Antitumoral effect of Ocoxin on acute myeloid leukemia

Elena Díaz-Rodríguez, Susana Hernández-García, Eduardo Sanz, Atanasio Pandiella, Elena Díaz-Rodríguez, Susana Hernández-García, Eduardo Sanz, Atanasio Pandiella

Abstract

Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy whose incidence is growing in developed countries. In the relapse setting, very limited therapeutic options are available and in most cases only palliative care can be offered to patients. The effect of a composite formulation that contains several antioxidants, Ocoxin Oral solution (OOS), was tested in this condition. When analyzed in vitro, OOS exhibited anti-AML action that was both time and dose dependent. In vivo OOS induced a ralentization of tumor growth that was due to a decrease in cell proliferation. Such effect could, at least partially, be due to an increase in the cell cycle inhibitor p27, although other cell cycle proteins seemed to be altered. Besides, OOS induced an immunomodulatory effect through the induction of IL6. When tested in combination with other therapeutic agents normally used in the treatment of AML patients, OOS demonstrated a higher antiproliferative action, suggesting that it may be used in combination with those standard of care treatments to potentiate their antiproliferative action in the AML clinic.

Keywords: acute myeloid leukemia; antioxidants; cell cycle; p27.

Conflict of interest statement

CONFLICTS OF INTEREST

E.S. is an employee of Catalysis S.L. The research costs of this work were partially supported by Catalysis S.L.

Figures

Figure 1. Efficacy of OOS on AML…
Figure 1. Efficacy of OOS on AML cell lines in vitro
Dose-dependent effect of OOS on the proliferation of HEL, KG1 or HL60 AML cells was assessed in vitro. Cells were incubated with OOS at the indicated dilution factors and MTT metabolization was measured at 24 (A), 48 (B), 72 (C) or 96 (D) hours. Mean absorbance values of untreated samples were taken as 100% and then mean values referred to that. Data are represented as mean ± SD of quadruplicates of an experiment that was repeated at least twice.
Figure 2. OOS is more effective in…
Figure 2. OOS is more effective in combination with standard of care treatments
The effect of OOS alone (1:50) or in combination with Ara C (250 nM, A), Doxorubicin (DXR, 100 nM, B) or Fludarabine (Fluda, 250 nM, C) were determined in MTT assays. The mean absorbance values of untreated samples from each were considered as 100%. Data are represented as mean ± SD of quadruplicates of an experiment that was repeated at least twice.
Figure 3. Efficacy of OOS on AML…
Figure 3. Efficacy of OOS on AML models in vivo
(A) OOS interferes with tumor growth. Female CB17-SCID athymic mice were injected with HEL cells. When tumors became palpable and were growing, they were randomized to different groups that were orally treated 5 days per week (Monday to Friday) with 100μl OOS/animal (■) or vehicle alone (water,♦), and tumor volumes were measured twice a week. Data are represented as mean tumor volume ± SEM of the animals on each group. (B) Effect of OOS on animal weight. Statistical significant differences are shown (*p

Figure 4. Action of OOS on immune…

Figure 4. Action of OOS on immune system cytokines

At the time of sacrifice, blood…

Figure 4. Action of OOS on immune system cytokines
At the time of sacrifice, blood from the animals under the different treatments was collected by intracardiac injection, and sera clarified and frozen. The amount of the indicated cytokines was determined by flow cytometry using a BD Cytometric Bead Array as described in the materials and methods section. The amount for each molecule and experimental condition are shown in the corresponding boxplots, as indicated. Statistical significant differences are shown (*p

Figure 5. OOS induces a decrease in…

Figure 5. OOS induces a decrease in tumor proliferation

(A) OOS does not induce apoptotic…

Figure 5. OOS induces a decrease in tumor proliferation
(A) OOS does not induce apoptotic cell death on AML tumors. For each experimental condition, two tumors were randomly processed for IHQ analysis and apoptotic cells were detected by tunnel staining. The number of apoptotic cells/field was quantified for each condition and its mean number ± SD is shown in the graph. Pictures of representative fields stained for this marker are shown on the right. (B) Similarly the number or endothelial cells was measured by CD31 staining. (C) Besides, to establish the proliferative status of the tumors, Ki67 marker was used. Pictures of representative fields stained for Ki-67 are shown on the right upper row and lower pictures show part of them (inset) at higher magnification. Statistical significant differences are shown (*p

Figure 6. OOS causes accumulation of p27…

Figure 6. OOS causes accumulation of p27 and G1 arrest

(A) Quantitation of the in…

Figure 6. OOS causes accumulation of p27 and G1 arrest
(A) Quantitation of the in vivo levels of cell cycle related proteins. The amount of the indicated proteins in the tumors was assessed by conventional WB. (B) The amount of p27 was quantified using the NIH image software, and the mean ± SD of such amount is represented in the graph. (C) Cell cycle analysis of HEL cells treated for 24 hours with OOS (1/50 dilution). The percentage of cells in the different phases of the cell cycle is shown. (D) Action of OOS (1/50) on p27 levels. HEL cells were treated for 24 hours with OOS and cell extracts prepared to analyze p27 by WB. GAPDH was used as a loading control.

Figure 7. In vivo effect of OOS…

Figure 7. In vivo effect of OOS on gene expression profiles

(A) Hierarchical clustering of…

Figure 7. In vivo effect of OOS on gene expression profiles
(A) Hierarchical clustering of the 6 tumors and the 37 genes deregulated after OOS treatment. Each row represents a gene and each column represents a tumor (1- control, 2- OOS treated). The expression level of each gene in each tumor is relative to its medium abundance across all the tumors and is depicted according to the color scale shown. Red and green indicate high or low expression levels, respectively. (B) The gene expression profile of three control tumors was compared to that of three tumors treated with 100 μl of OOS. A list with those genes whose expression changed above 2 times with the treatment is shown in the figure.
All figures (7)
Similar articles
Cited by
References
    1. Aleem E, Arceci RJ. Targeting cell cycle regulators in hematologic malignancies. Front Cell Dev Biol. 2015;3:16. - PMC - PubMed
    1. Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, Anderson JE, Petersdorf SH. Age and acute myeloid leukemia. Blood. 2006;107:3481–3485. - PMC - PubMed
    1. Juliusson G, Antunovic P, Derolf A, Lehmann S, Mollgard L, Stockelberg D, Tidefelt U, Wahlin A, Hoglund M. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 2009;113:4179–4187. - PubMed
    1. Leone G, Mele L, Pulsoni A, Equitani F, Pagano L. The incidence of secondary leukemias. Haematologica. 1999;84:937–945. - PubMed
    1. Renella R, Verkooijen HM, Fioretta G, Vlastos G, Kurtz J, Sappino AP, Schafer P, Neyroud-Caspar I, Bouchardy C. Increased risk of acute myeloid leukemia after treatment for breast cancer. Breast. 2006;15:614–619. - PubMed
Show all 27 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4. Action of OOS on immune…
Figure 4. Action of OOS on immune system cytokines
At the time of sacrifice, blood from the animals under the different treatments was collected by intracardiac injection, and sera clarified and frozen. The amount of the indicated cytokines was determined by flow cytometry using a BD Cytometric Bead Array as described in the materials and methods section. The amount for each molecule and experimental condition are shown in the corresponding boxplots, as indicated. Statistical significant differences are shown (*p

Figure 5. OOS induces a decrease in…

Figure 5. OOS induces a decrease in tumor proliferation

(A) OOS does not induce apoptotic…

Figure 5. OOS induces a decrease in tumor proliferation
(A) OOS does not induce apoptotic cell death on AML tumors. For each experimental condition, two tumors were randomly processed for IHQ analysis and apoptotic cells were detected by tunnel staining. The number of apoptotic cells/field was quantified for each condition and its mean number ± SD is shown in the graph. Pictures of representative fields stained for this marker are shown on the right. (B) Similarly the number or endothelial cells was measured by CD31 staining. (C) Besides, to establish the proliferative status of the tumors, Ki67 marker was used. Pictures of representative fields stained for Ki-67 are shown on the right upper row and lower pictures show part of them (inset) at higher magnification. Statistical significant differences are shown (*p

Figure 6. OOS causes accumulation of p27…

Figure 6. OOS causes accumulation of p27 and G1 arrest

(A) Quantitation of the in…

Figure 6. OOS causes accumulation of p27 and G1 arrest
(A) Quantitation of the in vivo levels of cell cycle related proteins. The amount of the indicated proteins in the tumors was assessed by conventional WB. (B) The amount of p27 was quantified using the NIH image software, and the mean ± SD of such amount is represented in the graph. (C) Cell cycle analysis of HEL cells treated for 24 hours with OOS (1/50 dilution). The percentage of cells in the different phases of the cell cycle is shown. (D) Action of OOS (1/50) on p27 levels. HEL cells were treated for 24 hours with OOS and cell extracts prepared to analyze p27 by WB. GAPDH was used as a loading control.

Figure 7. In vivo effect of OOS…

Figure 7. In vivo effect of OOS on gene expression profiles

(A) Hierarchical clustering of…

Figure 7. In vivo effect of OOS on gene expression profiles
(A) Hierarchical clustering of the 6 tumors and the 37 genes deregulated after OOS treatment. Each row represents a gene and each column represents a tumor (1- control, 2- OOS treated). The expression level of each gene in each tumor is relative to its medium abundance across all the tumors and is depicted according to the color scale shown. Red and green indicate high or low expression levels, respectively. (B) The gene expression profile of three control tumors was compared to that of three tumors treated with 100 μl of OOS. A list with those genes whose expression changed above 2 times with the treatment is shown in the figure.
All figures (7)
Similar articles
Cited by
References
    1. Aleem E, Arceci RJ. Targeting cell cycle regulators in hematologic malignancies. Front Cell Dev Biol. 2015;3:16. - PMC - PubMed
    1. Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, Anderson JE, Petersdorf SH. Age and acute myeloid leukemia. Blood. 2006;107:3481–3485. - PMC - PubMed
    1. Juliusson G, Antunovic P, Derolf A, Lehmann S, Mollgard L, Stockelberg D, Tidefelt U, Wahlin A, Hoglund M. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 2009;113:4179–4187. - PubMed
    1. Leone G, Mele L, Pulsoni A, Equitani F, Pagano L. The incidence of secondary leukemias. Haematologica. 1999;84:937–945. - PubMed
    1. Renella R, Verkooijen HM, Fioretta G, Vlastos G, Kurtz J, Sappino AP, Schafer P, Neyroud-Caspar I, Bouchardy C. Increased risk of acute myeloid leukemia after treatment for breast cancer. Breast. 2006;15:614–619. - PubMed
Show all 27 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 5. OOS induces a decrease in…
Figure 5. OOS induces a decrease in tumor proliferation
(A) OOS does not induce apoptotic cell death on AML tumors. For each experimental condition, two tumors were randomly processed for IHQ analysis and apoptotic cells were detected by tunnel staining. The number of apoptotic cells/field was quantified for each condition and its mean number ± SD is shown in the graph. Pictures of representative fields stained for this marker are shown on the right. (B) Similarly the number or endothelial cells was measured by CD31 staining. (C) Besides, to establish the proliferative status of the tumors, Ki67 marker was used. Pictures of representative fields stained for Ki-67 are shown on the right upper row and lower pictures show part of them (inset) at higher magnification. Statistical significant differences are shown (*p

Figure 6. OOS causes accumulation of p27…

Figure 6. OOS causes accumulation of p27 and G1 arrest

(A) Quantitation of the in…

Figure 6. OOS causes accumulation of p27 and G1 arrest
(A) Quantitation of the in vivo levels of cell cycle related proteins. The amount of the indicated proteins in the tumors was assessed by conventional WB. (B) The amount of p27 was quantified using the NIH image software, and the mean ± SD of such amount is represented in the graph. (C) Cell cycle analysis of HEL cells treated for 24 hours with OOS (1/50 dilution). The percentage of cells in the different phases of the cell cycle is shown. (D) Action of OOS (1/50) on p27 levels. HEL cells were treated for 24 hours with OOS and cell extracts prepared to analyze p27 by WB. GAPDH was used as a loading control.

Figure 7. In vivo effect of OOS…

Figure 7. In vivo effect of OOS on gene expression profiles

(A) Hierarchical clustering of…

Figure 7. In vivo effect of OOS on gene expression profiles
(A) Hierarchical clustering of the 6 tumors and the 37 genes deregulated after OOS treatment. Each row represents a gene and each column represents a tumor (1- control, 2- OOS treated). The expression level of each gene in each tumor is relative to its medium abundance across all the tumors and is depicted according to the color scale shown. Red and green indicate high or low expression levels, respectively. (B) The gene expression profile of three control tumors was compared to that of three tumors treated with 100 μl of OOS. A list with those genes whose expression changed above 2 times with the treatment is shown in the figure.
All figures (7)
Figure 6. OOS causes accumulation of p27…
Figure 6. OOS causes accumulation of p27 and G1 arrest
(A) Quantitation of the in vivo levels of cell cycle related proteins. The amount of the indicated proteins in the tumors was assessed by conventional WB. (B) The amount of p27 was quantified using the NIH image software, and the mean ± SD of such amount is represented in the graph. (C) Cell cycle analysis of HEL cells treated for 24 hours with OOS (1/50 dilution). The percentage of cells in the different phases of the cell cycle is shown. (D) Action of OOS (1/50) on p27 levels. HEL cells were treated for 24 hours with OOS and cell extracts prepared to analyze p27 by WB. GAPDH was used as a loading control.
Figure 7. In vivo effect of OOS…
Figure 7. In vivo effect of OOS on gene expression profiles
(A) Hierarchical clustering of the 6 tumors and the 37 genes deregulated after OOS treatment. Each row represents a gene and each column represents a tumor (1- control, 2- OOS treated). The expression level of each gene in each tumor is relative to its medium abundance across all the tumors and is depicted according to the color scale shown. Red and green indicate high or low expression levels, respectively. (B) The gene expression profile of three control tumors was compared to that of three tumors treated with 100 μl of OOS. A list with those genes whose expression changed above 2 times with the treatment is shown in the figure.

References

    1. Aleem E, Arceci RJ. Targeting cell cycle regulators in hematologic malignancies. Front Cell Dev Biol. 2015;3:16.
    1. Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, Anderson JE, Petersdorf SH. Age and acute myeloid leukemia. Blood. 2006;107:3481–3485.
    1. Juliusson G, Antunovic P, Derolf A, Lehmann S, Mollgard L, Stockelberg D, Tidefelt U, Wahlin A, Hoglund M. Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 2009;113:4179–4187.
    1. Leone G, Mele L, Pulsoni A, Equitani F, Pagano L. The incidence of secondary leukemias. Haematologica. 1999;84:937–945.
    1. Renella R, Verkooijen HM, Fioretta G, Vlastos G, Kurtz J, Sappino AP, Schafer P, Neyroud-Caspar I, Bouchardy C. Increased risk of acute myeloid leukemia after treatment for breast cancer. Breast. 2006;15:614–619.
    1. Appelbaum FR, Rosenblum D, Arceci RJ, Carroll WL, Breitfeld PP, Forman SJ, Larson RA, Lee SJ, Murphy SB, O'Brien S, Radich J, Scher NS, Smith FO, et al. End points to establish the efficacy of new agents in the treatment of acute leukemia. Blood. 2007;109:1810–1816.
    1. Lamson DW, Brignall MS. Antioxidants in cancer therapy; their actions and interactions with oncologic therapies. Altern Med Rev. 1999;4:304–329.
    1. Yang CS, Chung JY, Yang G, Chhabra SK, Lee MJ. Tea and tea polyphenols in cancer prevention. J Nutr. 2000;130(2S Suppl):472S–478S.
    1. Milkovic L, Siems W, Siems R, Zarkovic N. Oxidative stress and antioxidants in carcinogenesis and integrative therapy of cancer. Curr Pharm Des. 2014;20:6529–6542.
    1. Hollman PC, Feskens EJ, Katan MB. Tea flavonols in cardiovascular disease and cancer epidemiology. Proc Soc Exp Biol Med. 1999;220:198–202.
    1. Liu L, Hudgins WR, Shack S, Yin MQ, Samid D. Cinnamic acid: a natural product with potential use in cancer intervention. Int J Cancer. 1995;62:345–350.
    1. Gomez EV, Perez YM, Sanchez HV, Forment GR, Soler EA, Bertot LC, Garcia AY, del Rosario Abreu Vazquez M, Fabian LG. Antioxidant and immunomodulatory effects of Viusid in patients with chronic hepatitis C. World J Gastroenterol. 2010;16:2638–2647.
    1. Dayem Uddin MAI, Mahmood I, Ghosha AK, Khatuns RA. Findings of the 3-Month supportive treatment with ocoxin solution beside the standard modalities of patients with different neoplastic diseases. TAJ. 2009;22:172–175.
    1. Hernandez-Garcia S, Gonzalez V, Sanz E, Pandiella A. Effect of Oncoxin Oral Solution in HER2-Overexpressing Breast Cancer. Nutr Cancer. 2015;67:1159–1169.
    1. Ocana A, Pandiella A. Personalized therapies in the cancer “omics” era. Mol Cancer. 2010;9:202.
    1. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257.
    1. Horak ER, Leek R, Klenk N, LeJeune S, Smith K, Stuart N, Greenall M, Stepniewska K, Harris AL. Angiogenesis, assessed by platelet/endothelial cell adhesion molecule antibodies, as indicator of node metastases and survival in breast cancer. Lancet. 1992;340:1120–1124.
    1. Weidner N. Current pathologic methods for measuring intratumoral microvessel density within breast carcinoma and other solid tumors. Breast Cancer Res Treat. 1995;36:169–180.
    1. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999;13:1501–1512.
    1. Chu I, Blackwell K, Chen S, Slingerland J. The dual ErbB1/ErbB2 inhibitor, lapatinib (GW572016), cooperates with tamoxifen to inhibit both cell proliferation- and estrogen-dependent gene expression in antiestrogen-resistant breast cancer. Cancer Res. 2005;65:18–25.
    1. Nicola NA, Babon JJ. Leukemia inhibitory factor (LIF) Cytokine Growth Factor Rev. 2015;26:533–544.
    1. Giamarellou H, Antoniadou A. Infectious complications of febrile leukopenia. Infect Dis Clin North Am. 2001;15:457–482.
    1. Klastersky J, Paesmans M, Rubenstein EB, Boyer M, Elting L, Feld R, Gallagher J, Herrstedt J, Rapoport B, Rolston K, Talcott J. The Multinational Association for Supportive Care in Cancer risk index: A multinational scoring system for identifying low-risk febrile neutropenic cancer patients. J Clin Oncol. 2000;18:3038–3051.
    1. Szer J. The prevalent predicament of relapsed acute myeloid leukemia. Hematology Am Soc Hematol Educ Program. 2012;2012:43–48.
    1. Colado E, Alvarez-Fernandez S, Maiso P, Martin-Sanchez J, Vidriales MB, Garayoa M, Ocio EM, Montero JC, Pandiella A, San Miguel JF. The effect of the proteasome inhibitor bortezomib on acute myeloid leukemia cells and drug resistance associated with the CD34+ immature phenotype. Haematologica. 2008;93:57–66.
    1. Diaz-Rodriguez E, Alvarez-Fernandez S, Chen X, Paiva B, Lopez-Perez R, Garcia-Hernandez JL, San Miguel JF, Pandiella A. Deficient spindle assembly checkpoint in multiple myeloma. PLoS One. 2011;6:e27583.
    1. de Alava E, Ocana A, Abad M, Montero JC, Esparis-Ogando A, Rodriguez CA, Otero AP, Hernandez T, Cruz JJ, Pandiella A. Neuregulin expression modulates clinical response to trastuzumab in patients with metastatic breast cancer. J Clin Oncol. 2007;25:2656–2663.

Source: PubMed

3
Iratkozz fel