Sei sulla pagina 1di 5

Vitamin C supplementation exhibits an inhibitory effect on proliferation of cancer cells

NDFS 434

Dr. Jeff Tessem

February 20, 2017

Abstract:

Several studies have examined the effect of vitamin C on cancer cell development and the
mechanisms through which the vitamins apparent inhibitory effect on cancer cells occurs. In one
study, researchers found that vitamin C combined with cisplatin enhances the drugs activity
without becoming toxic to cells, which consequentially reduces the required dose of cisplatin to
kill cancer cells. In a similar study, vitamin C was combined with doxorubicin, an anthracycline
drug that inhibits the growth of breast cancer cells. Researchers observed significant changes in
26 cellular proteins and improved anti-cancer results in cells treated with both vitamin C and
doxorubicin. In another study, mice with or without endogenous vitamin C synthesis were
injected with cancer cells, then administered varied levels of vitamin C. All mice without
endogenous vitamin C production developed tumors, while mice at the highest dosage of vitamin
C developed no tumors. In a study examining the effect of vitamin C supplementation on cancer
cells, researchers determined that high-dose vitamin C induces oxidative stress in cancer cells.
Together, these studies suggest that vitamin C does have an anti-proliferative effect on cells, and
may enhance the activity of anti-cancer drugs while decreasing needed dose, thus reducing
toxicity.
Research has demonstrated that vitamin C can slow proliferation of cancer cells by

degrading the extracellular matrix (1) and inducing oxidative stress and cell death. However,

administration of oral high-dose vitamin C has not produced significant results on cancer cells in

clinical trials (2). The question remains whether oral or intravenous administration of vitamin C

has a greater effect on cancer cell death and whether this is a safe and viable treatment for cancer

in humans.

In a study by Cha et al., researchers compared wild-type mice to mice with two human

metabolic features associated with carcinogenesis: production of human Lp(a) and lack of

endogenous vitamin C production. Both groups of mice were injected with breast cancer cells

then fed one of four diets for six weeks, ranging from a low-ascorbate diet to a high-ascorbate

diet. As a result, all wild-type mice developed tumors, while Lp(a)+; Gulo(-/-) mice developed

one-third less tumors on the low-ascorbate diet and no tumors on the high-ascorbate diet. These

results suggest that dietary vitamin C may reduce proliferation of cancer cells, however this

study was not completed on human subjects, and it is unexplained why wild-type mice were

relatively unaffected by vitamin C administration (1).

Further studies have argued that intravenous administration might be a more effective

method of vitamin C supplementation over the oral route, though perhaps not the safest option.

Zhang et al. examined the role of erythrocytes and the pentose phosphate pathway (PPP) in

response to oxidative stress. When vitamin C enters the body in high doses, it is converted to

DHA, then taken into cells through GLUT-1, where it is reduced back to vitamin C at the

expense of cellular antioxidants such as GSH, thus inducing oxidative stress on the cell. In

cancer cells, where there is overexpression of GLUT-1, vitamin C can induce

significant oxidative damage. However, when researchers cultured cancer


cells (HCT116, A375, and SK-MEL-28) with erythrocytes, they appeared to be protected

from such damage, while the erythrocytes responded to the high dose of vitamin C by

undergoing hemolysis. Erythrocytes regularly protect body cells from oxidative stress using the

PPP, and since they possess a high number of GLUT-1 receptors, similarly to cancer cells, they

intake a large amount of vitamin C at high doses and subsequently incur substantial oxidative

damage before surrounding tissues (including cancerous tissue). Thus, intravenous

administration of vitamin C may not be a safe or effective treatment for cancer in humans,

though we cannot immediately come to this conclusion without performing tests outside of

culture medium (2).

Another study was carried out by Bober et al. to experiment with combining doxorubicin,

a chemotherapy drug used to inhibit growth of breast cancer cells, and vitamin C. Researchers

found that vitamin C supplementation by itself did not exhibit a significant anti-proliferative

effect on the MCF-7 breast cancer cell line, but when 200 uM of vitamin C was combined with 1

uM of doxorubicin, the combined anti-proliferative effect was significantly greater than that of

doxorubicin alone (3). Similar research was completed by Leeha et al. examining the effect of

cisplatin-based chemotherapy (CDPP) and vitamin C on HEK and SiHa (cervical cancer) cell

lines. Cells were maintained in culture and treated with doses ranging from 5 to 200 uM CDPP

for 24 hours and 25, 50, and 100 ug/ml vitamin C for 24, 48, and 72 hours. The greatest

cytotoxicity was found at 100 uM CDPP and 100 ug/ml of vitamin C (4). Because many

chemotherapeutic agents cause cumulative toxitity in patients, supplementation with vitamin C

may reduce the dose of such drugs required to treat cancer and subsequently mitigate harmful

side effects. However, a significant limitation to these studies is that they were performed only
on cultured cells, not on live test subjects. Thus, researchers gathered no data regarding the effect

of oral versus intravenous administration of vitamin C.

High-dose vitamin C induces cell lysis in erythrocytes when cultured with cancer cells

(2), however, few studies have been completed on live test subjects to see if cell lysis also occurs

in the body. In this study, mice with induced cancer growth will be administered

chemotherapeutic drugs with varied doses of intravenous vitamin C, and oxidative stress will be

measured in erythrocytes and cancer cells. The hypothesis is that at optimal levels of vitamin C

and chemotherapeutic drugs, as suggested by aforementioned studies (200 uM vitamin C

combined with 1 uM doxorubicin, and 100 ug/ml vitamin C combined with 100 uM CDPP) will

cause greater oxidative stress and cell death in cancer cells compared to erythrocytes, and

therefore produce the greatest anti-proliferative effect on cancer cells.

The first test group (each group will consist of four mice) will only be administered

chemotherapeutic drugs (doxorubicin or CDPP), a second group will only be administered

vitamin C, and the remaining groups will receive either the optimal level of doxorubicin

combined with vitamin C or the optimal level of CDPP combined with vitamin C (as outlined

above). Additional groups will receive the same levels of the chemotherapy drugs with 50 units

more or less vitamin C to examine relative oxidative stress in erythrocytes and cancer cells at

varying levels of intravenous vitamin C administration.

Because research has demonstrated the importance of the pentose phosphate pathway

(PPP) in dealing with oxidative stress (2), the amount of oxidative stress undergone by cells in

each of the test mice will be measured by production of NADPH, a product of the PPP.

Erythrocyte samples and cancer cell samples will be taken from each test subject after one day,

one week, and two weeks of treatment to examine oxidative stress. Higher levels of NADPH
found in erythrocytes compared to cancer cells will indicate that vitamin C administration is

doing more damage to healthy tissue than target cancer cells.

References

1. Cha J, Roomie MW, Kalinovsky T, Niedzwiecki A, Rath M. Lipoprotein(a) and vitamin C


impair development of breast cancer tumors in Lp(a); Gulo-/- mice. Int J Oncol. 2016;49(3)895-
902.

2. Zhang ZZ, Lee EE, Sudderth J, et al. Glutathione depletion, pentose phosphate pathway
activation and hemolysis in erythrocytes protecting cancer cells from vitamin C-induced
oxidative stress. J Biol Chem. 2016;291(44):22861-22867.

3. Bober P, Alexovic M, Talian I, et al. Proteomic analysis of the vitamin C effect on the
doxyrubicin cytotoxicity in the MCF-7 breast cancer cell line. J Cancer Res Clin Oncol.
2017;143(1):35-42.

4. Leekha A, Gurjar BS, Tyagi A, Rizvi MA, Verma AK. Vitamin C in synergism with cisplatin
induces cell death in cervical cancer cells through altered redox cycling and p53 upregulation. J
Cancer Res Clin Oncol. 2016;142(12):2503-2514.

Potrebbero piacerti anche