Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Dec 2.
Published in final edited form as: In Vivo. 2010 May-Jun;24(3):249–255.

Pharmacological Ascorbic Acid Suppresses Syngeneic Tumor Growth and Metastases in Hormone-refractory Prostate Cancer

HARVEY B POLLARD 1, MARK A LEVINE 2, OFER EIDELMAN 1, MORRIS POLLARD 3
PMCID: PMC9718485  NIHMSID: NIHMS1830796  PMID: 20554995

Abstract

Aim:

The aim of this study was to test for the influence of ascorbic acid on tumorigenicity and metastases of implanted PAIII prostate cancer adenocarcinoma cells in syngeneic LW rats.

Materials and Methods:

Hormone-refractory prostate cancer PAIII cells were implanted subcutaneously into immunologically intact, Lobund-Wistar (LW) rats. Intraperitoneal pharmacological doses of ascorbic acid were administered each day for the ensuing 30 days. On the 40th day, animals were sacrificed. Local tumor weights were measured, and metastases were counted.

Results:

At the end of the 40 day experimental period, the primary tumors were found to be significantly reduced in weight (p=0.026). In addition, sub-pleural lung metastases were even more profoundly reduced in number and size (p=0.009). Grossly enlarged ipsilateral lymph node metastases declined from 7 of 15 rats to 1 of 15 rats.

Conclusion:

Pharmacological doses of ascorbic acid suppress tumor growth and metastases in hormone-refractory prostate cancer.

Keywords: Ascorbic acid, metastases, tumor, syngeneic, prostate


Pharmacological doses of ascorbic acid have recently been shown to reduce the growth and weight of human, rat and murine tumor xenografts in athymic, nude mice (1). Specific tumor cell types tested included ovary (Ovcar5), pancreas (Pan02) and glioblastoma (9L). In experiments with the gliobastoma cell line, metastases were entirely absent from the treated cohort, but present in 30% of the controls. The process by which ascorbic acid suppresses tumor xenografts in these model systems appears to depend on the generation of hydrogen peroxide (H2O2) in extracellular fluid (2, 3). Hydrogen peroxide induces necrosis in tumor cells, but not in normal cells (2). A presently unknown metalloprotein mediator is believed to link the formation of pharmacological ascorbate concentrations to H2O2 (3). Similar results have been reported for the TLT mouse hepatoma cell line implanted in the irradiated NMRI/nude mouse (4). However, all of these models have compromised immune systems, which may contribute to the apparent efficacy of ascorbic acid. If the same result could be obtained in an animal model with an intact immune system, the use of ascorbic acid to treat cancer would have important translational consequences (5).

One of the most challenging human tumors to treat is hormone-resistant prostate cancer (HRPC) (68). Patients can develop irreversible, aggressive, therapy-resistant metastatic disease if untreated, or if relapse occurs following failed therapy (9). Therefore, to study a human-like rodent model for HRPC, we have turned our attention to the implanted PAIII hormone-resistant prostate cancer cell in the syngeneic Lobund-Wistar (LW) rat (10). The PAIII cell line is derived from a germfree LW rat with spontaneous advanced stage II HRPC (11), and has been routinely shown to form large primary tumors when injected subcutaneously (12). In 99% of cases, these tumors also develop metastases to local lymph nodes and to lung (9). This pattern of disease has remained constant over time (9, 13, 14). In addition to its faithful metastatic character, the PAIII cell is androgen-receptor-negative (15). As expected, it is also insensitive to testosterone deprivation (9). However, PAIII cells are positive for the estrogen receptors, ERα and ERβ (16), and are suppressed by the selective ER modulator trioxifene (LY133314) (16). Finally, the tumors formed by PAIII cells appear histologically as human-like, poorly differentiated anaplastic lesions (14, 18). Considering all of these properties together, the PAIII/LW rat system appears to be a very compelling small animal model for hormone insensitive prostate cancer in the normal, immunocompetent human population.

Results obtained with tumor xenografts in athymic nude mice demonstrate ascorbate-dependent reduction in both the primary tumor mass and the incidence of metastases. However, all of these models have compromised immune systems, which may contribute either to an enhanced or diminished efficacy of ascorbic acid. We therefore hypothesized that if carcinotoxic actions of ascorbate were independent of a compromised immune system then ascorbic acid should similarly suppress both the PAIII-dependent primary tumor mass and the incidence of PAIII lung metastases in a normal, syngeneic LW rats. We report here that ascorbic acid treatment does lead to both the suppression of the primary tumor mass, and also to the reduction in the incidence of lung metastases. Additionally, we found that ascorbic acid treatment changes the quantitative relationship between primary tumor weight and the number of lung metastases from random to essentially linear. This is the first report of ascorbic acid effects on tumor biology in a syngeneic, immune-competent rodent system.

Materials and Methods

Cells and tumor induction/treatment protocol.

PAIII tumor cells were passaged in LW rats, as previously described (11). To initiate the experiment, 106 cells were implanted subcutaneously into the flanks of 30 adult LW rats, aged 3 months, (12). For the present experiment, 15 of these rats were also treated pharmacologically with ascorbic acid, as described in the protocol below. Two complete experiments were performed. The pioneer experiment, with fewer rats, had very similar results to the second, large-scale experiment, as described in this paper.

Following a ten-day period for primary tumor growth, neutralized ascorbic acid (4 g/kg body weight; ca. 600 mg/day/rat, depending on initial weight; neutralized to pH 7 with NaOH) was injected intraperitoneally (IP) every day for the next 30 days. Control rats received equivalent volumes of sterile distilled water. At the end of the experiment, animals were sacrificed to obtain the primary tumor weight, to record ipsilateral axillary lymph node size, and to analyze lungs for sub-pleural metastases. Animals were cared for in accord with University of Notre Dame guidelines

Measurement of tumor progression.

This measurement was performed exactly as previously described (12). Briefly, rats were weighed, and then sacrificed by deep anesthesia with inhaled isofluorene, and exsanguinated from the exposed heart. This improved the visibility of metastatic foci. The primary tumor was dissected out and weighed. The lungs were removed and inflated through the trachea with Bouin’s fixative. The whole lung was then placed in Bouin’s fixative for 24 hours, and the fixative then replaced with 70% ethanol. Lungs were examined for sub-pleural metastases at ×5 magnification. Total numbers of metastases for each lung were then recorded. Sub-pleural tumors were observed here, as in the past, to expand into the parenchyma of the lung tissue.

Statistics.

Statistics were calculated using Student’s t-test. A p-value of <0.05 was taken as indicative of a significant difference between compared data. Receiver operating charactic (ROC) curves were calculated by standard methods (25).

Results

Implantated PAIII cells develop large primary tumors, and metastatic lesions in lymph nodes and lung. Figure 1a shows the location of a typical subcutaneous primary tumor in the flank of one of the control rats used for this study. Also shown is an ipsilateral, axial lymph node, which is frequently found to be grossly enlarged due to tumor cells. In this experiment, 7 out of 15 control rats typically showed this gross lesion, compared with 1 of 15 rats in those treated with ascorbic acid. Sub-pleural metastases, occurring typically in all PAIII-implanted rats, are also shown. Figure 1b shows subpleural metastases in lungs from a control implanted rat. These tumors initiate as subpleural metastases, and penetrate the parenchyma of the lung. Both large and small metastatic lesions are found.

Figure 1.

Figure 1.

Images of primary PAIII tumor, axillary lymph node, and lung metastases in LW rat. a: Gross image of LW rat implanted with PAIII tumor cells. Primary PAIII tumor (labeled arrow) is shown for a control rat inoculated with 106 tumor cells. An enlarged, ipsilateral axillary lymph node is shown (see labeled arrow). Sub-pleural metastases are shown (see labeled thin arrow). b: Sub-pleural lung metastases. Lung metastases are grossly shown on rat lung surface. Large and small metastases are seen. The data in this paper are based on total sub-pleural metastases observed using ×5 magnification.

Ascorbate treatment reduces primary tumor weight and number of lung metastases. As shown in Figure 2a, treatment of LW rats with implanted PAIII cells resulted in all rats developing primary tumors. The tumor weights averaged 19.9±5.6 (S.D.) g. However, when the parallel cohort of implanted rats were also treated with ascorbic acid, the average tumor weight declined to 15.6±3.8 (S.D.) g. This was a significant difference (t-test: p=0.026). As anticipated, all PAIII-implanted rats developed sub-pleural lung metastases (see Figure 2b). However, when these implanted animals were treated with ascorbate, there was a substantial reduction in numbers of lung metastases. The reduction was approximately 50%, from 11.6±7.6 (S.D.) to 5.4±3.4 (S.D.). The t-test indicates that the difference is very significant (p=0.009). These data are also summarized in Tables I and II. Classical ROC curves are shown for the effect of ascorbate treatment on tumor weight (Figure 3a) and cognate number of lung metastases (Figure 3b). The values for the area under the curve (AUC) for both tumor weight and number of metastases are 0.75 and 0.74, respectively. Perfect separation of data would correspond to an AUC of 1.0. Thus the ROC curves are consistent with the observed degree of data overlap between control and treated animals for both measures.

Figure 2.

Figure 2.

Influence of ascorbate on primary tumor weight and number of lung metastases. a: Influence of ascorbate treatment on primary tumor weight. Fifteen LW rats were treated with 106 PAIII cells, as described. Fifteen other rats were inoculated with the same number of PAIII cells, but also treated with ascorbate (4 g/kg), as described. After 40 days, rats were sacrificed and primary tumors dissected out for measurement of weight. The difference in weight between treated and untreated rats is significant (*p=0.026). b: Influence of ascorbate treatment on number of lung metastases. Lungs from the animals analyzed in Part a were fixed in Bouin’s Fixative, and the total number of metastases counted. The difference in numbers of lung metastases in the two conditions is significant (#p=0.009).

Table I.

Statistical analysis of ascorbate effect on transplanted PAIII prostate cancer.

Body weight Tumor weight Lung MZ, #

Control Avg 366.6 19.9 11.6
SD  26.1  5.6  7.6
ASCORB Avg 344.6 15.6  5.4
SD  23.7  3.8  3.4
t-Test     0.02781    0.02566     0.00987

Table II.

Dataset for PAIII-implanted hormone-resistant prostate cancer cells in LW rats.

Rat# Control
ASCORB
Body weight Tumor weight Lung MZ, # Rat# Body weight Tumor weight Lung MZ, #

 1 399 16.47 16  1 356 19.66 11
 2 379 21.7  9  2 385 15.04  6
 3 376 22.56  4  3 335 21.38 10
 4 358 13.03  4  4 330 14.42  9
 5 357 23.45  7  5 374 13.14  5
 6 382 17.04 20  6 303  9.87  2
 7 342 19.18 27  7 303 11.86  0
 8 328 26.63 19  8 330 17.23  7
 9 405 25.3  5  9 337 10.64  2
10 385  8.49 21 10 345 17.9  7
11 377 29.41 12 11 355 21.7  7
12 385 15.08  7 12 357 17.39  4
13 323 21.46  7 13 350 12.67  4
14 336 18.84  4 14 364 15.65  1
15 270 15.02  Lost 15 350 13.51  Lost
Average  366.6 19.9  11.6  344.6 15.6   5.4
SD   26.1  5.6  7.6   23.7  3.8   3.4

Figure 3.

Figure 3.

Receiver operating condition (ROC) curves for ascorbate effects on primary tumor weight and number of lung metastases. a: ROC curve for ascorbate effects on primary tumor weight. Area under the curve (AUC) is 0.75. A perfect discriminating cut-off point would have a value of 1.0. b: ROC curve for ascorbate effects on the number of lung metastases. Area under the curve (AUC) is 0.74. A perfect discriminating cut-off point would have a value of 1.0.

Ascorbate treatment changes the quantitative relationship between primary tumor weight and number of lung metastases. Figure 4 shows the relationship between weight of the primary tumor for each rat, and the number of lung metastases in each of the rats. In the case of the control rats (untreated with ascorbate), the distribution of tumor weights and cognate metastatic numbers appears to be somewhat random. In an attempt to determine a relationship, we tested whether a linear relationship could be determined. Based on the slope of −0.26 and the R2 value of 0.036, we conclude that a linear relationship is not present (see solid blue diamonds). However, for those rats treated with ascorbate, there is a proportional, and objectively linear relationship, based on the slope of +0.64 and an R2 value of 0.52 (see solid red squares). Furthermore, based on the negative intercept on the x-axis, ascorbate treatment appears to create a mathematically defined lag between primary tumor development and initiation of observable metastases. Such a lag cannot be defined in untreated animals. Thus ascorbate treatment appears to change the fundamental relationship between primary tumor mass and dependent metastases.

Figure 4.

Figure 4.

Ascorbate effects on the relationship between primary tumor weight and number of lung metastases. Data relating tumor weight to metastatic number in the absence of ascorbate treatment are shown in solid diamonds. The R2 value is only ∼0.04, indicating essentially random distribution. Data relating tumor weight to metastatic number in the presence of ascorbate treatment are shown in solid squares. The R2 value is ca. 0.5, indicating a somewhat linear relationship. The vertical intercept is ca. −4.7, indicating a discrete lag between primary tumor weight and metastatic number.

Discussion

These data show that pharmacological ascorbic acid inhibits both the growth and the metastatic potential of hormone-refractory PAIII cells, when these cells are implanted in the syngenic, immune-competent LW rat. This is the first report of pharmacological ascorbic acid effects on tumor biology in a syngeneic, immune-competent rodent system. Thus the observations made by Chen et al. (2008) (1), with tumor xenografts in athymic nude mice, appear to be reproducible in a small animal system with significant, documented clinical similarities to HRPC in man. Pharmacological concentrations of ascorbate only partially suppresses tumor growth and metastases mediated by PAIII cells. These data are thus consistent with the reports for xenotransplants into athymic nude mice (1), or into the NMRI/ heterozygous nude mouse (4). The suppressive effect is qualitatively significant for the incidence of ipsilateral lymph node metastases, and quantitatively significant for sub-pleural lung metastases. The ROC curves (Figure 3) indicate that the effects are quantitatively similar for both the growth of the primary tumor and the occurrence of metastases. Inasmuch as the PAIII system is among the most robust and aggressive of the transplanted prostate cancer systems, we suggest that the pharmacological ascorbate effects documented here would most likely be even more powerful if tested with less aggressive systems. The attractive aspects of ascorbate as a therapeutic agent for humans include that a phase I trial has already been completed to test the safety of pharmacological doses of ascorbate, and there are few documented side-effects (18). Known limitations do exist for the patients with renal insufficiency or G6PD deficiency (18, 19).

The PAIII tumor system is sensitive to other drugs. For example, pretreatment of LW rats with N-(4-hydroxyphenyl)retinamide suppresses tumorigenesis and subpleural lung metastases induced by in vivo-passaged PAIII cells, as used in this paper (20). In addition, for a modified system of cultured PAIII cells injected into the tail vein of LW rats, tumor suppression is observed with the selective estrogen receptor modulators trioxifene (LY133314; (16)) and raloxifene (LY156758; (21)); and the anticoagulants warfarin (22) and hirudin (23). However, in the present study, tumors were allowed to develop for ten days prior to the ascorbic acid treatment. The consequence is that the metastatic program for the PAIII tumor seems to be significantly altered by the ascorbate treatment protocol. For example, as shown in Figure 3, in the presence of continuous dosing with ascorbic acid, the relationship between primary tumor weight and number of metastases becomes essentially linear. By contrast, without ascorbate, the relationship between tumor weight and number of lung metastases is essentially random. Furthermore, in the presence of ascorbate, there is a discrete, mathematically defined lag between the growth of the primary tumor and the eventual occurrence of detectable metastases in the lung. By contrast, no such lag is apparent in untreated control LW rats.

The most likely interpretation of the surprising observation of linearity is the possibility that in the presence of ascorbate there is a critical mass of the primary tumor that must be reached before metastases can occur. The lag mass seemed to be the same for both ascorbate and control tumors, the difference for ascorbate being the reduction in number of metastases and the linearity with tumor mass. Thus the use of pharmacological ascorbate may open a hitherto unknown, limited window of therapeutic opportunity into the metastatic process. Clearly this relationship offers an opportunity for further study of ascorbic acid action on the biology of the PAIII cell system. The recent report that dehydroascorbic acid (oxidized ascorbate) lacks antitumor properties shows that tumor suppression depends on the specific chemical structure of ascorbic acid (24) Dehydroascorbate also opposes the action of some anti-cancer drugs, a property opposite to that reported for pharmacological ascorbic acid in the NMRI/nude mouse system (4). We therefore conclude that pharmacological ascorbate needs to be considered for addition to the limited therapeutic armamentarium that can currently be deployed against HRPC.

Acknowledgements

MP acknowledges technical assistance from Christina Sensabaugh and Cheryl Payne. The authors acknowledge Bette S. Pollard for editorial help.

This work was supported by: (i) Intramural Research Program, National Institute for Diabetes, Digestive and Kidney (NIDDK), National Institutes of Health, Bethesda, MD. 20892, [Z01 DK053212–02] to [MAL]; (ii) National Heart Lung and Blood Institute (NHLBI), National Institutes of Health: [NO1-HV-28187] to [HBP]; (iii) National Cancer Institute, National Institutes of Health [CA 17559] and National Center for Research Resources, National Institutes of Health [RR00294]; The Coleman Foundation of Chicago; The Retirement Research Foundation of Chicago; the Scully Trust; the United States Soyboard; Lobund Institute Endowement; and the University of Notre Dame to [MP]. The authors did not receive any specific support from private or Institutional bodies that might have commercial interests in the project.

References

  • 1.Chen Q, Espey MG, Sun AY, Pooput C, Kirk KL, Krishna MC,Khosh DB, Drisko J and Levine M: Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc Nat Acad Sci USA 105: 11105–11109, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Chen Q, Espey MG, Krishna MC, Mitchell JB, Corpe CP,Beutner GR, Shacter E and Levine M: Pharmacologic ascorbic acid concentrations selectively kill cancer cells: Action as a prodrug to deliver hydrogen peroxide to tissues. Proc Nat Acad Sci USA 102: 13604–13609, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chen Q, Espey MG, Sun AY, Lee JH, Krishna MC, Shacter E,Choyke PL, Pooput C, Kirk KL, Beutner GR and Levine M: Ascorbate in pharmacologic concentrations selectively generates ascorbate radical and hydrogen peroxide in extracellular fluid in vivo. Proc Nat Acad Sci USA 104: 8749–8754, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Verrax J and Calderon PB: Pharmacologic concentrations ofascorbate are achieved by parenteral administration and exhibit antitumoral effects. Free Radical Biology and Medicine 47: 32–40, 2009. [DOI] [PubMed] [Google Scholar]
  • 5.Paddayatti SJ, Riordan HD, Hewitt SM, Katz A, Hoffer LJ andLevine M: Intravenously administered vitamin C as cancer therapy: three cases. Can Med Assoc J 174: 937–942, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Furth J, Kim U and Clifton KH: On evolution of the neoplastic state: progression from dependence to autonomy. National Cancer Institute Monograph No 2: 149–177, 1960. [PubMed] [Google Scholar]
  • 7.Torti F: Hormonal therapy for prostate cancer. N Eng J Med 311: 1313–1314, 1984. [DOI] [PubMed] [Google Scholar]
  • 8.Waxman J: Hormonal aspects of prostatic cancer: a review. J Roy Soc Med 78: 129–135, 1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pollard M and Suckow MA: Hormone-refractory prostate cancerin the Lobund-Wistar rat. Exp Biol Med 230: 520–526, 2005. [DOI] [PubMed] [Google Scholar]
  • 10.Bostwick DG (Rappoteur): Workshop I. Rodent models ofprostate cancer. Prostate 36: 49–55, 1998. [DOI] [PubMed] [Google Scholar]
  • 11.Pollard M and Luckert PH: Transplantable metastasizing prostateadenocarcinoma in rats. J Natl Cancer Inst 54: 643–649, 1975. [PubMed] [Google Scholar]
  • 12.Chang CF and Pollard M: In vitro propagation of prostate adenocarcinoma cells from rats. Inves Urol 14: 331–334, 1977. [PubMed] [Google Scholar]
  • 13.Neubauer BL, Bemis KL, Best KL, Goode RL, Merriman RL,Smith GF, Tanzer LR and Hoover DM: Metastatic spread of the PAIII prostatic adenocarcinoma after implantation in the tail of the rat. Prostate 8: 265–276, 1986. [DOI] [PubMed] [Google Scholar]
  • 14.Koutsilieris M: PA-III rat prostate adenocarcinoma cells. In Vivo 6: 199–204, 1992. [PubMed] [Google Scholar]
  • 15.Bentel JM, Pickering MA, Pollard M, Clements JA and Tilley WD: Androgen receptor expression in primary prostate cancer of Lobund-Wistar rats and in tumor-derived cell lines. Cell Dev Biol Animal 35: 655–662, 1999. [DOI] [PubMed] [Google Scholar]
  • 16.Neubauer BL, McNulty AM, Chedid M, Chen K, Goode RL,Johnson MA, Jones CD, Krishman V, Lynch R, Osborne HE and Graff JR: The selective estrogen receptor modulator trioxifene (LY133314) inhibits metastasis and extends survival in the PAIII prostatic carcinoma rat model. Cancer Res 63: 6056–6062, 2003. [PubMed] [Google Scholar]
  • 17.Cohen MB, Heidger PM and Lubaroff DM: Gross andmicroscopic pathology of induced prostatic complex tumors arising in Lobund-Wistar rats. Cancer Res 54: 626–628, 1994. [PubMed] [Google Scholar]
  • 18.Hoffer LJ, Levine M, Assouline S, Melnychuk D, Padayatty SJ,Rosadiuk K, Rousseau C, Robitaille L and Miller WH Jr: Phase I clinical trial of i.v. ascorbic acid in advanced malignancy Ann Oncol 19: 1969–1974, 2008. [DOI] [PubMed] [Google Scholar]
  • 19.Riordan HD, Casciari JJ, González MJ, Riordan NH, Miranda-Massari JR, Taylor P and Jackson JA: A pilot clinical study of continuous intravenous ascorbate in terminal cancer patients PR Health Sci J 24: 269–276, 2005. [PubMed] [Google Scholar]
  • 20.Pollard M and Luckert PH: The inhibitory effect of 4-hydroxyphenylretinamide (4HPR) on metastases of prostate adenocarcinoma-III cells in Lobund-Wistar rats. Cancer Lett 59: 159–163, 1991. [DOI] [PubMed] [Google Scholar]
  • 21.Neubauer BL, Best KL, Counts DF, Goode RL, Hoover DM, Jones CD, Sarosdy MF, Shaar CJ, Tanzer LR and Merriman RL: Raloxifene (LY156758) produces antimetastatic responses and extends survival in the PAIII rat prostatic adenocarcinoma model. Prostate 27: 220–229, 1995. [DOI] [PubMed] [Google Scholar]
  • 22.Neubauer BL, Bemis KG, Best KL, Goode RL, Hoover DM, Smith GF, Tanzer LR and Merriman RL: Inhibitory effect of warfarin on the metastasis of the PAIII prostatic adenocarcinoma in the rat. J Urol 135: 163–166, 1986. [DOI] [PubMed] [Google Scholar]
  • 23.Neubauer BL, Besat KL, Goode RL, Sundboom J, Tanzer LRand Merriman RL: Anticoagulant and inhibitory effects on the lymphatic metastases of the PAIII prostatic adenocarcinoma in male rats by recombinant-derived hirudin (rHV2) Cell Pharmacol 1: 113–119, 1994. [Google Scholar]
  • 24.Heaney ML, Gardner JR, Karasavvas N, Golde DW, Scheinberg DA, Smith EA and O’Connor OA: Vitamin C antagonizes the cytotoxic effects of antineoplastic drugs. Cancer Res 68: 80318038, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zweig MH and Campbell G: Receiver-operating characteristic (ROC) plots: a fundamental evaluation tool in clinical medicine. Clin Chem 39: 561–577, 1993. [PubMed] [Google Scholar]

RESOURCES