacologic studies, including pharmacogenomic and pharmacoproteomic studies, presents the different pharmacological effects of vitamin C currently under investigation.
Keywords: high concentrations of vitamin C, pharmacogenomics, pharmacoproteomics

1. Introduction

There is increasing evidence that vitamin C (ascorbate) is selectively toxic to some types of tumor cells, functioning as a pro-oxidant [1,2,3]. Vitamin C at concentrations of 10 nM–1 mM induced apoptosis in neuroblastoma and melanoma cells [4] and was shown to be an important modulator for the growth of mouse myeloma cells in an in vitro colony assay [5]. Studies have established that the growth of leukemic progenitor cells from patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) can be significantly modulated by vitamin C [6,7]. Intravenous (i.v.) administration of sodium 5,6-benzylidene-l-ascorbate (SBA) to inoperable cancer patients induced a significant reduction in tumor volume without any adverse side effects [8]. Furthermore, recent clinical studies have reported that manipulation of vitamin C levels in vivo can result in clinical benefit for patients with AML and solid tumors [9].
Recent pharmacokinetic studies reported that 10 g of ascorbate given by i.v. produced plasma concentrations of nearly 6 mM, which were 25-fold higher than concentrations resulting from the same oral dose [10,11,12]. Depending on the dose, as much as a 70-fold difference in plasma concentration was found between oral and i.v. administration [13]. Complementary and alternative medicine practitioners worldwide currently use ascorbate i.v. in patients, in part because there are no apparent harmful effects [14,15,16]. Physiological concentration of vitamin C is under 0.1 mM, while plasma vitamin C concentrations that cause toxicity to cancer cells in vitro (1 mM–10 mM, depending on cell lines) can be achieved clinically by intravenous administration, which means a high dose of vitamin C.
In this review, in vitro and in vivo studies are summarized, showing that ascorbate killed cancer cells (Table 1). In addition, the mechanisms and physiologic relevance under investigation are also described. The results suggest that doses of vitamin C induce oxidative stress in cancer cells. Our previous results indicated that treatment of malignant lymphocytic cell lines with vitamin C (0.25–1 mM) for 24 h led to a marked dose-dependent decrease of cell proliferation [17]. The responsive cell lines were human myeloid leukemia cell line HL-60, retinoic acid (RA)-sensitive acute promyelocytic leukemia (APL) cell line NB4 and RA-resistant APL cell line NB4-R1. Different types of leukemia cells, such as K562 (chronic myelogenous leukemia cell line) [17] and KG1 (myeloblast cell line) [18], were also responsive to vitamin C. A similar result was obtained with cells containing over 90% of blasts from patients with AML. In these cell lines, induction of apoptosis by vitamin C demonstrated a dose-dependent effect. In addition, vitamin C weakly induced apoptosis in ovarian cell lines, including SK-OV-3, OVCAR-3 and 2774 [17]. For many of the cancer cell lines, ascorbate concentrations caused a 50% decrease in cell survival. The half maximal concentration (IC50) values were less than 5 mM, and all tested normal cells were insensitive to 20 mM ascorbate [13].
Table 1
Effects of vitamin C treatment on cell survival [13,17,18].

2. Molecular Mechanisms Induced by Vitamin C

In our previous study, vitamin C at concentrations of 0.25–2.0 mM significantly induced apoptosis in AML cell lines. Vitamin C induced oxidation of glutathione (GSH) to its oxidized form (GSSG). As a result, H2O2 accumulated in a concentration-dependent manner, in parallel with the induction of apoptosis. The direct role of H2O2 in the induction of apoptosis in AML cells was demonstrated by the finding that catalase could completely abrogate vitamin C-induced apoptosis [17].
A 30-min incubation of NB4 cells with 0–10 mM vitamin C resulted in its uptake in a concentration-dependent manner [17,19]. In accordance with its proposed pro-oxidant activity, vitamin C treatment reduced the GSH/GSSG ratio, which correlated with increased intracellular H2O2 in the NB4 cell line. Levine et al. suggested that the effect was due only to extracellular and not intracellular ascorbate, and that ascorbate-mediated cell death was probably due to protein-dependent extracellular H2O2 generation, via ascorbate radical formation from ascorbate as the electron donor [13].
Although H2O2 induced by ascorbate was first generated extracellularly, it is possible that it could diffuse across the plasma membrane into the intracellular space. Although studies in yeast and bacteria have shown that diffusion of H2O2 across membranes is limited, some reports have suggested that selected aquaporin homologues from plants and mammals can channel H2O2 across these membranes [20,21,22,23,24,25]. The susceptibility of cancer cells to ascorbate treatment might therefore be related to the permeability of hydrogen peroxide.
In our studies, vitamin C dramatically increased intracellular GSH oxidation and reactive oxygen species (ROS) levels within 3 h in a concentration-dependent manner. However, this GSH oxidation and the ROS accumulation did not last for a long period of time. After 3 h, the increase in GSH has been observed and hypothesized to be a defense mechanism [18]. No additional ROS accumulation resulted from the change in GSH. However, based upon our studies, the dramatic changes of intracellular oxidation state within 3 h seem to be enough to induce intracellular signaling. The studies showed that treatment with 1 mM vitamin C for only 30–60 min, followed by removal when replacing media, could induce apoptosis in both HL-60 and NB4 cells. This observation is consistent with initiation of apoptosis, resulting from generation of H2O2 after treatment with ascorbate. However, treatment with As2O3 resulted in less ROS accumulation than with vitamin C, and it was not in a concentration-dependent manner. However, ROS accumulation increased up to 24 h, with a long-lasting effect. Treatment with vitamin C combined with As2O3 increased ROS accumulation, as well as sustained the effect for up to 24 h. These results are consistent with cellular data showing that apoptosis induced by As2O3 is evident even at three days [26,27].

3. Proteomics Studies

Proteomics provides important qualitative information on post-translational modifications to proteins and quantitative data on protein expression in response to a particular stimulus. This information is particularly important when it provides data on early cellular events, such as the stimulus and signaling cascades triggered independently of protein neosynthesis. In accordance with its proposed pro-oxidant activity, vitamin C-mediated reduction in the GSH/GSSG ratio correlated with an intracellular H2O2 increase in the NB4 cell line [17,19]. This type of change in regional oxidation state could cause changes in the cellular milieu that could result in changes in protein structure. This is especially true of the oxidation state of cysteine sulfur, which is important for determining the tertiary structure of proteins. The immediate effects of cell stimuli are associated with protein post-translational modifications, such as phosphorylation, glutathionylation and cysteine oxidation. To study these early modifications, NB4 human leukemia cells were treated with 0.5 mM vitamin C and then analyzed by two-dimensional analysis. Approximately 240 different spots that were focused in a pH range of 4–7 were detected per sample.
After exposing cells to vitamin C, we observed one new spot, three intensified spots and five attenuated spots [19]. Each of these spots were excised, digested with trypsin and analyzed by matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF). Peptide mass fingerprint analysis and non-redundant sequence database matching allowed the unambiguous identification of all of the analyzed species [19]. An important protein identified from the proteomic analysis was a thiol/disulfide exchange catalyst, protein disulfide isomerase (PDI), which was a marker of the effect of vitamin C on NB4 cells [19]. PDI belongs to the superfamily of thiol/disulfide exchange catalysts, which act as protein-thiol-oxidoreductase enzymes, sharing sequence homology with thioredoxin [28]. PDI is composed of four domains, which have similarities with thioredoxin folds (i.e., a-b-b′-a′) [29]. In our study [19], the intensity of the spot corresponding to the PDI b subunit was decreased in vitamin C-treated cells as compared with control cells. These results demonstrated that thiol/disulfide exchange proteins are regulated in NB4 cells after vitamin C exposure. This is consistent with our study showing that intracellular GSH/GSSG exchange occurs shortly after vitamin C exposure [17].
When we measured cysteine uptake in leukemia cell lines exposed to vitamin C by using 35S-labeled-L-Cys in the media, the time-dependent rate of cysteine uptake in the cell culture increased significantly. The rate of uptake, determined under conditions without vitamin C, was very low. The glutathione synthesis inhibitor, buthionine sulfoximine, potently inhibited this increase, suggesting that incorporation of cysteine that corresponded to the amount of increased glutathione was mediated by glutathione synthesis. Overall, our results indicated that vitamin C-induced glutathione synthesis was accompanied by intracellular cysteine uptake.
Glutathione-S transferases (GSTs) are enzymes that catalyze the conjugation of electrophilic substitution to GSH, which protects cells by removing reactive oxygen species and regenerating S-thiolated proteins [30]. Intracellular total GSH levels in AML cells incubated with vitamin C peaked around 3 h, then declined, while the increase in incorporated [35S]-L-Cys peaked at 3 h and remained high. These results showed that [35S]-L-Cys transported into cells through cysteine uptake was incorporated and transferred intracellularly, strongly suggesting that the sulfhydryl transfer system is affected by vitamin C [30].
We therefore hypothesize that the biochemical pathway leading to thiol/disulfide redox regulation could be activated by vitamin C. Interestingly, of the proteins whose expressions changed by vitamin C treatment, immunoglobulin heavy chain binding protein (BiP, identical to Hsp70 chaperone) [19], like PDI, is also a multi-domain chaperone. BiP associated with the α-subunit of prolyl 4-hydroxylase (P4-H) by a disulfide bond [31]. P4-H consists of two distinct polypeptides, the catalytically more important α-subunit and the β-subunit, which is identical to the multifunctional enzyme, PDI [31]. Thus, BiP associated with the α-subunit of P4-H, which is another partner of PDI. The interaction of PDI with its substrates was due to a change in disulfide bonds, indicating that intrachain disulfide bonds between domains and substrates had been reduced [19]. Taken together, these results suggested that vitamin C oxidizes intracellular-reduced glutathione and affects disulfide bond formation in proteins [30].
Tropomyosin was also identified as a marker of the oxidative effect of vitamin C in NB4 cells. The spot corresponding to tropomyosin was positioned at an isoelectric point (pI) of approximately 5.0 and was attenuated in vitamin C-treated cells. In addition, a new spot having almost the same molecular weight was detected, which was positioned at a pI of 4.9 [19]. This new spot was also identified as tropomyosin, suggesting that post-translational chemical modification had affected its pI value. This result is consistent with previous data showing that the extracellular signal-regulated kinase (ERK)-mediated phosphorylation of tropomyosin-1 promoted cytoskeleton remodeling in response to oxidative stress [32]. The acidic shift of the spot with pI 5.0 to the phosphorylated tropomyosin spot by treatment with vitamin C was found to be abrogated by co-treatment with PD98059 [19], demonstrating that phosphorylation of tropomyosin was responsible for the observed acidic shift.
The significance of this observation may be related to differences in the regulation of the actomyosin contractile system in non-muscle cells as compared with that present in muscle cells. In addition, proteins that specifically reacted with sera from chronic myeloid leukemia patients included structural proteins, such as β-tubulin and tropomyosin isoforms [33]. Although the function of these proteins in myeloid leukemia needs further investigation, tropomyosin may have value as a leukemia-associated antigen and as a molecular target in antigen-based immunotherapy. In this regard, it is important to note that vitamin C causes a tropomyosin isoform to be modified during the immediate early response.

4. In Vivo Studies

Levine et al. reported that reaction products obtained from ascorbate in vitro are also found in vivo [34]. They showed that after i.v. injection, ascorbate baseline concentrations of 50–100 µM in blood and extracellular fluids peaked to >8 mM. After intraperitoneal injection, peaks approached 3 mM in both fluids. They hypothesized that in vivo, ascorbate was a prodrug for selective delivery of ascorbate radical and H2O2 to the extracellular space. Moreover, a regimen of daily pharmacologic ascorbate treatment significantly decreased growth rates of ovarian (p < 0.005), pancreatic (p < 0.05) and glioblastoma (p < 0.001) tumors established in mice [35]. In addition to inducing oxidative stress, high concentrations of ascorbic acid inhibited cell migration and the gap filling capacity of endothelial progenitor cells (EPCs) [36], and it has been reported that ascorbic acid inhibited corneal neovascularization in a rat model [37].
High concentrations of ascorbic acid also inhibited tumor growth in BALB/C mice implanted with sarcoma 180 cancer cells [38]. The survival rate increased by 20% in the group that received high doses of ascorbic acid, compared to controls. Gene expression studies from biopsy and wound healing analysis in vivo and in vitro suggested that the carcinostatic effect induced by high doses of ascorbic acid were related to inhibition of angiogenesis [39]. In addition, intraperitoneal administration of high doses of ascorbic acid quantitatively upregulated Raf kinase inhibitory protein (RKIP) and annexin A5 expression in a group of BALB/C mice implanted with S-180 sarcoma cancer cells. The increase in RKIP protein levels suggested that these proteins are involved in the ascorbic acid-mediated suppression of tumor formation [39]. Moreover, high doses of ascorbic acid (~1 mM) enhanced the apoptosis of cancer cells during co-treatment with paclitaxel, and the combinational treatment of paclitaxel and ascorbic acid ameliorated the side effects caused by paclitaxel in BALB/c mice implanted with or without sarcoma 180 cancer cells [40].

5. Clinical Studies

Cases of apparent responses of malignancies to intravenous vitamin C therapy have been reported, although they were reported without sufficient detail [15,41,42,43,44,45,46]. A recent study showed that oral administration of the maximum tolerated dose of vitamin C (18 g/day) produced peak plasma concentrations of only 0.22 mM, whereas intravenous administration of the same dose produced plasma concentrations approximately 25-fold higher. Larger doses (50–100 g) given intravenously resulted in plasma concentrations of approximately 14 mM [41].
Some case reports stated that high dose i.v. vitamin C has been used by Complementary and Alternate Medicine (CAM) practitioners [47]. Phase I evaluation of intravenous vitamin C in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer data did not reveal increased toxicity with the addition of ascorbic acid [48]. No side effects were reported for most patients, while 59 were reported to have lethargy or fatigue out of 11,233 patients that received intravenous vitamin C in 2006 and 8876 in 2008 [47]. Overall, it was reported that high dose intravenous vitamin C did not appear to cause serious side effects in patients.
Another clinical study reported the depletion of L-ascorbic acid alternating with its supplementation in the treatment of patients with acute myeloid leukemia or myelodysplastic syndromes [49]. During the supplementation phase, patients received daily i.v. administration of vitamin C. A pre-therapy in vitro assay was performed for vitamin C sensitivity of leukemic cells from individual patients. Of the nine patients with the in vitro assay indicating their leukemic cells were sensitive to vitamin C, seven exhibited a clinical response, compared with none of the six patients who were insensitive to vitamin C.

6. Conclusions

Previous studies have shown that vitamin C is involved in the mechanism of action of the intracellular microenvironment (oxidation) state changes that improve therapeutic potential, including apoptosis and necrosis. Although it is difficult to postulate precise vitamin C-specific mechanisms at this time, identification of genes or proteins that are specifically regulated by vitamin C in certain cellular phenotypes could improve the efficacy of therapies.

Conflicts of Interest

The author declares no conflicts of interest.

References…



                         Anti- Kancer lek
Kombinacija Magnezijum Askorbata i Vitamina K3 (100:1)
Izvor: US Patent US 8,507,555 B2
(NON-TOXIC ANTI-CANCER DRUG COMBINING ASCORBATE, MAGNESIUM AND A NAPHTHOQUINONE)
Inventors: Thomas M. Miller, El Cajon, CA (US);
James M. Jamison, Stow, OH (US)

IZVODI IZ PATENTA:

…by the Examples of types of cancers that are successfully treated
present compositions and methods are presented in the
list below and in Table 1, which is not intended to be limiting.
Thus the present invention is directed to the treatment of
pancreatic carcinomas, renal cell carcinomas, small cell lung
carcinoma, non-small cell lung carcinoma, prostatic carci
noma, bladder carcinoma, colorectal carcinomas, breast, ova
rian, endometrial and cervical cancers, gastric adenocarci
noma, primary hepatocellular carcinoma, genitourinary
adenocarcinoma, thyroid adenoma and adenocarcinoma,
melanoma, retinoblastoma, neuroblastoma, mycosis fun
goides, pancreatic carcinoma, prostatic carcinoma, bladder
carcinoma, myeloma, diffuse histiocytic and other lympho
mas, Wilms’ tumor, Hodgkin’s disease, adrenal tumors
(adrenocortical or adrenomedullary), osteogenic sarcoma,
soft tissue sarcoma, EWing’s sarcoma, rhabdomyosarcoma
and acute or chronic leukemias, islet cell cancer, cervical,
testicular, adrenocortical, or adrenomedullary cancers, cho
riocarcinoma, embryonal rhabdomyosarcoma, Kaposi’s sar
coma, etc.
TABLE 1
List of Cancers/Tumors
acoustic neuroma
adenocarcinoma
angiosarcoma
astrocytoma
basal cell carcinoma
bile duct carcinoma
bladder carcinoma
breast cancer
bronchogenic carcinoma
cervical cancer
chondrosarcoma
choriocarcinoma
colorectal carcinomas
craniopharyngioma
cystadenocarcinoma
embryonal carcinoma
endotheliosarcoma
ependymoma
esophageal carcinoma
EWing’s tumor
?brosarcoma
gastric carcinoma
Glioma/glioblastoma
Head and neck cancers
Hemangioblastoma
Hepatocellular carcinoma
Hepatoma
Kaposi’s sarcoma
leiomyosarcoma
liposarcoma
lung carcinoma
lymphangiosarcoma
lymphangioendotheliosarcoma
Lymphoma
Leukemia
medullary carcinoma
medulloblastoma
Melanoma
meningioma
mesothelioma
Multiple myeloma
Myxosarcoma
Nasopharyngeal carcinoma
Neuroblastoma
oligodendroglioma
osteogenic sarcoma
ovarian cancer
pancreatic cancer
papillary adenocarcinomas
pinealoma
prostate cancer
renal cell carcinoma
retinoblastoma
rhabdomyosarcoma
sebaceous gland carcinoma
seminoma
small cell lung carcinoma
squamous cell carcinoma
sweat gland carcinoma
synovioma
testicular tumor
Thyroid cancer
Wilms’ tumor

  ------

Example III
Formulations
A. Capsules
The preferred embodiment of the method utilizes oral
delivery. Capsules of a combination of MgVC2/V K3 are pre
pared with the agents in a predetermined ratio. For example,
0.5 g of Mg ascorbate (L-Ascorbic acid magnesium salt) is
combined with 0.005 g of water soluble vitamin K3 (mena
dione sodium bisulfite). Both vitamins are mixed in the pow-
dered form and placed in capsules without supplementary
ingredients at the predetermined ratio such as is 100:1…




         Medicinska studija Lečenja raka prostate sa Apatone®                          (Magnezijum askorbat + Vitamin K3 u odnosu 100:1)

Izvor: http://www.medsci.org/v05p0062.htm


Int J Med Sci 2008; 5(2):62-67. doi:10.7150/ijms.5.62
Research Paper
A 12 Week, Open Label, Phase I/IIa Study Using
Apatone® for the Treatment of Prostate Cancer
Patients Who Have Failed Standard Therapy
Basir Tareen1, Jack L. Summers1, James M. Jamison1, Deborah R. Neal1,
Karen McGuire1, Lowell Gerson1, Ananias Diokno 2
1. Summa Health System, Department of Urology, Akron, Ohio, USA
2. William Beaumont Hospital, Department of Urology, Royal Oak, Michigan, USA
This is an open access article distributed under the terms of the Creative
Commons Attribution (CC BY-NC) License. See http://ivyspring.com/terms
for full terms and conditions.
How to cite this article:
Tareen B, Summers JL, Jamison JM, Neal DR, McGuire K, Gerson L, Diokno
A. A 12 Week, Open Label, Phase I/IIa Study Using Apatonefor the
Treatment of Prostate Cancer Patients Who Have Failed Standard Therapy.
Int J Med Sci 2008; 5(2):62-67. doi:10.7150/ijms.5.62. Available from

Abstract
Purpose: To evaluate the safety and efficacy of oral Apatone® (Vitamin C and
Vitamin K3) administration in the treatment of prostate cancer in patients who
failed standard therapy.
Materials and Methods: Seventeen patients with 2 successive rises in PSA
after failure of standard local therapy were treated with (5,000 mg of VC and
50 mg of VK3 each day) for a period of 12 weeks. Prostate Specific Antigen
(PSA) levels, PSA velocity (PSAV) and PSA doubling times (PSADT) were
calculated before and during treatment at 6 week intervals. Following the
initial 12 week trial, 15 of 17 patients opted to continue treatment for an
additional period ranging from 6 to 24 months. PSA values were followed for
these patients.
Results: At the conclusion of the 12 week treatment period, PSAV decreased
and PSADT increased in 13 of 17 patients (p ≤ 0.05). There were no
dose-limiting adverse effects. Of the 15 patients who continued on Apatone
after 12 weeks, only 1 death occurred after 14 months of treatment.
Conclusion: Apatone showed promise in delaying biochemical progression in
this group of end stage prostate cancer patients.
Keywords: Prostate, Prostate neoplasms, ascorbic acid, menadione, Vitamin
K3, Apatone, Cancer

Introduction
The PSA era has led to a stage migration in the clinical course of prostate
cancer. While this success has dramatically lowered the death rate from
prostate cancer, it remains the most common cancer in men with 234,460 new
cases and 27,350 deaths in the US in 2007.1 While hormonal therapy is
typically initiated when the disease has advanced beyond local involvement
and delays the time to PSA recurrence, it has not improved overall survival2 of
patients with metastatic disease and has significant side effects.3 Newer
chemotherapeutic regimens for metastatic prostate cancer show promise,4 but
there are few therapy options for androgen independent prostate cancer
(AIPC) patients. Therefore, there is a substantial need for new therapeutic
options.
Because of their relatively low systemic toxicity, vitamin C (VC) and vitamin
K3 (VK3), have been evaluated for their abilities to prevent and treat cancer.5
VC exhibited selective toxicity against a variety of malignant cell lines,
prevented the induction of experimental tumors, acted as a chemosensitizer,
and acted in vivo as a radiosensitizer. However, variable clinical results were
obtained with VC because of the difficulty of attaining clinically active doses.6
VK3 exhibited selective antitumor activity alone and in conjunction with many
chemotherapeutic agents in human cancer cell lines. However, while
intravenous VK3 acted as a chemosensitizing and radiosensitizing agent in
patients, 30% of the patients exhibited hematologic toxicity (at higher
doses).7 When VC and VK3 were combined in a ratio of 100:1 (Apatone) and
administered to human tumor cell lines, including androgen independent
prostate cancer cells (DU145), they exhibited a synergistic inhibition of cell
growth and induced cell death by apoptosis at concentrations that were 10 to
50 times lower than for the individual vitamins.8, 9, 10 In addition, oral
Apatone significantly (P << 0.01) increased the mean survival time of nude
mice inoculated i.p. with DU145 cells and significantly reduced the growth rate
of solid tumors in nude mice (P < 0.05) without inducing any significant bone
marrow toxicity, changes in organ weight or pathologic changes of these
organs.9
The purpose of this study was to evaluate the safety and efficacy of oral
Apatone administered throughout the day in prostate cancer in patients who
failed standard therapy.

Materials and Methods
Patient selection
Prostate cancer patients who had failed standard therapy were enrolled at
William Beaumont, Royal Oak, MI and Summa Health Systems, Akron, OH.
Standard therapy was defined to include radical prostatectomy, radiotherapy
and hormonal ablation. We did not include docetaxol chemotherapy in our
inclusion criteria for failure of standard therapy. A patient was required to
have a biopsy with proven prostate cancer and 2 successive rises in PSA to be
included in the study. The patient could not be currently undergoing
chemotherapy, radiotherapy, or androgen deprivation.
All patients exhibited acceptable renal function with blood urea nitrogen lower
than 40 mg/dl and creatinine levels lower than 3 mg/dl and lacked clinical
signs of obstructive liver disease as demonstrated by SGOT levels below 75
U/l; SGPT levels below 80 U/l and Alkaline Phosphatase levels below 200 U/l.
TPatients using anticoagulants, chemotherapeutic agents, vitamin K, or
vitamin C were excluded from the study. This protocol was reviewed and
approved by the Institutional Review Board, and all patients provided their
voluntary, written informed consent.
Evaluations
Each subject was interviewed by the study coordinator and examined by an
urologist. Pretreatment evaluation included: a complete history and physical
examination with a digital rectal examination for prostate configuration, size
and symmetry; a medication audit; AUA Pain Scale and Symptom Score
analysis; a complete blood count with differential, comprehensive chemistry
panel including liver and renal panel, coagulation studies and a PSA test. We
did not include standard bone scans or other radiographic studies as part of
our study protocol. Patients were always seen by the study coordinator and
the same examining urologist.
Treatment
All patients were treated with Vitamin C: K3 (5,000 mg. of VC and 50 mg. of
VK3 each day, Apatone) for a total of 12 weeks. Apatone in capsular form (500
mg VC as ascorbate and 5mg VK3 as bisulfite) at a dose of 2 capsules on
arising, then 1 capsule every two hours for six doses followed by two capsules
at bedtime for a total of ten capsules per day. Following the 12 week study,
two of the three “non-responders” in the study who had large body mass index
values were given double the dose of Apatone by doubling the number of
capsules in the previous regimen.
Analysis of PSA changes and Statistics
PSA velocity (PSAV) and doubling time (PSADT) were calculated using the
Prostate Cancer Research Institute Algorithms.12 Successful outcome was
considered a PSADT increase and a PSAV decrease. The binomial expansion
was used to calculate the exact probability of the number of successful
outcomes among the enrolled patients. A probability of p <0.05 was taken as
indicative of an Apatone effect. Matched t-tests were employed to test for
significant difference in PSA velocity and doubling times before and after
treatment.13 Linear spline fit analysis was used to measure and compare PSA
values before, during and after therapy.12

Results
Of thirty-three patients approached for participation, fourteen were not
eligible; one withdrew; and one did not have two documented PSA values prior
to enrollment. The characteristics of the remaining seventeen patients are
detailed in Table 1. The median patient age was 71.5 (range 56 – 85 years),
AUA performance status 6.5 (range 1 – 14) and median number of prior
chemotherapy regimens was two.
Table 1
Patient Characteristics
N = 17
Age: median (range) 74.5 (56 – 85)
AUA Symptom Score: median (range) 6 (1 – 14)
Race:
Caucasian 15
African American 1
Prior therapies (1 or more treatments):
Hormonal 10
Radiation 8
Surgery 17
Chemotherapy:
None 0
One 0
Two 0
N = 17
Three or more 1
Pre-treatment PSAV ranged from 1.05 to 696 ng/ml/year (median 21.6
ng/ml/yr), while in-trial PSAV ranged from -12 to 256 ng/ml/year (median
6.39 ng/ml/yr). Conversely, pre-treatment PSADT values ranged from 2.0 to
54.4 months (median 3.12 months), while in-trial PSADT values ranged from
-39 to 57.1 months (median 7.88 months).
Linear spline fit analysis was performed using PSA levels before treatment,
during treatment and following treatment (Figure 1). Representative curves
are shown for a patient with a pre-treatment PSA > 30 ng/ml (Fig. 1a), a
patient with 30 ng/ml > PSA > 10 ng/ml (Fig. 1b) and for a patient with a PSA
< 10 ng/ml (Fig. 1c). In all 3 cases, the rate of PSA increase is significantly
decreased during Apatone treatment, but increases at a rate similar to that
seen before treatment once treatment ended (Fig. 1a and 1b). Thirteen of the
17 patients had a successful outcome; a decrease in PSAV and a lengthening
of PSDT (Table 2). The probability of 13/17 successful outcomes is 0.008
suggesting the 76 % response we observed, was unlikely due to chance. The 3
“non-responders” each volunteered to have their dose of Apatone doubled
following the trial. There were no adverse effects and two of these three
patients subsequently had a decrease in PSA velocity and increase in PSA
doubling time. No patient had a significant decrease in absolute PSA.
Figure 1
Natural Log transformations of PSA measurements for patients with PSA
greater than 30ng/ml (a); between 10-30ng/ml (b) and less than 10ng/ml
(c). Before and after treatment with Apatone plotted against time in weeks
fitted with a linear spline with knots at -60 weeks, the start of Apatone
therapy and end of therapy. () indicate where patients went off Apatone or
started alternative therapy.
(Click on the image to enlarge.)
(Click on the image to enlarge.)
(Click on the image to enlarge.)
Table 2
PSA Velocity and Doubling Time in Months
Patient PSA Velocity PSA Doubling Time
Pre-trial In-trial Change Pre-trial In-trial Change
1 1.74 - 12.0 Decreased 54.4 - 5.86 Increased
2 26.1 - 3.01 Decreased 2.51 - 21.2 Increased
3 257 158 Decreased 3.00 6.30 Increased
Patient PSA Velocity PSA Doubling Time
Pre-trial In-trial Change Pre-trial In-trial Change
4 14.6 9.14 Decreased 27.6 57.05 Increased
5 4.38 3.65 Decreased 2.76 9.17 Increased
6 19.3 - 8.11 Decreased 12.1 - 39.5 Increased
7 9.95 2.74 Decreased 2.72 13.7 Increased
8 1.05 0.00 Decreased 10.6 > 60 Increased
9 696 256 Decreased 2.03 9.24 Increased
10 46.5 12.6 Decreased 3.23 20.4 Increased
11 0 0 Unchanged 0 0.00 Unchanged
12 2.09 0.00 Decreased 5.23 > 60 Increased
13 352 163 Decreased 2.79 8.90 Increased
14 21.1 81.7 Increased 7.24 4.30 Decreased
15 54.2 112 Increased 2.91 2.88 Decreased
16 22.0 30.9 Increased 6.54 6.58 Increased
Following the 12 week trial, 15 of 17 patients opted to continue Apatone
therapy. Any decision to remain on Apatone therapy was left entirely to the
patient. Anecdotally, most patients reported feeling “better” and more
“energetic.” This coupled with stabilization of rising PSA along with no
significant side effects led the men to continue therapy. Four continued
therapy for 6 months and 11 continued for at least 1 year with one patient
continuing for more than 2 years. Therapy was not discontinued in any patient
due to vitamin toxicity or for other safety reasons. The PSA values of these
patients were checked at various intervals while on treatment and remained
stable. Patients terminating Apatone therapy experienced sharp increases in
PSA levels as seen in the linear spline fit analysis (Figure 1). Of the 11
patients on therapy for greater than 1 year, only one (initial PSA 256, PSADT=
3 months, and PSAV 157ng/ml/yr) passed away after 14 months.
No noteworthy changes were observed in the patient's complete blood counts,
biochemistry panels or coagulation studies. No dose limiting toxicity or
adverse events were experienced. Mild intermittent gastro-esophageal reflux
symptoms was observed in 16 of 17 patients, but was eliminated when the
Apatone was taken with meals or with antacids. The average AUA symptom
score prior to beginning therapy was 7.9 (Table 3). This fell to 7.2 upon
completion of the 12 week trial (P = .07). The average pain score based on
the standard index was 3.2 initially, 2.3 at 6 weeks and returned to 3.2 at
twelve weeks (Table 4).
Table 3
AUA Symptom Scores
AUA Score In
Points
Number of
Patients
Initial
Visit
Six Week
Visit
Twelve Week
Visit
Mild (0-7) 7 4.14 ア
0.40
4.27 ア
0.51
3.43 ア 0.53
Moderate (8-19) 9 10.9 ア
0.71
12.0 ア
0.21
10.0 ア 0.80
Severe (20-35) 0 -- -- --
= Data expressed as the mean ア standard error of the mean
Table 4

Pain Scores
AUA Pain Score In
Points
Initial Visit Six Week
Visit
Twelve Week
Visit
3.19 ア
0.79
2.31 ア 0.66 3.19 ア 0.70
= Data expressed as the mean ア standard error of the mean

Discussion
In a previously published, prospective, randomized trial, patients with
pathologically proven prostate cancer in advanced stages (M1), osseous
metastasis and resistance to hormone therapy were given two, 7 day courses
of oral Apatone (VC at 5 g/m2/day and VK3 at 50 mg/m2/day), VC alone, VK3
alone, or a placebo.14 The 7day courses of treatment occurred during the first
and fourth week of the study with two weeks of follow up after each treatment
period. For the vitamin combination, homocysteine (a marker of tumor cell
death induced by Apatone) assays showed an immediate and statistically
significant drop (p<<0.01) in tumor cell numbers, while PSA serum levels rose
in the two initial weeks and then fell to levels that were significantly different
(p << 0.01) from the control group. For VC and VK3 alone, a non-significant
difference was observed between the serum levels of homocysteine and PSA
compared to the control group which suggested that the decreased PSA levels
were due to tumor cell death.14 In this study, Apatone was administered daily
in a single oral dose which was 2.5 to 3 times higher than the dose employed
during the initial 12 weeks of our study. This dose resulted in a significant
decrease in patient PSA levels which was ascribed to Apatone- induced tumor
cell death by autoschizis. Conversely, the lower Apatone doses employed in
the current study, led to increased PSADT without decreasing patient PSA
levels.
In the previous study, Apatone was given in a single daily dose.14 However,
Apatone was designed as an adjunctive therapy for existing treatment
regimens with Apatone being administered intravenously in a bolus
immediately prior to chemotherapy or radiotherapy and then in daily oral
maintenance doses between therapies to prevent tumor growth following
washout of the chemotherapeutic agent. In addition, pharmacokinetic studies
indicated serum vitamin C levels returned to steady-state values within 5 to 6
hours of oral administration.15 For these reasons, Apatone was given every 5
to 6 hours in this study. During the 12 week course of the study, PSADT was
the primary endpoint. Using this criterion, thirteen of 17 patients had
significant increases in PSA doubling time. Following the initial 12 week trial,
two of the three “non-responders” in the study who had large body mass index
values were given increased Apatone doses adjusted to compensate for their
elevated BMI values. Both patients subsequently became “responders”. In
addition, 15 of 17 patients opted to continue Apatone therapy following the 12
week trial. The PSA values of these patients were checked at various intervals
while on treatment and remained stable. Therapy was not discontinued in any
patient due to vitamin toxicity or for other safety reasons.
PSADT has been useful in predicting treatment outcome before definitive
therapy. For example, PSADT significantly correlated with biochemical
recurrence16, linearly correlated with the interval to clinical relapse after PSA
failure following radiation therapy for prostate cancer17, and was the most
powerful indicator of disease activity in men under observation alone.18 When
pretreatment variables in patients with androgen-independent prostate cancer
were analyzed to determine the effect on PSA response after initiating
maximum androgen blockade, increased PSADT was the only significant
predictor of response.19 These results and others have led D'Amico to conclude
that PSADT is sufficiently robust as a surrogate marker of prostate cancer
survival to serve as a valid endpoint in trials of patients with hormonerefractory
disease.17
More recently, PSADT has been used as an effective in vivo method for
screening nontoxic agents, such as dihydroxyvitamin D3 (calcitriol), that
increase PSADT without concomitantly decreasing PSA and yet become
clinically valuable when used in combination with other anticancer agents.11
Our results demonstrate that oral Apatone significantly increased the PSADT of
almost all the patients without concomitantly decreasing PSA, while
co-administration of Apatone with known chemotherapeutic agents in other
cancers resulted in a synergistic increase in antitumor activity.8,20 These
results suggest that Apatone may find use in the clinic as a co-adjuvant
therapy potentially in addition to docetaxol. Our decision not to include
patients with alkaline phosphatase over 200 U/l may have excluded a number
of men with osteoblastic bone lesions from metastases. This inherent selection
bias does not allow us to examine the potential role of Apatone as salvage
therapy, potentially after failure of docetaxol chemotherapy in hormone
refractory patients.

Conclusions
Apatone is safe and effective with thirteen of the 17 prostate cancer patients
having a statistically significant (P-value < 0.05) increase in PSADT and a
decrease in PSADV after taking Apatone for 12 weeks. The long-term impact
of Apatone on disease progression is unknown and remains to be
demonstrated by further clinical study. Additional studies appear warranted for
the use of Apatone as a co–adjuvant, or for emerging salvage chemotherapy in
the treatment of late stage prostate cancer.

Acknowledgements
This research was supported by grants from The Beaumont Foundation, Royal
Oak, Michigan, IC-MedTech, Inc, San Diego, California and The Summa Health
System Foundation, Akron, Ohio.
Conflict of interest
The authors have declared that no conflict of interest exists.
References…

Author contact
Correspondence to: Basir Tareen, M.D., Department of Urology, New York
University, 550 First Avenue, New York, NY 10016.
tareen@medicine.nodak.edu
Received 2008-1-27
Accepted 2008-3-23
Published 2008-3-24
Copyright ゥ2017 Ivyspring International Publisher. Terms of Use & Privacy