Skip to main content
Top
Published in: Metabolic Brain Disease 2/2014

01-06-2014 | Original Paper

Identifying the optimal dose of ritonavir in the treatment of malignancies

Author: Emad Y. Moawad

Published in: Metabolic Brain Disease | Issue 2/2014

Login to get access

Abstract

Identifying the optimal dose of ritonavir therapy overcomes the chemical resistance may exhibit in some cases due to poor prognosis of imprecise staging. Dose modeling was performed by analyzing previously published data of ritonavir cancer growth inhibition in vitro and in vivo. In-vitro 3H-Thymidine-based cell proliferation assay was performed on samples of the GL15 cell line incubated with 0, 1, 10 and 100 μ M of ritonavir. Proliferation inhibition was quantified to identify energy of the used doses as described before in earlier studies. Models involving in-vivo growth of established breast cancer tumor (MDA-MB-231), KSIMM tumor and EL4-T cell thymomas in mice were used. The effects of 40 mg/kg/day for 52 days, 30 mg/kg/day for 15 days and 8.8 mg/mouse/day for about 1 week of ritonavir in those xenograft growths respectively were monitored and quantified to identify energy of those doses as described before in earlier studies. Ritonavir demonstrated an in-vitro reduction in proliferation rate in dose dependent manner. The energy of the in-vitro influences following ritonavir therapy were perfectly correlated (r = 1) with ritonavir dose, allowed to establish an efficient energy-model with a perfect fit (R2=1) describes the energy yield by ritonavir doses, enables to administer the appropriate dose. Ritonavir had also a significant influence in-vivo on all sizes of treated tumors compared to the control animals such that the energy yield by the administered drug as derived from the energy-model was 100 % identical to the induced influence in tumor energy. The in-vitro determination of inhibition to proliferation by ritonavir doses is useful to characterize the response of cancer to ritonavir therapy targeting patient-personalized cancer medicine. The molecular method of response determination by 3H-TDR incorporation and ritonavir dose-energy model are reliable to avoid chemo-resistance by identifying the optimal dosing regimens and schedules prior therapy allowing the use of much lower dose of ritonavir and thus decreases the drug side effects and risks of relapse.
Literature
go back to reference Andre P, Groettrup M, Klenerman P et al (1998) An inhibitor of HIV-1 protease modulates proteasome activity, antigen presentation, and T cell responses. Proc Natl Acad Sci U S A 95:13120–13124PubMedCentralPubMedCrossRef Andre P, Groettrup M, Klenerman P et al (1998) An inhibitor of HIV-1 protease modulates proteasome activity, antigen presentation, and T cell responses. Proc Natl Acad Sci U S A 95:13120–13124PubMedCentralPubMedCrossRef
go back to reference Bocchini V, Casalone R, Collini P, Rebel G, Lo Curto F (1991) Changes in glial fibrillary acidic protein and karyotype during culturing of two cell lines established from human glioblastoma multiforme. Cell Tissue Res 265:73–81PubMedCrossRef Bocchini V, Casalone R, Collini P, Rebel G, Lo Curto F (1991) Changes in glial fibrillary acidic protein and karyotype during culturing of two cell lines established from human glioblastoma multiforme. Cell Tissue Res 265:73–81PubMedCrossRef
go back to reference Bower M, Fox P, Fife K et al (1999) Highly active anti-retroviral therapy (HAART) prolongs time to treatment failure in Kaposi’s sarcoma. AIDS 13:2105–2111PubMedCrossRef Bower M, Fox P, Fife K et al (1999) Highly active anti-retroviral therapy (HAART) prolongs time to treatment failure in Kaposi’s sarcoma. AIDS 13:2105–2111PubMedCrossRef
go back to reference Collier AC, Coombs RW, Schoenfeld DA et al (1996) Treatment of human immunodeficiency virus infection with saquinavir, zidovudine, and zalcitabine. AIDS Clinical Trials Group. N Engl J Med 334:1011–1017PubMedCrossRef Collier AC, Coombs RW, Schoenfeld DA et al (1996) Treatment of human immunodeficiency virus infection with saquinavir, zidovudine, and zalcitabine. AIDS Clinical Trials Group. N Engl J Med 334:1011–1017PubMedCrossRef
go back to reference Cornetta K, Moore A, Johannessohn M, Sledge GW (1994) Clonal dominance detected in metastases but not primary tumors of retrovirally marked human breast carcinoma injected into nude mice. Clin Exp Metastasis 12:3–12PubMedCrossRef Cornetta K, Moore A, Johannessohn M, Sledge GW (1994) Clonal dominance detected in metastases but not primary tumors of retrovirally marked human breast carcinoma injected into nude mice. Clin Exp Metastasis 12:3–12PubMedCrossRef
go back to reference Gaedicke S, Firat-Geier E, Constantiniu O et al (2002) Antitumor effect of the human immunodeficiency virus protease inhibitor ritonavir: induction of tumor-cell apoptosis associated with perturbation of proteasomal proteolysis. Cancer Res 62:6901–6908PubMed Gaedicke S, Firat-Geier E, Constantiniu O et al (2002) Antitumor effect of the human immunodeficiency virus protease inhibitor ritonavir: induction of tumor-cell apoptosis associated with perturbation of proteasomal proteolysis. Cancer Res 62:6901–6908PubMed
go back to reference Goldberg AL, Rock K (2002) Not just research tools—proteasome inhibitors offer therapeutic promise. Nat Med 8:338–340PubMedCrossRef Goldberg AL, Rock K (2002) Not just research tools—proteasome inhibitors offer therapeutic promise. Nat Med 8:338–340PubMedCrossRef
go back to reference Helbig G, Christopherson KW II, Bhat-Nakshatri P et al (2003) NF-κB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem 278:21631–21638PubMedCrossRef Helbig G, Christopherson KW II, Bhat-Nakshatri P et al (2003) NF-κB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem 278:21631–21638PubMedCrossRef
go back to reference Kyriazis AP, Kyriazis AA, Scarpelli DG, Fogh J, Rao MS, Lepera R (1982) Human pancreatic adenocarcinoma line Capan-1 in tissue culture and the nude mouse: morphologic, biologic, and biochemical characteristics. Am J Pathol 106:250–260PubMedCentralPubMed Kyriazis AP, Kyriazis AA, Scarpelli DG, Fogh J, Rao MS, Lepera R (1982) Human pancreatic adenocarcinoma line Capan-1 in tissue culture and the nude mouse: morphologic, biologic, and biochemical characteristics. Am J Pathol 106:250–260PubMedCentralPubMed
go back to reference Laurent N, de Bouard S, Guillamo JS et al (2004) Effects of the proteasome inhibitor ritonavir on glioma growth in vitro and in vivo. Mol Cancer Ther 3:129–136PubMed Laurent N, de Bouard S, Guillamo JS et al (2004) Effects of the proteasome inhibitor ritonavir on glioma growth in vitro and in vivo. Mol Cancer Ther 3:129–136PubMed
go back to reference Levitz SM (1998) Improvement in CD4_ cell counts despite persistently detectable HIV load. N Engl J Med 338:1074–1075PubMedCrossRef Levitz SM (1998) Improvement in CD4_ cell counts despite persistently detectable HIV load. N Engl J Med 338:1074–1075PubMedCrossRef
go back to reference Moawad E (2010) Isolated system towards a successful radiotherapy treatment. Nucl Med Mol Imaging 44:123–136CrossRef Moawad E (2010) Isolated system towards a successful radiotherapy treatment. Nucl Med Mol Imaging 44:123–136CrossRef
go back to reference Moawad EY (2011) Radiotherapy and risks of tumor regrowth or inducing second cancer. Cancer Nanotechnol 2:81–93CrossRef Moawad EY (2011) Radiotherapy and risks of tumor regrowth or inducing second cancer. Cancer Nanotechnol 2:81–93CrossRef
go back to reference Moawad EY (2012b) Clinical and pathological staging of the cancer at the nanoscale. Cancer Nano 3:37–46CrossRef Moawad EY (2012b) Clinical and pathological staging of the cancer at the nanoscale. Cancer Nano 3:37–46CrossRef
go back to reference Nguyen HQ, Magaret AS, Kitahata MM et al (2008) Persistent Kaposi sarcoma in the era of highly active antiretroviral therapy: characterizing the predictors of clinical response. AIDS 22:937–945PubMedCentralPubMedCrossRef Nguyen HQ, Magaret AS, Kitahata MM et al (2008) Persistent Kaposi sarcoma in the era of highly active antiretroviral therapy: characterizing the predictors of clinical response. AIDS 22:937–945PubMedCentralPubMedCrossRef
go back to reference Pajonk F, Himmelsbach J, Riess K et al (2002) The human immunodeficiency virus (HIV)-1 protease inhibitor saquinavir inhibits proteasome function and causes apoptosis and radiosensitization in non-HIV-associated human cancer cells. Cancer Res 62:5230–5235PubMed Pajonk F, Himmelsbach J, Riess K et al (2002) The human immunodeficiency virus (HIV)-1 protease inhibitor saquinavir inhibits proteasome function and causes apoptosis and radiosensitization in non-HIV-associated human cancer cells. Cancer Res 62:5230–5235PubMed
go back to reference Pati S, Pelser CB, Dufraine J, Bryant JL, Reitz MS Jr, Weichold FF (2002) Antitumorigenic effects of HIV protease inhibitor ritonavir: inhibition of Kaposi sarcoma. Blood 99(10):3771–3779PubMedCrossRef Pati S, Pelser CB, Dufraine J, Bryant JL, Reitz MS Jr, Weichold FF (2002) Antitumorigenic effects of HIV protease inhibitor ritonavir: inhibition of Kaposi sarcoma. Blood 99(10):3771–3779PubMedCrossRef
go back to reference Perrin L, Telenti A (1998) HIV treatment failure: testing for HIV resistance in clinical practice. Science 280:1871–1873PubMedCrossRef Perrin L, Telenti A (1998) HIV treatment failure: testing for HIV resistance in clinical practice. Science 280:1871–1873PubMedCrossRef
go back to reference Sgadari C et al (2003) Use of HIV protease inhibitors to block Kaposi’s sarcoma and tumour growth. Lancet Oncol 4(9):537–547PubMedCrossRef Sgadari C et al (2003) Use of HIV protease inhibitors to block Kaposi’s sarcoma and tumour growth. Lancet Oncol 4(9):537–547PubMedCrossRef
go back to reference Sloand EM, Kumar PN, Zella D, Young NS, Gallo RC, Weichold FF (1997) HIV-1 protease inhibitor modulates activation of peripheral blood derived CD4+ T-cells and decreases their susceptibility to apoptosis in vitro [abstract]. Blood 90:2568 Sloand EM, Kumar PN, Zella D, Young NS, Gallo RC, Weichold FF (1997) HIV-1 protease inhibitor modulates activation of peripheral blood derived CD4+ T-cells and decreases their susceptibility to apoptosis in vitro [abstract]. Blood 90:2568
go back to reference Sriranga MA, Milani M, Mitra R, Guo Z, Rodriguez M et al (2011) The human immunodeficiency virus protease inhibitor ritonavir inhibits lung cancer cells, in part, by inhibition of survivin. J Thorac Oncol 6:661–670CrossRef Sriranga MA, Milani M, Mitra R, Guo Z, Rodriguez M et al (2011) The human immunodeficiency virus protease inhibitor ritonavir inhibits lung cancer cells, in part, by inhibition of survivin. J Thorac Oncol 6:661–670CrossRef
go back to reference Srirangam A, Mitra R, Wang M et al (2006) Effects of HIV protease inhibitor ritonavir on Akt-regulated cell proliferation in breast cancer. Clin Cancer Res 12:1883–1896PubMedCentralPubMedCrossRef Srirangam A, Mitra R, Wang M et al (2006) Effects of HIV protease inhibitor ritonavir on Akt-regulated cell proliferation in breast cancer. Clin Cancer Res 12:1883–1896PubMedCentralPubMedCrossRef
go back to reference Torre-Cisneros J, Pozo F, Serrano R et al (2000) Patterns of lymphotropic herpesvirus viraemia in HIV-infected patients with Kaposi’s sarcoma treated with highly active antiretroviral therapy and liposomal daunorubicin. AIDS 14:2215–2217PubMedCrossRef Torre-Cisneros J, Pozo F, Serrano R et al (2000) Patterns of lymphotropic herpesvirus viraemia in HIV-infected patients with Kaposi’s sarcoma treated with highly active antiretroviral therapy and liposomal daunorubicin. AIDS 14:2215–2217PubMedCrossRef
go back to reference Weichold FF, Bryant JL, Pati S et al (1999) HIV-1 protease inhibitor ritonavir modulates susceptibility to apoptosis of uninfected T cells. J Hum Virol 2:261–269PubMed Weichold FF, Bryant JL, Pati S et al (1999) HIV-1 protease inhibitor ritonavir modulates susceptibility to apoptosis of uninfected T cells. J Hum Virol 2:261–269PubMed
Metadata
Title
Identifying the optimal dose of ritonavir in the treatment of malignancies
Author
Emad Y. Moawad
Publication date
01-06-2014
Publisher
Springer US
Published in
Metabolic Brain Disease / Issue 2/2014
Print ISSN: 0885-7490
Electronic ISSN: 1573-7365
DOI
https://doi.org/10.1007/s11011-013-9448-5

Other articles of this Issue 2/2014

Metabolic Brain Disease 2/2014 Go to the issue