IP6

 

Supplement: International Research Conference on Food, Nutrition, and Cancer
Cancer Inhibition by Inositol Hexaphosphate (IP6) and Inositol: From Laboratory to Clinic1,2
Ivana Vucenik*,,3 and AbulKalam M. Shamsuddin

*Department of Medical and Research Technology and Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201

3 To whom correspondence should be addressed. E-mail: ivucenik@umaryland.edu.


ABSTRACT
TOP
ABSTRACT
LITERATURE CITED


Inositol hexaphosphate (IP6) is a naturally occurring polyphosphorylated carbohydrate that is present in substantial amounts in almost all plant and mammalian cells. It was recently recognized to possess multiple biological functions. A striking anticancer effect of IP6 was demonstrated in different experimental models. Inositol is also a natural constituent possessing moderate anticancer activity. The most consistent and best anticancer results were obtained from the combination of IP6 plus inositol. In addition to reducing cell proliferation, IP6 increases differentiation of malignant cells, often resulting in a reversion to normal phenotype. Exogenously administered IP6 is rapidly taken into the cells and dephosphorylated to lower-phosphate inositol phosphates, which further interfere with signal transduction pathways and cell cycle arrest. Enhanced immunity and antioxidant properties can also contribute to tumor cell destruction. However, the molecular mechanisms underlying this anticancer action are not fully understood. Because it is abundantly present in regular diet, efficiently absorbed from the gastrointestinal tract, and safe, IP6 holds great promise in our strategies for the prevention and treatment of cancer. IP6 plus inositol enhances the anticancer effect of conventional chemotherapy, controls cancer metastases, and improves the quality of life, as shown in a pilot clinical trial. The data strongly argue for the use of IP6 plus inositol in our strategies for cancer prevention and treatment. However, the effectiveness and safety of IP6 plus inositol at therapeutic doses needs to be determined in phase I and phase II clinical trials in humans.

--------------------------------------------------------------------------------

KEY WORDS: • prevention • treatment • differentiation • phytic acid

Cancer remains a major health problem in the United States and in other developed countries (1). In our continuing effort to reduce the public health burden of cancer, there is a constant search for more effective cancer treatment, and increased interest in the concept of prevention, as a promising approach to the control of cancer (2).

A novel anticancer function of inositol hexaphosphate (IP6;4 also InsP6 and phytic acid) has been shown both in vivo and in vitro (3–5). IP6 is a polyphosphorylated carbohydrate, contained in high concentrations (0.4–6.4%) in cereals and legumes (6). Myo-inositol is a parent compound of IP6. Only myo-inositol hexaphosphate has been found in plants; neo-, chiro-, and scyllo-inositol hexaphosphates have been isolated from soil (7). The phosphate grouping in positions 1, 2, and 3 (axial-equatorial-axial) is unique for IP6, providing a specific interaction with iron to completely inhibit its ability to catalyze hydroxyl radical formation, making IP6 a strong antioxidant, probably still the only role of IP6 that is widely recognized and accepted.

Almost all mammalian cells contain IP6 and much smaller amounts of its forms with fewer phosphate groups (IP1-5), which are important for regulating vital cellular functions. Inositol occurs ubiquitously in cell membranes in conjugation with lipids, as phosphatidylinositol. Recently, inositol phospholipids in the plasma membrane have received much attention because of their biological significance for signal transduction systems. Phosphatidylinositol 4,5-bisphosphate (PIP2), a phosphoinositide, is a precursor for several informational molecules in signal transduction—inositol 1,4,5-P3 (IP3), 1,2-diacylglycerol, and phosphatidylinositol 3,4,5-trisphosphate—linking receptor stimulation to Ca2+ mobilization (8). A second messenger role in intracellular Ca2+ homeostasis for IP4 was also shown. It is now recognized that subsequent to PIP2 hydrolysis a cascade of inositol phosphate metabolites are formed and that these multiple isomers show a complex pattern of interconversion (8–10). Inositol phosphates are versatile molecules with important roles in controlling diverse cellular activities (9,10). IP6 may serve as a natural antioxidant (11) and possibly as a neurotransmitter (10). Different binding proteins for inositol polyphosphates have been isolated, indicating their importance for the cellular functions (12) such as effects on ion channels and protein trafficking (13,14), endocytosis (15), exocytosis (16), and efficient export of mRNA from the nucleus to the cell (17).

How can exogenously administered IP6 affect tumor growth? Pioneering experiments showing this novel anticancer feature of IP6 were performed by Shamsuddin et al. (18–20), who were intrigued by the epidemiologic data indicating that only diets containing a high IP6 content (cereals and legumes) showed a negative correlation with colon cancer. Almost 15 y ago, Shamsuddin et al. hypothesized that IP6 can be internalized by the cells and dephosphorylated to IP1-5 and then can enter into the intracellular inositol phosphate pool and inhibit tumor growth. It was also hypothesized that the addition of inositol, a precursor of inositol phosphates and also a natural carbohydrate, to IP6 may enhance the anticancer function of IP6 (18–20). Because inositol phosphates are common molecules involved in signal transduction in most mammalian cell systems, it was further hypothesized that the anticancer action of inositol phosphates would be observed in different cells and tissue systems (18–20). All these proposed hypotheses have been confirmed.

Contrary to the dogma and skepticism at that time, we showed that IP6 is taken up by malignant cells (21) and that orally administered IP6 can reach target tumor tissue distant from the gastrointestinal tract (22). Because of the highly charged nature of IP6, it was a common misconception that it could not be transported into the cells. Analyzing absorption, intracellular distribution, and metabolism of IP6 in HT-29 human colon carcinoma and cells of hematopoietic lineage (K-562, human erythroleukemia and YAC-1, mouse lymphoma cells), we found that IP6 is rapidly taken up by mechanisms probably involving pinocytosis or receptor-mediated endocytosis, transported intracellularly, and dephosphorylated into inositol phosphates with fewer phosphate groups (21). Similar data were obtained when MCF-7 human breast cancer cells were incubated with [3H]-IP6 (SA 444 GBq/mmol, 370 Bq/106 cells): as early as 1 min after incubation, 3.1% of IP6-associated radioactivity was taken up by MCF-7 cells, and 9.5% after 1 h. By differential centrifugation 86% radioactivity was recovered from the cell cytosol. Anion-exchange chromatography showed that 58% of the absorbed radioactivity was in IP6 form. When [3H]-IP6 was administered intragastrically to rats, it was quickly absorbed from the stomach and upper intestine and distributed to various organs as early as 1 h after administration (22). Although the radioactivity isolated from gastric epithelium at this time was associated with inositol and IP1-6, the radioactivity in the plasma and urine was associated with inositol and IP1. These data indicate that the intact molecule was transported inside the gastric epithelial cells, wherein it was rapidly dephosphorylated, and that the metabolism of IP6 was very rapid. In our preliminary studies, [3H]-IP6 was given via oral gavage to rats bearing 7,12-dimethylbenz[a]anthracene-induced mammary tumors. A substantial amount of radioactivity (19.7% of all radioactivity recovered in collected tissues) was found in tumor tissue as early as 1 h after administration, providing at least partial explanation for the antineoplastic activity of IP6 at sites distant from the gastrointestinal tract. In this study only 50% of the radioactivity was excreted in urine within 72 h after administration; in addition feces accounted for another 10% of radioactivity, suggesting that at least 40% of the IP6-associated radioactivity was distributed within the animal tissues. These data indicate that IP6 can reach and concentrate at cellular targets. Chromatographic analysis of tumor tissue revealed the presence of inositol and IP1, similar to plasma.

Using a novel and highly sensitive method combining gas chromatography–mass spectrometry analysis and HPLC, Grases et al. (23,24) were able to identify IP6 in human urine and plasma and detect IP6 and its less-phosphorylated forms (IP3-5) in mammalian cells and in body fluids as they occur naturally. They also showed that the levels of IP6 and its less phosphorylated forms fluctuate depending on the intake of IP6.

That the extracellularly applied IP6 enters the cell and that this intracellular delivery is followed by a dephosphorylation of IP6 was recently confirmed by Ferry et al. (25).

Anticancer action of IP6

As hypothesized, it was demonstrated that IP6 is a broad-spectrum antineoplastic agent, affecting different cells and tissue systems. In vitro studies with IP6 are summarized in Table 1.


View this table:
[in this window]
[in a new window]
TABLE 1 Antitumor effect of inositol hexaphosphate (IP6) in vitro


IP6 inhibited the growth of all tested cell lines in a dose- and time-dependent manner. The growth of cells of hematopoietic lineage was inhibited: human leukemic hematopoietic cell lines, such as K-562 (26,27) and human normal and leukemic hematopoietic cells (27). The antiproliferative activity of IP6 was further reported in human colon cancer HT-29 cells (28), estrogen receptor–positive and estrogen receptor–negative human breast cancer cells (32), cervical cancer (25), prostate cancer (15,33,34), and HepG2 hepatoma cell lines (31). IP6 also inhibited the growth of mesenchymal tumors, murine fibrosarcoma (39), and human rhabdomyosarcoma (38). However, cells from different origin have different sensitivity to IP6 (the leukemic cell lines seem to be highly susceptible to IP6), suggesting that IP6 may affect different cell types through different mechanisms of action.
The potential of IP6 to induce differentiation and maturation of malignant cells, often resulting in reversion to the normal phenotype, was first demonstrated in K-562 hematopoietic cells (26). IP6 was further shown to increase differentiation of human colon carcinoma HT-29 cells (28,29), prostate cancer cells (33), breast cancer cells (32), and rhabdomyosarcoma cells (38).

The cancer preventive activity of IP6 in vitro was first tested in a benzo[a]pyrene-induced transformation in the rat tracheal cell culture transformation assay (30) and then was tested in a model using BALB/c mouse 3T3 fibroblasts (37) with modest efficacy. The observation that IP6 impaired the transformation induced by epidermal growth factor or phorbol ester in JB6 (mouse epidermal) cells (35) strongly suggested the potential role of IP6 as a cancer preventive agent, because this model has been a well-characterized cell system for studying the tumor promotion and molecular mechanisms of antitumor agents. Furthermore, IP6 reduced 12-O-tetradecanoylphorbol-13-acetate–induced ornithine decarboxylase activity, an essential event in tumor promotion in HEL-30 cells, a murine keratinocyte cell line (36).

A summary of in vivo studies using IP6 and inositol is shown in Table 2. Although experts in the field of nutrition and cancer have been performing in vivo experiments by adding IP6 to the diet, in all our cancer prevention studies, IP6 was given via drinking water in concentrations ranging from 0.4% to 2.0%. We were able to obtain comparable or even stronger tumor inhibition with much lower concentrations of IP6 when it was given in drinking water. For example, much stronger tumor inhibition was achieved with 0.4% IP6 in drinking water compared with the same amount given in a 20% high fiber diet (52).


View this table:
[in this window]
[in a new window]
TABLE 2 Antitumor effect of IP6 and inositol in vivo


The effectiveness of IP6 as a cancer preventive agent was shown in colon cancer induced in different species (rats and mice) with different carcinogens (1,2-dimethylhydrazine and azoxymethane) (18–20,40–46). IP6 was effective in a dose-dependent manner given either before or after carcinogen administration. The finding that IP6 was able to reduce the development of large intestinal cancer 5 mo after carcinogen administration, when IP6-treated animals demonstrated a significantly lower tumor number and size, has suggested its potential use as a therapeutic agent (20). IP6 decreased the incidence of aberrant crypts when they were used as an intermediate biomarker for colon cancer (43,44). Studies using other experimental models showed that antineoplastic properties of IP6 were not restricted to the colon. IP6 significantly reduced experimental mammary carcinoma in Sprague-Dawley rats induced either by 7,12-dimethylbenz[a]anthracene (51–54) or N-methylnitrosourea (42). Using a two-stage mouse skin carcinogenesis model, Ishikawa et al. (55) investigated the effect of IP6 on skin cancer and found a reduction in skin papillomas when IP6 was given during the initiation stage but not when given during the promotion stage (55).
The therapeutic properties of IP6 were demonstrated in the FSA-1 mouse model of transplantable and metastatic fibrosarcoma (39). After subcutaneous inoculation of mouse fibrosarcoma FSA-1 cells, mice were treated with intraperitoneal injections of IP6 and a significant inhibition of tumor size and survival over untreated controls was observed. In this model experimental lung metastases are developed after intravenous injections of FSA-1 cells; intraperitoneal injections of IP6 resulted in a significant reduction of metastatic lung colonies (39). A strong anticancer activity of IP6 was also demonstrated against human rhabdomyosarcoma RD cells transplanted in nude mice (38), where the efficacy of IP6 was tested on the tumor-forming capacity of RD cells. Peritumoral treatment with IP6 (40 mg/kg) initiated 2 d after subcutaneous injection of rhabdomyosarcoma cells suppressed the tumor growth by 25–49-fold (38). IP6 was also potent in inhibiting experimental hepatoma (31,48). We tested the effect of IP6 on tumorigenicity and tumor regression in this model. A single treatment of HepG2 cells in vitro by IP6 resulted in the complete loss of the ability of these cells to form tumors when inoculated subcutaneously in nude mice (48). Additionally, the preexisting liver cancers regressed when they were treated directly with IP6 (48).

Myo-inositol itself was also demonstrated to have anticancer function, albeit modest. It inhibited pulmonary adenoma formation in mice (49,50). We found that inositol alone or in combination with IP6 can prevent the formation and incidence of several cancers in experimental animals: in soft tissue, colon, metastatic lung, and mammary cancers. Additionally, we showed that inositol potentiates both the antiproliferative and antineoplastic effects of IP6 in vivo (3–5,19,39,51,52). Synergistic cancer inhibition by IP6 when combined with inositol was observed in colon cancer (Table 3) (19) and mammary cancer studies (Table 4) (51,52). Similar results were seen in the metastatic lung cancer model (39). Thus, the combination of IP6 and inositol was significantly better in different cancers than was either one alone.


View this table:
[in this window]
[in a new window]
TABLE 3 Synergistic cancer inhibition by IP6 when combined with inositol (Ins) 1,2-dimethylhydrazine (DMH)- induced colon carcinoma in mice


View this table:
[in this window]
[in a new window]
TABLE 4 7,12-Dimethylbenz[a]nthracene (DMBA)-induced mammary carcinoma in rats


Mechanisms of action of IP6
The mechanisms involved in the anticancer activity of inositol compounds are not fully understood. It is known that virtually all animal cells contain inositol phosphates and that the inositol phosphates with fewer phosphate groups, especially IP3 and IP4, have an important role in cellular signal transduction, regulation of cell function, growth, and differentiation (8,9). We hypothesized that one of the several ways by which IP6 plus inositol exerts its action is via lower-phosphate inositol phosphates. Measurement of intracellular inositol phosphates after IP6 treatment showed an increased level of lower-phosphate inositol phosphates (IP1-3) (21,24–26); their involvement in signal transduction pathways can affect cell cycle regulation, growth, and differentiation of malignant cells (3–5). Derivatives of phosphatidylinositol transmit cellular signals in response to extracellular stimuli, and enzymes responsible for the phosphorylation and hydrolysis of these signaling lipids play an important role in a broad range of biological effects. A central molecule is a phosphatidylinositol-3 kinase, which primarily phosphorylates the lipid phosphatidylinositol on the 3 position of the D-myo-inositol ring, yielding phosphatidylinositol-3-phosphate, but also can use phosphorylated forms of phosphatidylinositol as substrates. IP6 inhibits phosphatidylinositol-3 kinase (35). This action is related to the IP6 structure that is similar to D-3-deoxy-3-fluoro-PtdIns, an inhibitor of phosphatidylinositol-3 kinase (35). In addition to the blocking of phosphatidylinositol-3 kinase and activating protein-1 by IP6 (35), protein kinase C (16,57) and mitogen-activated protein kinases (15,35) are involved in IP6-mediated anticancer activity. The role of IP6 among these multiple signaling pathways and their cross-talk in regulation of cell functions needs to be addressed in the future.

IP6 can also modulate cellular response at the level of receptor binding. IP6, after sterically blocking the heparin-binding domain of basic fibroblast growth factor, disrupted further receptor interactions (58). This modulation in binding and the activity of basic fibroblast growth factor is thought to be due to the chair conformation of IP6 mimicking that of the pyranose ring structure in heparin (58).

The observed anticancer effect of inositol compounds could be mediated through several other mechanisms. The antioxidant role of IP6 is known and widely accepted; this function of IP6 occurs by chelation of Fe3+ and suppression of ·OH formation (11). Therefore, IP6 can reduce carcinogenesis mediated by active oxygen species and cell injury via its antioxidative function. This activity seems to be closely related to its unique structure. The phosphate grouping in positions 1,2,3 (axial-equatorial-axial) is unique to IP6, specifically interacting with iron to completely inhibit its ability to catalyze hydroxyl radical formation, making IP6 a strong antioxidant. This anticancer action of IP6 may be further related to mineral binding ability; IP6 by binding with Zn2+ can affect thymidine kinase activity, an enzyme essential for DNA synthesis, or remove iron, which may augment colorectal cancer (3–5,41,46).

Besides affecting tumor cells, IP6 can act on a host by restoring its immune system. IP6 augments natural killer cell activity in vitro and normalizes the carcinogen-induced depression of natural killer cell activity in vivo (59).

Value of IP6 as a therapeutic and preventive agent for cancer

Safety. IP6 is a natural compound and an important dietary component. Some concerns have been expressed regarding the mineral deficiency that results from an intake of foods high in IP6 that might reduce the bioavailability of dietary minerals. However, recent studies demonstrate that this antinutrient effect of IP6 can be manifested only when large quantities of IP6 are consumed in combination with a diet poor in oligoelements (60–63). A long-term intake of IP6 in food (60,61) or in a pure form (64) did not cause such a deficiency in humans. Studies in experimental animals showed no significant toxic effects on body weight, serum, or bone minerals (Table 5) or any pathological changes in either male F344 or female Sprague-Dawley rats for 40 wk (40,51,52). Grases et al. (65) confirmed our findings and also reported that abnormal calcification was prevented in rats given IP6.


View this table:
[in this window]
[in a new window]
TABLE 5 Effect of inositol compounds on bone minerals


IP6 does not affect normal cells. The most important expectation of a good anticancer agent is for it to only affect malignant cells and not affect normal cells and tissues. That property was recently shown for IP6. When the fresh CD34+ cells from bone marrow was treated with different doses of IP6, a toxic effect (inhibition of the clonogenic growth or as cytotoxicity on liquid cultures) was observed that was specific to leukemic progenitors from chronic myelogenous leukemia patients but no cytotoxic or cytostatic effect was observed on normal bone marrow progenitor cells under the same conditions (27). Recently, we (66) showed that IP6 inhibited the colony formation of Kaposi's sarcoma (KS) cell lines, KS Y-1 (AIDS-related KS) and KS SLK (Iatrogenic KS), and CCRF-CEM (human adult T lymphoma) cells in a dose-dependent manner (66). However, in striking contrast to taxol, used as a control, IP6 did not affect the ability of normal cells (peripheral blood mononuclear cells and T-cell colony-forming cells) to form colonies in a semisolid methylcellulose medium. Malignant and normal cells are known to have a different metabolism, growth rate, expression of receptors, etc., but the mechanism for this different selectivity of IP6 for normal and malignant cells needs to be further investigated.
IP6 acts synergistically with standard chemotherapeutics. Current cancer treatment recognizes the importance of using combination therapy to increase efficacy and decrease side effects of conventional chemotherapy. Another important aspect of cancer treatment is overcoming acquired drug resistance. Our recent data demonstrate that IP6 acts synergistically with doxorubicin and tamoxifen, being particularly effective against estrogen receptor–negative and doxorubicin-resistant cell lines, both conditions that are challenging to treat (67). These data are particularly important because tamoxifen is usually given as a chemopreventive agent in the posttreatment period and doxorubicin has enormous cardiotoxicity and its use is associated with doxorubicin resistance.

IP6 affects principal pathways of malignancy. Our goal is to identify agents that can target tumors at vulnerable sites and interrupt specific pathways of carcinogenesis. From the behavior and characteristics of malignant cells, several principal pathways of malignancy have been established, such as proliferation, cell cycle progression, metastases and invasion, angiogenesis, and apoptosis; interestingly, IP6 targets and acts on all of them.

Uncontrolled proliferation is a hallmark of malignant cells, and IP6 can reduce the cell proliferation rate of many different cell lines of different lineage and of both human and rodent origin (3–5,26,28,31–33,38). Although normal cells divide at a controlled and limited rate, malignant cells escape from the control mechanisms that regulate the frequency of cell multiplication and usually have lost the checkpoint controls that prevent replication of defective cells. IP6 can regulate the cell cycle to block uncontrolled cell division and force malignant cells either to differentiate or go into apoptosis. IP6 induces G1 phase arrest and a significant decrease of the S phase of human breast (68,69), colon (69), and prostate (34) cancer cell lines. However, IP6 causes the accumulation of human leukemia cells in the G2M phase of the cell cycle; a cDNA microarray analysis showed a down-modulation of multiple genes involved in transcription and cell-cycle regulation by IP6 (27).

One important characteristic of malignancy is the ability of tumor cells to metastasize and infiltrate normal tissue. A significant reduction in the number of lung metastatic colonies by IP6 was observed in a mouse metastatic tumor model using FSA-1 cells (39). Using highly invasive MDA-MB 231 human breast cancer cells, we demonstrated that IP6 inhibits metastasis in vitro through effects on cancer cell adhesion, migration, and invasion (70,71). Tumor cells emit substances known as matrix metalloproteinases that allow metastatic cells to pass into the blood vessels; IP6 significantly inhibited secretion of MMP-9 from MDA-MB 231 cells (70).

Tumors depend on the formation of new blood vessels to support their growth and metastasis. Many tumors produce large amounts of vascular endothelial growth factor, a cytokine that signals normal blood vessels to grow. IP6 inhibited the growth and differentiation of endothelial cells (66,72) and inhibited the secretion of vascular endothelial growth factor from malignant cells (27,66,72). IP6 can also adversely affect angiogenesis as antagonist of fibroblast growth factor (58).

Apoptosis is a hallmark of action of many anticancer drugs. It has been reported that IP6 induces apoptosis in vivo (45) and in vitro in prostate (34) and cervical cancer (25) cell lines, involving cleavage of caspase 3, caspase 9, and poly ADP-ribose polymerase, an apoptotic substrate, in a time- and dose-dependent manner.

Effectiveness of IP6 as a cancer preventive agent. Possible mechanisms of the cancer preventive action of IP6 include carcinogen blocking activities, antioxidant activities, and antiproliferation and antiprogression activities (73). Therefore, the strategy of chemoprevention is to use agents that will inhibit mutagenesis, induce apoptosis, induce maturation and differentiation, and inhibit proliferation (74). The antioxidant activity of IP6 is widely accepted and indisputable (11), and IP6 possesses antiproliferative and antiprogression activities. Its induction of terminal differentiation (26,28,29,32,33,38), restoration of immune response (59), modulation of growth factors (58), modulation of signal transduction pathways (15,16,35,57), induction of apoptosis (25,34,45), and possibly inhibition of oncogene activity and restoration of tumor suppressor function are well documented. IP6 not only inhibits the activities of some liver enzymes (75,76) but also significantly increases the hepatic levels of glutathione S-transferase (44,77), both of which indicate its possible role in carcinogen-blocking activities and cancer protection.

Although IP6 may belong to almost all previously mentioned categories of cancer preventive drugs, affecting almost all phases of cancer prevention, it still appears that IP6 is not a direct antagonist to the carcinogen because of its moderate efficacy in vitro when tested and compared with other chemopreventive agents (30) and a lack of dramatic decrease in cancer incidence when tested in vivo. However, because cancer prevention is a long process, a long administration of cancer preventive agent is generally needed, requiring usually 10–40 y of continuous treatment (2,73), and, therefore, it is very important that cancer preventive agents have low or almost no toxicity. IP6, a natural compound with virtually no toxicity, can satisfy this special and very important requirement for cancer prevention.

IP6 plus inositol and patients

An enhanced antitumor activity without compromising the patient's quality of life was demonstrated in a pilot clinical trial involving six patients with advanced colorectal cancer (Dukes C and D) with multiple liver and lung metastasis (78). IP6 plus inositol was given as an adjuvant to chemotherapy according to Mayo protocol. One patient with liver metastasis refused chemotherapy after the first treatment, and she was treated only with IP6 plus inositol; her control ultrasound and abdominal computed tomography scan 14 mo after surgery showed a significantly reduced growth rate. A reduced tumor growth rate was noticed overall and in some cases a regression of lesions was noted. Additionally, when IP6 plus inositol was given in combination with chemotherapy, side effects of chemotherapy (drop in leukocyte and platelet counts, nausea, vomiting, alopecia) were diminished and patients were able to perform their daily activities (78). Further controlled randomized clinical trials are necessary to confirm these observations.

Other biological effects of IP6

In humans, IP6 not only has almost no toxic effects, but it has many other beneficial health effects such as inhibition of kidney stone formation and reduction in risk of developing cardiovascular disease. IP6 was administered orally either as the pure sodium salt or in a diet to reduce hypercalciuria and to prevent formation of kidney stones, and no evidence of toxicity was reported (64,65,79,80). A potential hypocholesterolemic effect of IP6 may be very significant in the clinical management of hyperlipidemia and diabetes (75,76,81). IP6 inhibits agonist-induced platelet aggregation (82) and efficiently protects myocardium from ischemic damage and reperfusion injury (83), both of which are important for the management of cardiovascular diseases.

Many potential beneficial actions of IP6 have been described. The inclusion of IP6 plus inositol in our strategies for prevention and treatment of cancer as well as other chronic diseases is warranted. However, the effectiveness and safety of IP6 plus inositol need to be determined in Phase I and Phase II clinical trials in humans.


FOOTNOTES

1 Presented as part of a symposium, "International Research Conference on Food, Nutrition, and Cancer," given by the American Institute for Cancer Research and the World Cancer Research Fund International in Washington, D.C., July 17–18, 2003. This conference was supported by Balchem Corporation; BASF Aktiengesellschaft; California Dried Plum Board; The Campbell Soup Company; Danisco USA, Inc.; Hill's Pet Nutrition, Inc.; IP-6 International, Inc.; Mead Johnson Nutritionals; Roche Vitamins, Inc.; Ross Products Division; Abbot Laboratories; and The Solae Company. Guest editors for this symposium were Helen A. Norman and Ritva R. Butrum.

2 Studies from the authors' laboratories were supported by the American Institute for Cancer Research, the Susan Komen Breast Cancer Foundation, the University of Maryland Designated Research Initiative Fund, and the University of Maryland Women Health Research Foundation.

4 Abbreviations used: Ins, inositol; IP6, inositol hexaphosphate; IP3, inositol 1,4,5-P3; KS, Kaposi's sarcoma; PIP2, phosphatidylinositol 4,5-bisphosphate.


LITERATURE CITED
TOP
ABSTRACT
LITERATURE CITED


1. Jemal, A., Murray, T., Samuels, A., Ghafoor, A., Ward, E. & Thun, M. (2003) Cancer statistics, 2003. CA Cancer J. Clin. 53: 5–26.[Abstract/Free Full Text]

2. Kelloff, G. J., Hawk, E. T., Karp, J. E., Crowell, J. A., Boone, C. W., Steele, V. E., Lubet, R. A. & Sigman, C. C. (1997) Progress in clinical chemoprevention. Semin. Oncol. 24: 241–252.[Medline]

3. Shamsuddin, A. M. (2002) Anti-cancer function of phytic acid. Int. J. Food Sci. Technol. 37: 769–782.

4. Shamsuddin, A. M., Vucenik, I. & Cole, K. E. (1997) IP6: a novel anti-cancer agent. Life Sci. 61: 343–354.[Medline]

5. Shamsuddin, A. M. (1995) Inositol phosphates have novel anticancer function. J. Nutr. 125: 725S–732S.

6. Harland, B. F. & Oberleas, D. (1987) Phytate in foods. World Rev. Nutr. Diet. 52: 235–259.[Medline]

7. Reddy, N. R., Sathe, S. K. & Salunke, D. K. (1982) Phytates in legumes and cereals. Adv. Food Res. 28: 1–89.[Medline]

8. Berridge, M. J. & Irvine, R. F. (1989) Inositol phosphates and cell signalling. Nature 341: 197–205.[Medline]

9. Menniti, F. S., Oliver, K. G., Pytney, J. W., Jr. & Shears, S. B. (1993) Inositol phosphates and cell signalling: new view of InsP5 and InsP6. Trends Biochem. Sci. 18: 53–65.[Medline]

10. Sasakawa, N., Sharif, M. & Hanley, M. R. (1995) Metabolism and biological activities of inositol pentakisphosphates and inositol hexakisphosphate. Biochem. Pharmacol. 50: 137–146.[Medline]

11. Graf, E. & Eaton, J. W. (1990) Antioxidant functions of phytic acid. Free Radi. Biol. Med. 8: 61–69.

12. Huisaman, B. & Lochner, A. (1996) Inositol polyphosphates and their binding proteins-a short review. Mol. Cell. Biochem. 157: 229–232.[Medline]

13. Shears, S. B. (1996) Inositol pentakis- and hexakisphosphate metabolism adds versatility to the actions of inositol polyphosphates: novel effects on ion channels and protein traffic. Subcell. Biochem. 26: 187–226.[Medline]

14. Larsson, O., Barker, C. J., Sjöholm, Å., Carlqvist, H., Mitchell, R. H., Bertorello, A., Nilsson, T., Honkanen, R. E., Mayr, G. W., Zwiller, J. & Berggren, P.-O. (1997) Inhibition of phosphatases and increased Ca2+ channel activity by inositol hexakisphosphate. Science 278: 471–474.[Abstract/Free Full Text]

15. Zi, X., Singh, R. P. & Agarwal, R. (2000) Impairment of erbB1 receptor and fluid phase endocytosis and associated mitogenic signaling by inositol hexaphosphate in human prostate carcinoma DU145 cells. Carcinogenesis 21: 2225–2235.[Abstract/Free Full Text]

16. Efanov, A. M., Zaitsev, S. V. & Berggren, P.-O. (1997) Inositol hexakisphosphate stimulates non-Ca2+-mediated and primes Ca2+-mediated exocytosis of insulin by activation of protein kinase C. Proc. Natl. Acad. Sci. U.S.A. 94: 4435–4439.[Abstract/Free Full Text]

17. York, J. D., Odom, A. R., Murphy, R., Ives, E. B. & Wente, S. R. (1999) A phospholipase C-dependent inositol polyphosphate kinase pathway required for efficient messenger RNA export. Science 285: 96–100.[Abstract/Free Full Text]

18. Shamsuddin, A. M., Elsayed, A. & Ullah, A. (1988) Suppression of large intestinal cancer in F344 rats by inositol hexaphosphate. Carcinogenesis 9: 577–580.[Abstract/Free Full Text]

19. Shamsuddin, A. M., Ullah, A. & Chakravarthy, A. (1989) Inositol and inositol hexaphosphate suppress cell proliferation and tumor formation in CD-1 mice. Carcinogenesis 10: 1461–1463.[Abstract/Free Full Text]

20. Shamsuddin, A. M. & Ullah, A. (1989) Inositol hexaphosphate inhibits large intestinal cancer in F344 rats 5 months after induction by azoxymethane. Carcinogenesis 10: 625–626.[Abstract/Free Full Text]

21. Vucenik, I. & Shamsuddin, A. M. (1994) [3H]-Inositol hexaphosphate (phytic acid) is rapidly absorbed and metabolized by murine and human malignant cells in vitro. J. Nutr. 124: 861–868.

22. Sakamoto, K., Vucenik, I. & Shamsuddin, A. M. (1993) [3H]Phytic acid (inositol hexaphosphate) is absorbed and distributed to various tissues in rats. J. Nutr. 123: 713–720.

23. Grases, F., Simonet, B. M., Vucenik, I., Prieto, R. M., Costa-Bauzá, A., March, J. G. & Shamsuddin, A. M. (2001) Absorption and excretion of orally administered inositol hexaphosphate (IP6 or phytate) in humans. Biofactors 15: 53–61.[Medline]

24. Grases, F., Simonet, B. M., Vucenik, I., Perelló, J., Prieto, R. M. & Shamsuddin, A. M. (2002) Effects of exogenous inositol hexakiphosphate (InsP6) on the levels of InsP6 and of inositol trisphosphate (InsP3) in malignant cells, tissues and biological fluids. Life Sci. 71: 1535–1546.[Medline]

25. Ferry, S., Matsuda, M., Yoshida, H. & Hirata, M. (2002) Inositol hexakisphosphate blocks tumor cell growth by activating apoptotic machinery as well as by inhibiting the Akt/NFB-mediated cell survival pathway. Carcinogenesis 23: 2031–2041.[Abstract/Free Full Text]

26. Shamsuddin, A. M., Baten, A. & Lalwani, N. D. (1992) Effect of inositol hexaphosphate on growth and differentiation in K562 erythroleukemia cell line. Cancer Lett. 64: 195–202.[Medline]

27. Deliliers, L. G., Servida, G., Fracchiolla, N. S., Ricci, C., Borsotti, C., Colombo, G. & Soligo, D. (2002) Effects of inositol hexaphosphate (IP6) on human normal and leukaemic hematopoietic cells. Br. J. Haematol. 117: 577–587.[Medline]

28. Sakamoto, K., Venkatraman, G. & Shamsuddin, A. M. (1993) Growth inhibition and differentiation of HT-29 cells in vitro by inositol hexaphosphate (phytic acid). Carcinogenesis 14: 1815–1819.[Abstract/Free Full Text]

29. Yang, G.-Y. & Shamsuddin, A. M. (1995) IP6-induced growth inhibition and differentiation of HT-29 human colon cancer cells: involvement of intracellular inositol phosphates. Anticancer Res. 15: 2479–2488.[Medline]

30. Arnold, J. T., Wilkinson, B. P., Sharma, S. & Steele, V. E. (1993) Evaluation of chemopreventative agents in different mechanistic classes using a rat epithelial cell culture transformation assay. Cancer Res. 73: 537–543.

31. Vucenik, I., Tantivejkul, K., Zhang, Z. S., Cole, K. E., Saied, I. & Shamsuddin, A. M. (1998) IP6 treatment of liver cancer. I. IP6 inhibits growth and reverses transformed phenotype in HepG2 human liver cancer cell line. Anticancer Res. 18: 4083–4090.[Medline]

32. Shamsuddin, A. M., Yang, G.-Y. & Vucenik, I. (1996) Novel anti-cancer functions of IP6: growth inhibition and differentiation of human mammary cancer cell lines in vitro. Anticancer Res. 16: 3287–3292.[Medline]

33. Shamsuddin, A. M. & Yang, G.-Y. (1995) Inositol hexaphosphate inhibits growth and induces differentiation of PC-3 human prostate cancer cells. Carcinogenesis 16: 1975–1979.[Abstract/Free Full Text]

34. Singh, R. P., Agarwal, C. & Agarwal, R. (2003) Inositol hexaphosphate inhibits growth, and induces G1 arrest and apoptotic death of prostate carcinoma DU145: modulation of CDKI-CDK-cyclin and pRb-related protein-E2F complexes. Carcinogenesis 24: 555–563.[Abstract/Free Full Text]

35. Huang, C., Ma, W.-Y., Hecht, S. S. & Dong, Z. (1997) Inositol hexaphosphate inhibits cell transformation and activator protein 1 activation by targeting phosphatidylinositol-3' kinase. Cancer Res. 57: 2873–2878.[Abstract/Free Full Text]

36. Nickel, K. P. & Belury, M. A. (1999) Inositol hexaphosphate reduces 12-O-tetradecanoylphorbol-13-acetate-induced ornithine decarboxylase independent of protein kinase C isoform expression in keratinocytes. Cancer Lett. 140: 105–111.[Medline]

37. Babich, H., Borenfreund, E. & Stern, A. (1993) Comparative toxicities of selected minor dietary non-nutrients with chemopreventive properties. Cancer Lett. 73: 127–133.[Medline]

38. Vucenik, I., Kalebic, T., Tantivejkul, K. & Shamsuddin, A. M. (1998) Novel anticancer function of inositol hexaphosphate (IP6): inhibition of human rhabdomyosarcoma in vitro and in vivo. Anticancer Res. 18: 1377–1384.[Medline]

39. Vucenik, I., Tomazic, V. J., Fabian, D. & Shamsuddin, A. M. (1992) Antitumor activity of phytic acid (inositol hexaphosphate) in murine transplanted and metastatic fibrosarcoma, a pilot study. Cancer Lett. 65: 9–13.[Medline]

40. Ullah, A. & Shamsuddin, A. M. (1990) Dose-dependent inhibition of large intestinal cancer by inositol hexaphosphate in F344 rats. Carcinogenesis 11: 2219–2222.[Abstract/Free Full Text]

41. Nelson, R. L., Yoo, S. J., Tanure, J. C., Andrianopoulos, G. & Misumi, A. (1989) The effect of iron on experimental colorectal carcinogenesis. Anticancer Res. 9: 1477–1482.[Medline]

42. Shivapurkar, N., Tang, Z. C., Frost, A. & Alabaster, O. (1996) A rapid dual organ rat carcinogenesis bioassay for evaluating the chemoprevention of breast and colon cancer. Cancer Lett. 100: 169–179.[Medline]

43. Pretlow, T. P., O'Riordan, M. A., Somich, G. A., Amini, S. B. & Pretlow, T. G. (1992) Aberrant crypts correlate with tumor incidence in F344 rats treated with azoxymethane and phytate. Carcinogenesis 13: 1509–1512.[Abstract/Free Full Text]

44. Challa, A., Rao, D. R. & Reddy, B. S. (1997) Interactive suppression of aberrant crypt foci induced by azoxymethane in rat colon by phytic acid and green tea. Carcinogenesis 18: 2023–2026.[Abstract/Free Full Text]

45. Jenab, M. & Thompson, L. U. (2000) Phytic acid in wheat bran affects cell morphology, differentiation and apoptosis. Carcinogenesis 21: 1547–1552.[Abstract/Free Full Text]

46. Thompson, L. U. & Zhang, L. (1991) Phytic acid and minerals: effect of early markers of risk for mammary and colon carcinogenesis. Carcinogenesis 12: 2041–2045.[Abstract/Free Full Text]

47. Hirose, M., Ozaki, K., Takaba, K., Fukushima, S., Shirai, T. & Ito, N. (1991) Modifying effects of the naturally occuring antioxidants -oryzanol, phytic acid, tannic acid and n-tritriacontane-16,18-dione in rat a wide-spectrum organ carcinogenesis model. Carcinogenesis 12: 1917–1921.[Abstract/Free Full Text]

48. Vucenik, I., Zhang, Z. S. & Shamsuddin, A. M. (1998) IP6 in treatment of liver cancer. II. Intra-tumoral injection of IP6 regresses pre-existing human liver cancer xenotransplanted in nude mice. Anticancer Res. 18: 4091–4096.[Medline]

49. Estensen, R. D. & Wattenberg, L. W. (1993) Studies of chemopreventive effects of myo-inositol on benzo[a]pyrene-induced neoplasia of the lung and forestomach of female A/J mice. Carcinogenesis 14: 1975–1977.[Abstract/Free Full Text]

50. Wattenberg, L. W. (1995) Chalcones, myo-inositol and other novel inhibitors of pulmonary carcinogenesis. J. Cell. Biochem. 22: 162–168.

51. Vucenik, I., Sakamoto, K., Bansal, M. & Shamsuddin, A. M. (1993) Inhibition of mammary carcinogenesis by inositol hexaphosphate (phytic acid). A pilot study. Cancer Lett. 75: 95–102.[Medline]

52. Vucenik, I., Yang, G.-Y. & Shamsuddin, A. M. (1995) Inositol hexaphosphate and inositol inhibit DMBA-induced rat mammary cancer. Carcinogenesis 16: 1055–1058.[Abstract/Free Full Text]

53. Vucenik, I., Yang, G. & Shamsuddin, A. M. (1997) Comparison of pure inositol hexaphosphate (IP6) and high-bran diet in the prevention of DMBA-induced rat mammary carcinogenesis. Nutr. Cancer 28: 7–13.[Medline]

54. Hirose, M., Hoshiya, T., Akagi, K., Futakushi, M. & Ito, N. (1994) Inhibition of mammary gland carcinogenesis by green tea catechin and other naturally occuring antioxidants in Sprague-Dawley rats pretreated with 7,12-dimethylbenz[a]anthracene. Cancer Lett. 83: 149–156.[Medline]

55. Ishikawa, T., Nakatsuru, Y., Zarkovic, M. & Shamsuddin, A. M. (1999) Inhibition of skin cancer by IP6 in vivo: initiation-promotion model. Anticancer Res. 19: 3749–3752.[Medline]

56. Jariwalla, R. J., Sabin, R., Lawson, S., Bloch, D. A., Prender, M., Andrews, V. & Herman, Z. S. (1988) Effects of dietary phytic acid (phytate) on the incidence and growth rate of tumors promoted in Fisher rats by a magnesium supplement. Nutr. Res. 8: 813–827.

57. Vucenik, I., Ramakrishna, G., Tantivejkul, K., Anderson, L. & Ramljak, D. (1999) Inositol hexaphosphate (IP6) differentially modulates the expression of PKC in MCF-7 and MDA-MB 231 cells. Proc. Am. Assoc. Cancer Res. 40: 653.

58. Morisson, R. S., Shi, E., Kan, M., Yamaguchi, F., McKeehan, W., Rudnicka-Nawrot, M. & Palczewski, K. (1994) Inositol hexaphosphate (InsP6): An antagonist of fibroblast growth factor receptor binding and activity. In vitro cell. Dev. Biol. 30A: 783–789.

59. Baten, A., Ullah, A., Tomazic, V. J. & Shamsuddin, A. M. (1989) Inositol-phosphate-induced enhancement of natural killer cell activity correlates with tumor suppression. Carcinogenesis 10: 1595–1598.[Abstract/Free Full Text]

60. Walker, A. R. P., Fox, F. W. & Irving, J. T. (1948) Studies in human mineral metabolism. I. The effect of bread rich in phytate phosphorous on the metabolism of certain mineral salts with special reference to calcium. Biochem. J. 42: 452–462.[Medline]

61. Cullumbine, H., Basnayake, V. & Lemottee, J. (1950) Mineral metabolism on rice diets. Br. J. Nutr. 4: 101–111.[Medline]

62. Kelsay, J. L. (1987) Effect of fiber, phytic acid, and oxalic acid in the diet on mineral bioavailability. Am. J. Gastroenterol. 278: 983–986.

63. Sandstrom, B., Bugel, S., McGaw, B. A., Price, J. & Reid, M. D. (2000) A high oat-bran intakes does not impair zinc absorption in human when added to a low-fiber animal protein-based diet. J. Nutr. 130: 594–599.[Abstract/Free Full Text]

64. Henneman, P. H., Benedict, P. H., Forbes, A. P. & Dudley, H. R. (1958) Idiopathic hypercalciuria. N. Engl. J. Med. 17: 802–807.

65. Grases, F., García-Gonsales, R., Torres, J. J. & Llobera, A. (1988) Effects of phytic acid on renal stone formation in rats. Scand. J. Urol. Nephrol. 32: 261–265.

66. Tran, H. C., Brooks, J., Gadwal, S., Bryant, J. L., Shamsuddin, A. M., Lunardi-Iskandar, Y. & Vucenik, I. (2003) Effect of inositol hexaphosphate (IP6) on AIDS neoplastic Kaposi's sarcoma, iatrogenic Kaposi's sarcoma and lymphoma. Proc. Am. Assoc Cancer Res. 44: 499.

67. Tantivejkul, K., Vucenik, I., Eiseman, J. & Shamsuddin, A. M. (2003) Inositol hexaphosphate (IP6) enhances the anti-proliferative effects of adriamycin and tamoxifen in breast cancer. Breast Cancer Res. Treat. 79: 301–312.[Medline]

68. Vucenik, I., Tantivejkul, K., Ramakrishna, G., Anderson, L. M. & Ramljak, D. (2000) Antiproliferative effect inositol hexaphosphate (IP6) in breast cancer cells is mediated by increase in p27 and decrease in Rb protein phosphorylation. Proc. Am. Assoc. Cancer Res. 41: 339.

69. El-Sherbiny, Y., Cox, M. C., Ismail, Z. A., Shamsuddin, A. M. & Vucenik, I. (2001) G0/G1 arrest and S phase inhibition of human cancer cell lines by inositol hexaphosphate (IP6). Anticancer Res. 21: 2393–2404.[Medline]

70. Tantivejkul, K., Vucenik, I. & Shamsuddin, A. M. (2003) Inositol hexaphosphate (IP6) inhibits key events of cancer metastases: I. In vitro studies of adhesion, migration and invasion of MDA-MB 231 human breast cancer cells. Anticancer Res. 23: 5 (in press).

71. Tantivejkul, K., Vucenik, I. & Shamsuddin, A. M. (2003) Inositol hexaphosphate (IP6) inhibits key events of cancer metastases: II. Effects on integrins and focal adhesions. Anticancer Res. 23: 5 (in press).

72. Vucenik, I., Passaniti, A., Tantivejkul, K., Eggleton, P., Vitolo, M. & Shamsuddin, A. M. (2002) Anti-angiogenic potential of inositol hexaphophate (IP6). Rev. Oncologia 4 (Suppl 1): 79.

73. Kelloff, G. J., Boone, C. W., Steele, V. E., Fay, J. R., Lubet, R. A., Crowell, J. A. & Sigman, C. C. (1994) Mechanistic consideration in chemopreventive drug development. J. Cell. Biochem. 20: 1–24.

74. Boone, C. W., Kelloff, G. J. & Malone, W. F. (1990) Identification of candidate cancer chemopreventive agents and their evaluation in animal models and human clinical trials. Cancer Res. 50: 2–9.[Abstract/Free Full Text]

75. Katayama, T. (1995) Effect of dietary sodium phytate on the hepatic and serum levels of lipids and on the hepatic activities of NADH-generating enzymes in rats fed on sucrose. Biosci. Biotechnol. Biochem. 59: 1159–1160.[Medline]

76. Katayama, T. (1997) Effects of dietary myo-inositol or phytic acid on hepatic concentrations of lipids and hepatic activities of lipogenic enzymes in rats fed on corn starch or sucrose. Nutr. Res. 4: 721–728.

77. Singh, A., Singh, S. P. & Bamezai, R. (1997) Modulatory influence of arecoline on the phytic acid-altered hepatic biotransformation system enzymes, sulfhydryl content and lipid peroxidation in a murine system. Cancer Lett. 117: 1–6.[Medline]

78. Druzijanic, N., Juricic, J., Perko, Z. & Kraljevic, D. (2002) IP-6 & inositol: adjuvant to chemotherapy of colon cancer. A pilot clinical trial. Rev. Oncologia 4: 171.

79. Ohkawa, T., Ebisuno, S., Kitagawa, M., Marimoto, S., Miyazaki, Y. & Yasukawa, S. (1984) Rice bran treatment for patients with hypercalciuric stones: experimental and clinical studies. J. Urol. 132: 1140–1145.[Medline]

80. Grases, F., March, J. G., Prieto, R. M., Simonet, B. M., Costa-Bauzá, A., Garcia-Raja, A. & Conte, A. (2000) Urinary phytate in calcium oxalate stone-formers and healthy people. Dietary effects on phytate excretion. Scand. J. Urol. Nephrol. 34: 162–164.[Medline]

81. Jariwalla, R. J., Sabin, R., Lawson, S. & Herman, Z. S. (1990) Lowering of serum cholesterol and triglycerides and modulations by dietary phytate. J. Appl. Nutr. 42: 18–28.

82. Vucenik, I., Podczasy, J. J. & Shamsuddin, A. M. (1999) Antiplatelet activity of inositol hexaposphate (IP6). Anticancer Res. 19: 3689–3693.[Medline]

83. Rao, P. S., Liu, X. K., Das, D. K., Weinstein, G. S. & Tyras, D. H. (1991) Protection of ischemic heart from reperfusion injury by myo-inositol hexaphosphate, a natural antioxidant. Ann. Thorac. Surg. 52: 908–912.[Abstract]

BACK