Skip to main content
Top
Published in: Clinical Pharmacokinetics 2/2005

01-02-2005 | Review Article

Interactions Between Antiretrovirals and Antineoplastic Drug Therapy

Authors: Mr Tony Antoniou, Alice L. Tseng

Published in: Clinical Pharmacokinetics | Issue 2/2005

Login to get access

Abstract

Despite the established impact of highly active antiretroviral therapy (HAART) in reducing HIV-related morbidity and mortality, malignancy remains an important cause of death. Patients who receive the combination of cancer chemotherapy and HAART may achieve better response rates and higher rates of survival than patients who receive antineoplastic therapy alone. However, the likelihood of drug interactions with combined therapy is high, since protease inhibitors (PIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) are substrates and potent inhibitors or inducers of the cytochrome P450 (CYP) system. Since many antineoplastic drugs are also metabolised by the CYP system, coadministration with HAART could result in either drug accumulation and possible toxicity, or decreased efficacy of one or both classes of drugs. Although formal, prospective pharmacokinetic interaction studies are not available in most instances, it is possible to infer the nature of drug interactions based on the metabolic fates of these agents.
Paclitaxel and docetaxel are both metabolised by the CYP system, although differences exist in the nature of the isoenzymes involved. Case reports describing adverse consequences of concomitant taxane-antiretroviral therapy exist. Although other confounding factors may have been present, these cases serve as reminders of the vigilant monitoring necessary when taxanes and HAART are coadministered. Similarly, vinca alkaloids are substrates of CYP3A4 and are, thus, vulnerable to PI- or NNRTI-mediated changes in their pharmacokinetics. Interactions with the alkylating agents cyclophosphamide and ifosfamide are complicated as a result of the involvement of the CYP3A4 and CYP2B6 isoenzymes in both the metabolic activation of these drugs and the generation of potentially neurotoxic metabolites. Existing data regarding the metabolic fate of the anthracyclines doxorubicin and daunorubicin suggest that clinically detrimental interactions would not be expected with coadministered HAART. Commonly used endocrine therapies are largely substrates of the CYP system and may, therefore, be amenable to modulation by concomitant HAART. In addition, tamoxifen itself has been associated with reduced concentrations of both anastrozole and letrozole, raising the concern that similar inducing properties may adversely affect the outcome of PI- or NNRTI-based therapy. Similarly, dexamethasone is both a substrate and concentration-dependent inducer of CYP3A4; enhanced corticosteroid pharmacodynamics may result with CYP3A4 inhibitors, while the efficacy of concomitant HAART may be compromised with prolonged dexamethasone coadministration. Since PIs and NNRTIs may also induce or inhibit the expression of P-glycoprotein, the potential for additional interactions to arise via modulation of this transporter also exists. Further research delineating the combined safety and pharmacokinetics of antiretrovirals and antineoplastic therapy is necessary.
Literature
1.
go back to reference Palella FJJ, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338: 853–60PubMed Palella FJJ, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338: 853–60PubMed
2.
go back to reference Cameron DW, Heath-Chiozzi M, Danner S, et al. Randomised placebo-controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV Disease Ritonavir Study Group. Lancet 1998; 351: 543–9PubMed Cameron DW, Heath-Chiozzi M, Danner S, et al. Randomised placebo-controlled trial of ritonavir in advanced HIV-1 disease. The Advanced HIV Disease Ritonavir Study Group. Lancet 1998; 351: 543–9PubMed
3.
go back to reference Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. AIDS Clinical Trials Group 320 Study Team. N Engl J Med 1997; 337: 725–33PubMed Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. AIDS Clinical Trials Group 320 Study Team. N Engl J Med 1997; 337: 725–33PubMed
4.
go back to reference Van Cleef G, Fisher EJ, Polk RE. Drug interaction potential with inhibitors of HIV protease. Pharmacotherapy 1997; 17: 774–8PubMed Van Cleef G, Fisher EJ, Polk RE. Drug interaction potential with inhibitors of HIV protease. Pharmacotherapy 1997; 17: 774–8PubMed
5.
go back to reference Preston SL, Postelnick M, Purdy BD, et al. Drug interactions in HIV-positive patients initiated on protease inhibitor therapy [letter]. AIDS 1998; 12: 228–30PubMed Preston SL, Postelnick M, Purdy BD, et al. Drug interactions in HIV-positive patients initiated on protease inhibitor therapy [letter]. AIDS 1998; 12: 228–30PubMed
6.
go back to reference von Moltke LL, Greenblatt DY, Grassi JM, et al. Protease inhibitors as inhibitors of human cytochromes P450: high risk associated with ritonavir. J Clin Pharmacol 1998; 38: 106–11 von Moltke LL, Greenblatt DY, Grassi JM, et al. Protease inhibitors as inhibitors of human cytochromes P450: high risk associated with ritonavir. J Clin Pharmacol 1998; 38: 106–11
7.
go back to reference von Moltke LL, Greenblatt DJ, Granda BW, et al. Inhibition of cytochrome P450 isoforms by non-nucleoside reverse transcriptase inhibitors. J Clin Pharmacol 2001; 41: 85–91 von Moltke LL, Greenblatt DJ, Granda BW, et al. Inhibition of cytochrome P450 isoforms by non-nucleoside reverse transcriptase inhibitors. J Clin Pharmacol 2001; 41: 85–91
8.
go back to reference Deeks SG, Smith M, Holodniy M, et al. HIV-1 protease inhibitors: a review for clinicians. JAMA 1997; 277: 145–53PubMed Deeks SG, Smith M, Holodniy M, et al. HIV-1 protease inhibitors: a review for clinicians. JAMA 1997; 277: 145–53PubMed
9.
go back to reference Barry M, Gibbons S, Back D, et al. Protease inhibitors in patients with HIV disease: clinically important pharmacokinetic considerations. Clin Pharmacokinet 1997; 32: 194–209PubMed Barry M, Gibbons S, Back D, et al. Protease inhibitors in patients with HIV disease: clinically important pharmacokinetic considerations. Clin Pharmacokinet 1997; 32: 194–209PubMed
10.
go back to reference Piscitelli SC, Flexner C, Minor JR, et al. Drug interactions in patients infected with HIV. Clin Infect Dis 1996; 23: 685–93PubMed Piscitelli SC, Flexner C, Minor JR, et al. Drug interactions in patients infected with HIV. Clin Infect Dis 1996; 23: 685–93PubMed
11.
go back to reference Barry M, Mulcahy F, Merry C, et al. Pharmacokinetics and potential interactions amongst antiretroviral agents used to treat patients with HIV infection. Clin Pharmacokinet 1999; 36: 289–304PubMed Barry M, Mulcahy F, Merry C, et al. Pharmacokinetics and potential interactions amongst antiretroviral agents used to treat patients with HIV infection. Clin Pharmacokinet 1999; 36: 289–304PubMed
12.
go back to reference Tseng AL, Foisy MM. Management of drug interactions in patients with HIV. Ann Pharmacother 1997; 31: 1040–58PubMed Tseng AL, Foisy MM. Management of drug interactions in patients with HIV. Ann Pharmacother 1997; 31: 1040–58PubMed
13.
go back to reference Efavirenz (Sustiva®) product monograph. Princeton (NJ): Bristol Myers Squibb Pharma, 2002 Feb Efavirenz (Sustiva®) product monograph. Princeton (NJ): Bristol Myers Squibb Pharma, 2002 Feb
14.
go back to reference Orrick JJ, Steinhart CR. Atazanavir. Ann Pharmacother 2004; 38: 1664–74PubMed Orrick JJ, Steinhart CR. Atazanavir. Ann Pharmacother 2004; 38: 1664–74PubMed
15.
go back to reference Walmsley S, Leith J, Katlama C, et al. Pharmacokinetics and safety of tipranavir/ritonavir (TPV/r) alone or in combination with saquinavir (SQV), amprenavir (APV), or lopinavir (LPV): interim analysis of BI1182.51 [abstract no. WeOrB1236]. XV International AIDS Conference; 2004 Jul 11–16; Bangkok Walmsley S, Leith J, Katlama C, et al. Pharmacokinetics and safety of tipranavir/ritonavir (TPV/r) alone or in combination with saquinavir (SQV), amprenavir (APV), or lopinavir (LPV): interim analysis of BI1182.51 [abstract no. WeOrB1236]. XV International AIDS Conference; 2004 Jul 11–16; Bangkok
16.
go back to reference van Heeswijk R, Sabo JP, Cooper C, et al. The pharmacokinetic interactions between tipranavir/ritonavir 500/200mg bid (TPV/r) and atorvastatin, antacid and CYP3A4 in healthy adult volunteers [abstract no. 5.2]. 5th International Workshop on Clinical Pharmacology of HIV Therapy; 2004 Apr 1–3; Rome van Heeswijk R, Sabo JP, Cooper C, et al. The pharmacokinetic interactions between tipranavir/ritonavir 500/200mg bid (TPV/r) and atorvastatin, antacid and CYP3A4 in healthy adult volunteers [abstract no. 5.2]. 5th International Workshop on Clinical Pharmacology of HIV Therapy; 2004 Apr 1–3; Rome
17.
go back to reference Bonnet F, Morlat P, Chene G, et al. Causes of death among HIV-infected patients in the era of highly active antiretroviral therapy, Bordeaux, France, 1998–1999. HIV Med 2002; 3: 195–9PubMed Bonnet F, Morlat P, Chene G, et al. Causes of death among HIV-infected patients in the era of highly active antiretroviral therapy, Bordeaux, France, 1998–1999. HIV Med 2002; 3: 195–9PubMed
18.
go back to reference Cohen MH, French AL, Benning L, et al. Causes of death among women with human immunodeficiency virus infection in the era of combination antiretroviral therapy. Am J Med 2002; 113: 91–8PubMed Cohen MH, French AL, Benning L, et al. Causes of death among women with human immunodeficiency virus infection in the era of combination antiretroviral therapy. Am J Med 2002; 113: 91–8PubMed
19.
go back to reference Louie JK, Hsu LC, Osmond DH, et al. Trends in causes of death among persons with acquired immunodeficiency syndrome in the era of highly active antiretroviral therapy, San Francisco, 1994–1998. J Infect Dis 2002; 186: 1023–7PubMed Louie JK, Hsu LC, Osmond DH, et al. Trends in causes of death among persons with acquired immunodeficiency syndrome in the era of highly active antiretroviral therapy, San Francisco, 1994–1998. J Infect Dis 2002; 186: 1023–7PubMed
20.
go back to reference Jones JL, Hanson DL, Sworkin MS, et al. Incidence and trends in Kaposi’s sarcoma in the era of effective antiretroviral therapy. J Acquir Immune Defic Syndr 2000; 24: 270–4PubMed Jones JL, Hanson DL, Sworkin MS, et al. Incidence and trends in Kaposi’s sarcoma in the era of effective antiretroviral therapy. J Acquir Immune Defic Syndr 2000; 24: 270–4PubMed
21.
go back to reference Ledergerber B, Egger M, Erard V, et al. AIDS-related opportunistic illnesses occurring after initiation of potent antiretroviral therapy: the Swiss HIV Cohort Study. JAMA 1999; 282: 2220–6PubMed Ledergerber B, Egger M, Erard V, et al. AIDS-related opportunistic illnesses occurring after initiation of potent antiretroviral therapy: the Swiss HIV Cohort Study. JAMA 1999; 282: 2220–6PubMed
22.
go back to reference Carrieri MP, Pradier C, Piselli P, et al. Reduced incidence of Kaposi’s sarcoma and of systemic non-Hodgkin’s lymphoma in HIV-infected individuals treated with highly active antiretroviral therapy. Int J Cancer 2003; 103: 142–4PubMed Carrieri MP, Pradier C, Piselli P, et al. Reduced incidence of Kaposi’s sarcoma and of systemic non-Hodgkin’s lymphoma in HIV-infected individuals treated with highly active antiretroviral therapy. Int J Cancer 2003; 103: 142–4PubMed
23.
go back to reference Kirk O, Pederson C, Cozzi-Lepri A, et al. Non-Hodgkin lymphoma in HIV-infected patients in the era of highly active antiretroviral therapy. Blood 2001; 98: 3406–12PubMed Kirk O, Pederson C, Cozzi-Lepri A, et al. Non-Hodgkin lymphoma in HIV-infected patients in the era of highly active antiretroviral therapy. Blood 2001; 98: 3406–12PubMed
24.
go back to reference Dal Maso L, Franceschi S. Epidemiology of non-Hodgkin lymphomas and other haemolymphopoietic neoplasms in people with AIDS. Lancet Oncol 2003; 4: 110–9PubMed Dal Maso L, Franceschi S. Epidemiology of non-Hodgkin lymphomas and other haemolymphopoietic neoplasms in people with AIDS. Lancet Oncol 2003; 4: 110–9PubMed
25.
go back to reference Dal Maso L, Serraino D, Franceschi S. Epidemiology of AIDS-related tumours in developed and developing countries. Eur J Cancer 2001; 37: 1188–201PubMed Dal Maso L, Serraino D, Franceschi S. Epidemiology of AIDS-related tumours in developed and developing countries. Eur J Cancer 2001; 37: 1188–201PubMed
26.
go back to reference Bertz RJ, Granneman GR. Use of in vitro and in vivo data to estimate the likelihood of metabolic pharmacokinetic interactions. Clin Pharmacokinet 1997; 32: 210–58PubMed Bertz RJ, Granneman GR. Use of in vitro and in vivo data to estimate the likelihood of metabolic pharmacokinetic interactions. Clin Pharmacokinet 1997; 32: 210–58PubMed
27.
go back to reference Gerard L, Galicier L, Boulanger E, et al. Improved survival in HIV-related Hodgkin’s lymphoma since the introduction of highly active antiretroviral therapy. AIDS 2003; 17: 81–7PubMed Gerard L, Galicier L, Boulanger E, et al. Improved survival in HIV-related Hodgkin’s lymphoma since the introduction of highly active antiretroviral therapy. AIDS 2003; 17: 81–7PubMed
28.
go back to reference Tulpule A, Groopman J, Saville MW, et al. Multicenter trial of low-dose paclitaxel in patients with AIDS-related Kaposi sarcoma. Cancer 2002; 95: 147–54PubMed Tulpule A, Groopman J, Saville MW, et al. Multicenter trial of low-dose paclitaxel in patients with AIDS-related Kaposi sarcoma. Cancer 2002; 95: 147–54PubMed
29.
go back to reference Gill PS, Tulpule A, Espina BM, et al. Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi’s sarcoma. J Clin Oncol 1999; 17: 1876–83PubMed Gill PS, Tulpule A, Espina BM, et al. Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi’s sarcoma. J Clin Oncol 1999; 17: 1876–83PubMed
30.
go back to reference Welles L, Saville MW, Lietzau J, et al. Phase II trial with dose titration of paclitaxel for the therapy of human immunodeficiency virus-associated Kaposi’s sarcoma. J Clin Oncol 1998; 16: 1112–21PubMed Welles L, Saville MW, Lietzau J, et al. Phase II trial with dose titration of paclitaxel for the therapy of human immunodeficiency virus-associated Kaposi’s sarcoma. J Clin Oncol 1998; 16: 1112–21PubMed
31.
go back to reference Cresteil T, Monsarrat B, Dubois J, et al. Regioselective metabolism of taxoids by human CYP3A4 and 2C8: structure-activity relationship. Drug Metab Dispos 2002; 30: 438–45PubMed Cresteil T, Monsarrat B, Dubois J, et al. Regioselective metabolism of taxoids by human CYP3A4 and 2C8: structure-activity relationship. Drug Metab Dispos 2002; 30: 438–45PubMed
32.
go back to reference Rahman A, Korzekwa KR, Grogan J, et al. Selective biotransformation of taxol to 6 α-hydroxytaxol by human cytochrome P450 2C8. Cancer Res 1994; 54: 5543–6PubMed Rahman A, Korzekwa KR, Grogan J, et al. Selective biotransformation of taxol to 6 α-hydroxytaxol by human cytochrome P450 2C8. Cancer Res 1994; 54: 5543–6PubMed
33.
go back to reference Cresteil T, Monsarrat B, Alvinerie P, et al. Taxol metabolism by human liver microsomes: identification of cytochrome P450 involved in its biotransformation. Cancer Res 1994; 54: 386–92PubMed Cresteil T, Monsarrat B, Alvinerie P, et al. Taxol metabolism by human liver microsomes: identification of cytochrome P450 involved in its biotransformation. Cancer Res 1994; 54: 386–92PubMed
34.
go back to reference Monsarrat B, Royer I, Wright M, et al. Biotransformation of taxoids by human cytochromes P450: structure-activity relationship. Bull Cancer 1997; 84: 125–33PubMed Monsarrat B, Royer I, Wright M, et al. Biotransformation of taxoids by human cytochromes P450: structure-activity relationship. Bull Cancer 1997; 84: 125–33PubMed
35.
go back to reference Kumar G, Ray S, Walle T, et al. Comparative in vitro cytotoxic effects of taxol and its major human metabolite 6 α-hydroxytaxol. Cancer Chemother Pharmacol 1995; 36: 129–35PubMed Kumar G, Ray S, Walle T, et al. Comparative in vitro cytotoxic effects of taxol and its major human metabolite 6 α-hydroxytaxol. Cancer Chemother Pharmacol 1995; 36: 129–35PubMed
36.
go back to reference Monsarrat B, Mariel E, Cros S, et al. Taxol metabolism: isolation and identification of three major metabolites of taxol in rat bile. Drug Metab Dispos 1990; 18: 895–901PubMed Monsarrat B, Mariel E, Cros S, et al. Taxol metabolism: isolation and identification of three major metabolites of taxol in rat bile. Drug Metab Dispos 1990; 18: 895–901PubMed
37.
go back to reference Harris JW, Katki A, Anderson LW, et al. Isolation, structural determination, and biological activity of 6 α-hydroxytaxol, the principal human metabolite of taxol. J Med Chem 1994; 37: 706–9PubMed Harris JW, Katki A, Anderson LW, et al. Isolation, structural determination, and biological activity of 6 α-hydroxytaxol, the principal human metabolite of taxol. J Med Chem 1994; 37: 706–9PubMed
38.
go back to reference Panday VRN, Hoetelmans RMW, van Heeswijk RPG, et al. Paclitaxel in the treatment of human immunodeficiency virus 1-associated Kaposi’s sarcoma: drug-drug interactions with protease inhibitors and a nonnucleoside reverse transcriptase inhibitor: a case report study. Cancer Chemother Pharmacol 1999; 43: 516–9 Panday VRN, Hoetelmans RMW, van Heeswijk RPG, et al. Paclitaxel in the treatment of human immunodeficiency virus 1-associated Kaposi’s sarcoma: drug-drug interactions with protease inhibitors and a nonnucleoside reverse transcriptase inhibitor: a case report study. Cancer Chemother Pharmacol 1999; 43: 516–9
39.
go back to reference Duchin K, Sun J, Tan M, et al. Pharmacokinetics of low-dose Paxene™ (paclitaxel) in patients with refractory or relapsed AIDS-related Kaposi’s sarcoma [abstract no. 829]. 33rd Annual Meeting of the American Society of Clinical Oncology; 1997 May 17–20; Denver Duchin K, Sun J, Tan M, et al. Pharmacokinetics of low-dose Paxene™ (paclitaxel) in patients with refractory or relapsed AIDS-related Kaposi’s sarcoma [abstract no. 829]. 33rd Annual Meeting of the American Society of Clinical Oncology; 1997 May 17–20; Denver
40.
go back to reference Schwartz JD, Howard M, Scadden DT. Potential interaction of antiretroviral therapy with paclitaxel in patients with AIDS-related Kaposi’s sarcoma. AIDS 1999; 13: 283–4PubMed Schwartz JD, Howard M, Scadden DT. Potential interaction of antiretroviral therapy with paclitaxel in patients with AIDS-related Kaposi’s sarcoma. AIDS 1999; 13: 283–4PubMed
41.
go back to reference Parameswaran R, Sweeney C, Einhorn LH. Interaction between highly active antiretroviral therapy (HAART) and taxanes: a report of two cases [abstract no. 2194]. 38th Annual Meeting of the American Society of Clinical Oncology; 2002 May 18–21; Orlando Parameswaran R, Sweeney C, Einhorn LH. Interaction between highly active antiretroviral therapy (HAART) and taxanes: a report of two cases [abstract no. 2194]. 38th Annual Meeting of the American Society of Clinical Oncology; 2002 May 18–21; Orlando
42.
go back to reference Marre F, Sanderink GJ, de Sousa G, et al. Hepatic biotransformation of docetaxel (Taxotere) in vitro: involvement of the CYP3A subfamily in humans. Cancer Res 1996; 56: 1296–302PubMed Marre F, Sanderink GJ, de Sousa G, et al. Hepatic biotransformation of docetaxel (Taxotere) in vitro: involvement of the CYP3A subfamily in humans. Cancer Res 1996; 56: 1296–302PubMed
43.
go back to reference Hirth J, Watkins PB, Strawderman M, et al. The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin Cancer Res 2000; 6: 1255–8PubMed Hirth J, Watkins PB, Strawderman M, et al. The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin Cancer Res 2000; 6: 1255–8PubMed
44.
go back to reference Royer I, Monsarrat B, Sonnier M, et al. Metabolism of docetaxel by human cytochromes P450: interactions with paclitaxel and other antineoplastic drugs. Cancer Res 1996; 56: 56–65 Royer I, Monsarrat B, Sonnier M, et al. Metabolism of docetaxel by human cytochromes P450: interactions with paclitaxel and other antineoplastic drugs. Cancer Res 1996; 56: 56–65
45.
go back to reference Bardelmeijer HA, Ouwehand M, Buckle T, et al. Low systemic exposure of oral docetaxel in mice resulting from extensive first-pass metabolism is boosted by ritonavir. Cancer Res 2002; 62: 6158–84PubMed Bardelmeijer HA, Ouwehand M, Buckle T, et al. Low systemic exposure of oral docetaxel in mice resulting from extensive first-pass metabolism is boosted by ritonavir. Cancer Res 2002; 62: 6158–84PubMed
46.
go back to reference Villikka K, Kivisto KT, Maenpaa H, et al. Cytochrome P450-inducing antiepileptics increase the clearance of vincristine in patients with brain tumors. Clin Pharmacol Ther 1999; 66: 589–93PubMed Villikka K, Kivisto KT, Maenpaa H, et al. Cytochrome P450-inducing antiepileptics increase the clearance of vincristine in patients with brain tumors. Clin Pharmacol Ther 1999; 66: 589–93PubMed
47.
go back to reference Bohme A, Ganser A, Hoelzer D. Aggravation of vincristine-induced neurotoxicity by itraconazole in the treatment of adult ALL. Ann Hematol 1995; 71: 311–2PubMed Bohme A, Ganser A, Hoelzer D. Aggravation of vincristine-induced neurotoxicity by itraconazole in the treatment of adult ALL. Ann Hematol 1995; 71: 311–2PubMed
48.
go back to reference Kamaluddin M, McNally P, Breatnach F, et al. Potentiation of vincristine toxicity by itraconazole in children with lymphoid malignancies. Acta Paediatr 2001; 90: 1204–7PubMed Kamaluddin M, McNally P, Breatnach F, et al. Potentiation of vincristine toxicity by itraconazole in children with lymphoid malignancies. Acta Paediatr 2001; 90: 1204–7PubMed
49.
go back to reference Zhou-Pan XR, Seree E, Zhou XJ, et al. Involvement of human liver cytochrome P450 3in vinblastine metabolism: drug interactions. Cancer Res 1993; 53: 5121–6PubMed Zhou-Pan XR, Seree E, Zhou XJ, et al. Involvement of human liver cytochrome P450 3in vinblastine metabolism: drug interactions. Cancer Res 1993; 53: 5121–6PubMed
50.
go back to reference Kajita J, Kuwabara T, Kobayashi H, et al. CYP3A4 is mainly responsible for the metabolism of a new vinca alkaloid, vinorelbine, in human liver microsomes. Drug Metab Dispos 2000; 28: 1121–7PubMed Kajita J, Kuwabara T, Kobayashi H, et al. CYP3A4 is mainly responsible for the metabolism of a new vinca alkaloid, vinorelbine, in human liver microsomes. Drug Metab Dispos 2000; 28: 1121–7PubMed
51.
go back to reference Yao D, Ding S, Burchell B, et al. Detoxification of vinca alkaloids by human P450 CYP3A4-mediated metabolism: implications for the development of drug resistance. J Pharmacol Exp Ther 2000; 294: 387–95PubMed Yao D, Ding S, Burchell B, et al. Detoxification of vinca alkaloids by human P450 CYP3A4-mediated metabolism: implications for the development of drug resistance. J Pharmacol Exp Ther 2000; 294: 387–95PubMed
52.
go back to reference Relling MV, Evans R, Dass C, et al. Human cytochrome P450 metabolism of teniposide and etoposide. J Pharmacol Exp Ther 1992; 261: 491–6PubMed Relling MV, Evans R, Dass C, et al. Human cytochrome P450 metabolism of teniposide and etoposide. J Pharmacol Exp Ther 1992; 261: 491–6PubMed
53.
go back to reference Relling MV, Nemec J, Schuetz EG, et al. O-demethylation of epipodophyllotoxins is catalyzed by human cytochrome P450 3A4. Mol Pharmacol 1994; 45: 352–8PubMed Relling MV, Nemec J, Schuetz EG, et al. O-demethylation of epipodophyllotoxins is catalyzed by human cytochrome P450 3A4. Mol Pharmacol 1994; 45: 352–8PubMed
54.
go back to reference Kawashiro T, Yamashita K, Zhao XJ, et al. A study on the metabolism of etoposide and possible interactions with antitumor or supporting agents by human liver microsomes. J Pharmacol Exp Ther 1998; 286: 1294–300PubMed Kawashiro T, Yamashita K, Zhao XJ, et al. A study on the metabolism of etoposide and possible interactions with antitumor or supporting agents by human liver microsomes. J Pharmacol Exp Ther 1998; 286: 1294–300PubMed
55.
go back to reference Zhao XJ, Kawashiro T, Ishizaki T. Mutual inhibition between quinine and etoposide by human liver microsomes: evidence for cytochrome P450 3A4 involvement in their major metabolic pathways. Drug Metab Dispos 1997; 26: 188–91 Zhao XJ, Kawashiro T, Ishizaki T. Mutual inhibition between quinine and etoposide by human liver microsomes: evidence for cytochrome P450 3A4 involvement in their major metabolic pathways. Drug Metab Dispos 1997; 26: 188–91
56.
go back to reference Mathijssen RHJ, van Alpen RJ, Verweij J, et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res 2001; 7: 2182–94PubMed Mathijssen RHJ, van Alpen RJ, Verweij J, et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res 2001; 7: 2182–94PubMed
57.
go back to reference Cersosimo RJ. Topotecan: a new topoisomerase I inhibiting antineoplastic agent. Ann Pharmacother 1998; 32: 1334–43PubMed Cersosimo RJ. Topotecan: a new topoisomerase I inhibiting antineoplastic agent. Ann Pharmacother 1998; 32: 1334–43PubMed
58.
go back to reference Garcia-Carbonero R, Supko JG. Current perspectives on the clinical experience, pharmacology and continued development of the camptothecins. Clin Cancer Res 2002; 8: 641–61PubMed Garcia-Carbonero R, Supko JG. Current perspectives on the clinical experience, pharmacology and continued development of the camptothecins. Clin Cancer Res 2002; 8: 641–61PubMed
59.
go back to reference Herben VMM, Schoemaker NE, Rosing H, et al. Urinary and fecal excretion of topotecan in patients with malignant solid tumours. Cancer Chemother Pharmacol 2002; 50: 59–64PubMed Herben VMM, Schoemaker NE, Rosing H, et al. Urinary and fecal excretion of topotecan in patients with malignant solid tumours. Cancer Chemother Pharmacol 2002; 50: 59–64PubMed
60.
go back to reference Chang TK, Weber GF, Crespi CL, et al. Differential activation of cyclophosphamide and ifosphamide by cytochromes P450 2B and 3in human liver microsomes. Cancer Res 1993; 53: 5629–37PubMed Chang TK, Weber GF, Crespi CL, et al. Differential activation of cyclophosphamide and ifosphamide by cytochromes P450 2B and 3in human liver microsomes. Cancer Res 1993; 53: 5629–37PubMed
61.
go back to reference Roy P, Yu LJ, Crespi CL, et al. Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab Dispos 1999; 27: 655–66PubMed Roy P, Yu LJ, Crespi CL, et al. Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab Dispos 1999; 27: 655–66PubMed
62.
go back to reference Huang Z, Roy P, Waxman DJ. Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Biochem Pharmacol 2000; 59: 961–72PubMed Huang Z, Roy P, Waxman DJ. Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Biochem Pharmacol 2000; 59: 961–72PubMed
63.
go back to reference Yu L, Waxman DJ. Role of cytochrome P450 in oxazaphosphorine metabolism: deactivation via N-dechloroethylation and activation via 4-hydroxylation catalyzed by distinct subsets of rat liver cytochromes P450. Drug Metab Dispos 1996; 24: 1254–62PubMed Yu L, Waxman DJ. Role of cytochrome P450 in oxazaphosphorine metabolism: deactivation via N-dechloroethylation and activation via 4-hydroxylation catalyzed by distinct subsets of rat liver cytochromes P450. Drug Metab Dispos 1996; 24: 1254–62PubMed
64.
go back to reference Granvil CP, Madan A, Sharkawi M, et al. Role of CYP2B6 and CYP3A4 in the in vitro N-dechloroethylation of (R)- and (S)-ifosfamide in human liver microsomes. Drug Metab Dispos 1999; 27: 533–41PubMed Granvil CP, Madan A, Sharkawi M, et al. Role of CYP2B6 and CYP3A4 in the in vitro N-dechloroethylation of (R)- and (S)-ifosfamide in human liver microsomes. Drug Metab Dispos 1999; 27: 533–41PubMed
65.
go back to reference Roy P, Tretyakov O, Wright J, et al. Stereoselective metabolism of ifosfamide by human P-450s 3A4 and 2B6: favourable metabolic properties of (R)-enantiomer. Drug Metab Dispos 1999; 27: 1309–18PubMed Roy P, Tretyakov O, Wright J, et al. Stereoselective metabolism of ifosfamide by human P-450s 3A4 and 2B6: favourable metabolic properties of (R)-enantiomer. Drug Metab Dispos 1999; 27: 1309–18PubMed
66.
go back to reference Lee FY, Workman P, Roberts JT, et al. Clinical pharmacokinetics of oral CCNU (lomustine). Cancer Chemother Pharmacol 1985; 14: 125–31PubMed Lee FY, Workman P, Roberts JT, et al. Clinical pharmacokinetics of oral CCNU (lomustine). Cancer Chemother Pharmacol 1985; 14: 125–31PubMed
67.
go back to reference Wheeler GP, Johnston TP, Bowdon BJ, et al. Comparison of the properties of metabolites of CCNU. Biochem Pharmacol 1977; 26: 2331–6PubMed Wheeler GP, Johnston TP, Bowdon BJ, et al. Comparison of the properties of metabolites of CCNU. Biochem Pharmacol 1977; 26: 2331–6PubMed
68.
go back to reference Chang TK, Chen G, Waxman DJ. Modulation of thiotepa antitumor activity in vivo by alteration of liver cytochrome P450-catalyzed drug metabolism. J Pharmacol Exp Ther 1995; 274: 270–5PubMed Chang TK, Chen G, Waxman DJ. Modulation of thiotepa antitumor activity in vivo by alteration of liver cytochrome P450-catalyzed drug metabolism. J Pharmacol Exp Ther 1995; 274: 270–5PubMed
69.
go back to reference van Maanen MJ, Huitema AD, Beijen JH. Influence of comedicated drugs on the biotransformation of thioTEPA to TEPA and thioTEPA-mercapturate. Anticancer Res 2000; 20(3A): 1711–6PubMed van Maanen MJ, Huitema AD, Beijen JH. Influence of comedicated drugs on the biotransformation of thioTEPA to TEPA and thioTEPA-mercapturate. Anticancer Res 2000; 20(3A): 1711–6PubMed
70.
go back to reference Jacobson PA, Green K, Birnbaum A, et al. Cytochrome P450 isozymes 3A4 and 2B6 are involved in the in vitro human metabolism of thiotepa to TEPA. Cancer Chemother Pharmacol 2002; 49: 461–7PubMed Jacobson PA, Green K, Birnbaum A, et al. Cytochrome P450 isozymes 3A4 and 2B6 are involved in the in vitro human metabolism of thiotepa to TEPA. Cancer Chemother Pharmacol 2002; 49: 461–7PubMed
71.
go back to reference Rae JM, Soukhova NV, Flockhart DA, et al. Triethylenethiophosphoramide is a specific inhibitor of cytochrome P450 2B6: implications for cyclophosphamide metabolism. Drug Metab Dispos 2002; 30: 525–30PubMed Rae JM, Soukhova NV, Flockhart DA, et al. Triethylenethiophosphoramide is a specific inhibitor of cytochrome P450 2B6: implications for cyclophosphamide metabolism. Drug Metab Dispos 2002; 30: 525–30PubMed
72.
go back to reference Reid JM, Kuffel MJ, Miller JK, et al. Metabolic activation of dacarbazine by human cytochromes P450: the role of CYP1A1, CYP1A2, and CYP2E1. Clin Cancer Res 1999; 5: 2192–7PubMed Reid JM, Kuffel MJ, Miller JK, et al. Metabolic activation of dacarbazine by human cytochromes P450: the role of CYP1A1, CYP1A2, and CYP2E1. Clin Cancer Res 1999; 5: 2192–7PubMed
73.
go back to reference Damia G, D’Incalci M. Clinical pharmacokinetics of altretamine. Clin Pharmacokinet 1995; 28: 439–48PubMed Damia G, D’Incalci M. Clinical pharmacokinetics of altretamine. Clin Pharmacokinet 1995; 28: 439–48PubMed
74.
go back to reference Karunanayake EH, Hearse DJ, Mellows G. The metabolic fate and elimination of streptozocin. Biochem Soc Trans 1975; 3: 410–4PubMed Karunanayake EH, Hearse DJ, Mellows G. The metabolic fate and elimination of streptozocin. Biochem Soc Trans 1975; 3: 410–4PubMed
75.
go back to reference Weiss RB. Streptozocin: a review of its pharmacology, efficacy, and toxicity. Cancer Treat Rep 1982; 66: 427–38PubMed Weiss RB. Streptozocin: a review of its pharmacology, efficacy, and toxicity. Cancer Treat Rep 1982; 66: 427–38PubMed
76.
go back to reference Goria-Gatti L, Iannone A, Tomasi A, et al. In vitro and in vivo evidence for the formation of methyl radical from procarbazine: a spin trapping study. Carcinogenesis 1992; 13: 799–805PubMed Goria-Gatti L, Iannone A, Tomasi A, et al. In vitro and in vivo evidence for the formation of methyl radical from procarbazine: a spin trapping study. Carcinogenesis 1992; 13: 799–805PubMed
77.
go back to reference Kasel D, Baumhakel M, Fuhr U. Biodegradation of procarbazine by human liver microsomes. Int J Clin Pharmacol Ther 2000; 38: 153–5 Kasel D, Baumhakel M, Fuhr U. Biodegradation of procarbazine by human liver microsomes. Int J Clin Pharmacol Ther 2000; 38: 153–5
78.
go back to reference Czerwinski M, Gibbs JP, Slattery JT. Busulfan conjugation by glutathione S-transferases α, μ, and pi. Drug Metab Dispos 1996; 24: 1015–9PubMed Czerwinski M, Gibbs JP, Slattery JT. Busulfan conjugation by glutathione S-transferases α, μ, and pi. Drug Metab Dispos 1996; 24: 1015–9PubMed
79.
go back to reference Gibbs JP, Czerwinski M, Slattery JT. Busulfan-glutathione conjugation catalyzed by human liver cytosolic glutathione S-transferases. Cancer Res 1996; 56: 3678–81PubMed Gibbs JP, Czerwinski M, Slattery JT. Busulfan-glutathione conjugation catalyzed by human liver cytosolic glutathione S-transferases. Cancer Res 1996; 56: 3678–81PubMed
80.
go back to reference Poonkuzhali B, Chandy M, Srivastava A, et al. Glutathione S-transferase activity influences busulfan pharmacokinetics in patients with beta thalassemia major undergoing bone marrow transplantation. Drug Metab Dispos 2001; 29: 264–7PubMed Poonkuzhali B, Chandy M, Srivastava A, et al. Glutathione S-transferase activity influences busulfan pharmacokinetics in patients with beta thalassemia major undergoing bone marrow transplantation. Drug Metab Dispos 2001; 29: 264–7PubMed
81.
go back to reference Marchand DH, Remmel RP, Abdel-Monem MM. Biliary excretion of a glutathione conjugate of busulfan and 1, 4-diiodobutane in the rat. Drug Metab Dispos 1993; 16: 85–92 Marchand DH, Remmel RP, Abdel-Monem MM. Biliary excretion of a glutathione conjugate of busulfan and 1, 4-diiodobutane in the rat. Drug Metab Dispos 1993; 16: 85–92
82.
go back to reference Buggia I, Zecca M, Alessandrino EP, et al. Itraconazole can increase systemic exposure to busulfan in patients given bone marrow transplantation. GITMO (Gruppo Italiano Trapianto di Midollo Osseo). Anticancer Res 1996; 16: 2083–8PubMed Buggia I, Zecca M, Alessandrino EP, et al. Itraconazole can increase systemic exposure to busulfan in patients given bone marrow transplantation. GITMO (Gruppo Italiano Trapianto di Midollo Osseo). Anticancer Res 1996; 16: 2083–8PubMed
83.
go back to reference McLean A, Newell D, Baker G, et al. The metabolism of chlorambucil. Biochem Pharmacol 1980; 29: 2039–47PubMed McLean A, Newell D, Baker G, et al. The metabolism of chlorambucil. Biochem Pharmacol 1980; 29: 2039–47PubMed
84.
go back to reference Alberts DS, Chang SY, Chen HS, et al. Comparative pharmacokinetics of chlorambucil and melphalan in man. Recent Results Cancer Res 1980; 74: 124–31PubMed Alberts DS, Chang SY, Chen HS, et al. Comparative pharmacokinetics of chlorambucil and melphalan in man. Recent Results Cancer Res 1980; 74: 124–31PubMed
85.
go back to reference Alberts DS, Chang SY, Chen HS, et al. Kinetics of intravenous melphalan. Clin Pharmacol Ther 1979; 26: 73–80PubMed Alberts DS, Chang SY, Chen HS, et al. Kinetics of intravenous melphalan. Clin Pharmacol Ther 1979; 26: 73–80PubMed
86.
go back to reference Gormley PE, Bull JM, LeRoy AF, et al. Kinetics of cisdichlorodiammineplatinum. Clin Pharmacol Ther 1979; 25: 351–7PubMed Gormley PE, Bull JM, LeRoy AF, et al. Kinetics of cisdichlorodiammineplatinum. Clin Pharmacol Ther 1979; 25: 351–7PubMed
87.
go back to reference Jacobs C, Kaiman SM, Tretton M, et al. Renal handling of cisdiamminedichloroplatinum(II). Cancer Treat Rep 1980; 64: 1223–6PubMed Jacobs C, Kaiman SM, Tretton M, et al. Renal handling of cisdiamminedichloroplatinum(II). Cancer Treat Rep 1980; 64: 1223–6PubMed
88.
go back to reference LeRoy AF, Lutz RJ, Dedrick RL, et al. Pharmacokinetic study of cis-diamminedichloroplatinum(II) (DDP) in the beagle dog: thermodynamic and kinetic behaviour of DDP in a biologic milieu. Cancer Treat Rep 1979; 63: 59–71PubMed LeRoy AF, Lutz RJ, Dedrick RL, et al. Pharmacokinetic study of cis-diamminedichloroplatinum(II) (DDP) in the beagle dog: thermodynamic and kinetic behaviour of DDP in a biologic milieu. Cancer Treat Rep 1979; 63: 59–71PubMed
89.
go back to reference Go RS, Adjei AA. Review of the comparative pharmacology and clinical activity of cisplatin and carboplatin. J Clin Oncol 1999; 17: 409–22PubMed Go RS, Adjei AA. Review of the comparative pharmacology and clinical activity of cisplatin and carboplatin. J Clin Oncol 1999; 17: 409–22PubMed
90.
go back to reference Product monograph. Mustargen (mechlorethamine). West Point (PA): Merck & Co Inc., 1999 Mar Product monograph. Mustargen (mechlorethamine). West Point (PA): Merck & Co Inc., 1999 Mar
91.
go back to reference Bachur NR. Adriamycin (NSC-123127) pharmacology. Cancer Chemother Rep 1975; 6: 153–8 Bachur NR. Adriamycin (NSC-123127) pharmacology. Cancer Chemother Rep 1975; 6: 153–8
92.
go back to reference Takanashi S, Bachur NR. Adriamycin metabolism in man. Drug Metab Dispos 1967; 4: 79–87 Takanashi S, Bachur NR. Adriamycin metabolism in man. Drug Metab Dispos 1967; 4: 79–87
93.
go back to reference Benjamin RS, Riggs CE, Bachur NR. Plasma pharmacokinetics of adriamycin and its metabolites in humans with normal renal and hepatic function. Cancer Res 1977; 37: 1416–20PubMed Benjamin RS, Riggs CE, Bachur NR. Plasma pharmacokinetics of adriamycin and its metabolites in humans with normal renal and hepatic function. Cancer Res 1977; 37: 1416–20PubMed
94.
go back to reference Riggs CE, Benjamin RS, Serpick AA, et al. Biliary disposition of adriamycin. Clin Pharmacol Ther 1977; 22: 234–41PubMed Riggs CE, Benjamin RS, Serpick AA, et al. Biliary disposition of adriamycin. Clin Pharmacol Ther 1977; 22: 234–41PubMed
95.
go back to reference Goeptar AR, Te Koppele JM, Lamme EK, et al. Cytochrome P450 2B1-mediated one-electron reduction of adriamycin: a study with rat liver microsomes and purified enzymes. Mol Pharmacol 1993; 44: 1267–77PubMed Goeptar AR, Te Koppele JM, Lamme EK, et al. Cytochrome P450 2B1-mediated one-electron reduction of adriamycin: a study with rat liver microsomes and purified enzymes. Mol Pharmacol 1993; 44: 1267–77PubMed
96.
go back to reference Goeptar AR, Groot EF, Scheerens H, et al. Cytotoxicity of mitomycin and adriamycin in freshly isolated rat hepatocytes: the role of cytochrome P450. Cancer Res 1994; 54: 2411–8PubMed Goeptar AR, Groot EF, Scheerens H, et al. Cytotoxicity of mitomycin and adriamycin in freshly isolated rat hepatocytes: the role of cytochrome P450. Cancer Res 1994; 54: 2411–8PubMed
97.
go back to reference Coukell AJ, Spencer CM. Polyethylene glycol-liposomal doxorubicin: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in the management of AIDS-related Kaposi’s Sarcoma. Drugs 1997; 53: 520–38PubMed Coukell AJ, Spencer CM. Polyethylene glycol-liposomal doxorubicin: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in the management of AIDS-related Kaposi’s Sarcoma. Drugs 1997; 53: 520–38PubMed
98.
go back to reference Amantea MA, Forrest A, Northfelt DW, et al. Population pharmacokinetics and pharmacodynamics of pegylated-liposomal doxorubicin in patients with AIDS-related Kaposi’s sarcoma. Clin Pharmacol Ther 1997; 61: 301–11PubMed Amantea MA, Forrest A, Northfelt DW, et al. Population pharmacokinetics and pharmacodynamics of pegylated-liposomal doxorubicin in patients with AIDS-related Kaposi’s sarcoma. Clin Pharmacol Ther 1997; 61: 301–11PubMed
99.
go back to reference Loveless H, Arena E, Felsted RL, et al. Comparative mammalian metabolism of adriamycin and daunorubicin. Cancer Res 1978; 38: 593–8PubMed Loveless H, Arena E, Felsted RL, et al. Comparative mammalian metabolism of adriamycin and daunorubicin. Cancer Res 1978; 38: 593–8PubMed
100.
go back to reference Takanashi S, Bachur NR. Daunorubicin metabolites in human urine. J Pharmacol Exp Ther 1975; 195: 41–9PubMed Takanashi S, Bachur NR. Daunorubicin metabolites in human urine. J Pharmacol Exp Ther 1975; 195: 41–9PubMed
101.
go back to reference Lown JW, Chen HH. Electron paramagnetic resonance characterization and conformation of daunorubicin semiquinone intermediate implicated in anthracycline metabolism, cardiotoxicity, and anticancer action. Can J Chem 1981; 59: 3212–7 Lown JW, Chen HH. Electron paramagnetic resonance characterization and conformation of daunorubicin semiquinone intermediate implicated in anthracycline metabolism, cardiotoxicity, and anticancer action. Can J Chem 1981; 59: 3212–7
102.
go back to reference Fumagalli LLF, Zucchetti M, Vigano MG, et al. Treatment of HIV-associated non-Hodgkin’s lymphoma with liposomal daunorubicin substituted CHOP and HAART: preliminary clinical results and CSF pharmacokinetic [abstract no. 136]. American Society of Clinical Oncology, 36th Annual Meeting; 2000 May 19–23; New Orleans Fumagalli LLF, Zucchetti M, Vigano MG, et al. Treatment of HIV-associated non-Hodgkin’s lymphoma with liposomal daunorubicin substituted CHOP and HAART: preliminary clinical results and CSF pharmacokinetic [abstract no. 136]. American Society of Clinical Oncology, 36th Annual Meeting; 2000 May 19–23; New Orleans
103.
go back to reference Girard PM, Bouchaud O, Goetschel A, et al. Phase II study of liposomal encapsulated daunorubicin in the treatment of AIDS-associated mucocutaneous Kaposi’s sarcoma. AIDS 1996; 10: 753–7PubMed Girard PM, Bouchaud O, Goetschel A, et al. Phase II study of liposomal encapsulated daunorubicin in the treatment of AIDS-associated mucocutaneous Kaposi’s sarcoma. AIDS 1996; 10: 753–7PubMed
104.
go back to reference Gill PS, Wernz J, Scadden DT, et al. Randomized phase III trial of liposomal daunorubicin versus doxorubicin, bleomycin, and vincristine in AIDS-related Kaposi’s sarcoma. J Clin Oncol 1996; 14: 2353–64PubMed Gill PS, Wernz J, Scadden DT, et al. Randomized phase III trial of liposomal daunorubicin versus doxorubicin, bleomycin, and vincristine in AIDS-related Kaposi’s sarcoma. J Clin Oncol 1996; 14: 2353–64PubMed
105.
go back to reference Melosky B, Gelmon K. Phase II trial of liposomal daunorubicin with concurrent protease inhibitors in AIDS-related Kaposi’s sarcoma [abstract no. 34]. J Acquir Immune Defic Syndr 2000; 23: A23 Melosky B, Gelmon K. Phase II trial of liposomal daunorubicin with concurrent protease inhibitors in AIDS-related Kaposi’s sarcoma [abstract no. 34]. J Acquir Immune Defic Syndr 2000; 23: A23
106.
go back to reference Fumagalli L, Zucchetti M, Parisi I, et al. The pharmacokinetics of liposomal encapsulated daunorubicin are not modified by HAART in patients with HIV-associated Kaposi’s sarcoma. Cancer Chemother Pharmacol 2000; 45: 495–501PubMed Fumagalli L, Zucchetti M, Parisi I, et al. The pharmacokinetics of liposomal encapsulated daunorubicin are not modified by HAART in patients with HIV-associated Kaposi’s sarcoma. Cancer Chemother Pharmacol 2000; 45: 495–501PubMed
107.
go back to reference Tattersall MHN, Sodergren JE, Sengupta SK, et al. Pharmacokinetics of actinomycin D in patients with malignant melanoma. Clin Pharmacol Ther 1975; 17: 701–8PubMed Tattersall MHN, Sodergren JE, Sengupta SK, et al. Pharmacokinetics of actinomycin D in patients with malignant melanoma. Clin Pharmacol Ther 1975; 17: 701–8PubMed
108.
go back to reference Mross K, Maessen P, van der Vijgh WJ, et al. Pharmacokinetics and metabolism of epidoxorubicin and doxorubicin in humans. J Clin Oncol 1988; 6: 517–26PubMed Mross K, Maessen P, van der Vijgh WJ, et al. Pharmacokinetics and metabolism of epidoxorubicin and doxorubicin in humans. J Clin Oncol 1988; 6: 517–26PubMed
109.
go back to reference Weenen H, Lankelma J, Penders PG, et al. Pharmacokinetics of 4-epi-doxorubicin in man. Invest New Drugs 1983; 1: 59–64PubMed Weenen H, Lankelma J, Penders PG, et al. Pharmacokinetics of 4-epi-doxorubicin in man. Invest New Drugs 1983; 1: 59–64PubMed
110.
go back to reference Tjuljandin SA, Doig RG, Sobol MM, et al. Pharmacokinetics and toxicity of two schedules of high dose epirubicin. Cancer Res 1990; 50: 5095–101PubMed Tjuljandin SA, Doig RG, Sobol MM, et al. Pharmacokinetics and toxicity of two schedules of high dose epirubicin. Cancer Res 1990; 50: 5095–101PubMed
111.
go back to reference Innocenti F, Iyer L, Ramirez J, et al. Epirubicin glucuronidation is catalyzed by human UDP-glucuronosyltransferase 2B7. Drug Metab Dispos 2001; 29: 686–92PubMed Innocenti F, Iyer L, Ramirez J, et al. Epirubicin glucuronidation is catalyzed by human UDP-glucuronosyltransferase 2B7. Drug Metab Dispos 2001; 29: 686–92PubMed
112.
go back to reference Cairo MS, Toy C, Sender L, et al. Effect of idarubicin and epirubicin on in vitro polymorphonuclear function: diminished superoxide radical formation compared to their parent compounds daunorubicin and doxorubicin. J Leukoc Biol 1990; 47: 224–33PubMed Cairo MS, Toy C, Sender L, et al. Effect of idarubicin and epirubicin on in vitro polymorphonuclear function: diminished superoxide radical formation compared to their parent compounds daunorubicin and doxorubicin. J Leukoc Biol 1990; 47: 224–33PubMed
113.
go back to reference Gillies HC, Herriot D, Liang R, et al. Pharmacokinetics of idarubicin (4-demethoxydaunorubicin; IMI-30; NSC 256439) following intravenous and oral administration in patients with advanced cancer. Br J Clin Pharmacol 1987; 23: 303–10PubMed Gillies HC, Herriot D, Liang R, et al. Pharmacokinetics of idarubicin (4-demethoxydaunorubicin; IMI-30; NSC 256439) following intravenous and oral administration in patients with advanced cancer. Br J Clin Pharmacol 1987; 23: 303–10PubMed
114.
go back to reference Camaggi CM, Strocchi E, Carisi P, et al. Idarubicin metabolism and pharmacokinetics after intravenous and oral administration in cancer patients: a crossover study. Cancer Chemother Pharmacol 1992; 30: 307–16PubMed Camaggi CM, Strocchi E, Carisi P, et al. Idarubicin metabolism and pharmacokinetics after intravenous and oral administration in cancer patients: a crossover study. Cancer Chemother Pharmacol 1992; 30: 307–16PubMed
115.
go back to reference Kerpel-Fronius S, Verwey J, Stuurman M, et al. Pharmacokinetics and toxicity of mitomycin in rodents, given alone, in combination, or after induction of microsomal drug metabolism. Cancer Chemother Pharmacol 1988; 22: 104–8PubMed Kerpel-Fronius S, Verwey J, Stuurman M, et al. Pharmacokinetics and toxicity of mitomycin in rodents, given alone, in combination, or after induction of microsomal drug metabolism. Cancer Chemother Pharmacol 1988; 22: 104–8PubMed
116.
go back to reference Crooke ST, Henderson M, Samson M, et al. Phase I study of oral mitomycin Cancer Treat Rep 1976; 60: 1633–6 Crooke ST, Henderson M, Samson M, et al. Phase I study of oral mitomycin Cancer Treat Rep 1976; 60: 1633–6
117.
go back to reference den Hartigh J, McVie JG, van Oort WJ, et al. Pharmacokinetics of mitomycin in humans. Cancer Res 1983; 43: 5017–21 den Hartigh J, McVie JG, van Oort WJ, et al. Pharmacokinetics of mitomycin in humans. Cancer Res 1983; 43: 5017–21
118.
go back to reference Cummings J, Spanswick VJ, Tomasz M, et al. Enzymology of mitomycin metabolic activation in tumour tissue. Biochem Pharmacol 1998; 56: 405–14PubMed Cummings J, Spanswick VJ, Tomasz M, et al. Enzymology of mitomycin metabolic activation in tumour tissue. Biochem Pharmacol 1998; 56: 405–14PubMed
119.
go back to reference Chiccarelli FS, Morrison JA, Cosulich DB, et al. Identification of human urinary mitoxantrone metabolites. Cancer Res 1986; 46: 4858–61PubMed Chiccarelli FS, Morrison JA, Cosulich DB, et al. Identification of human urinary mitoxantrone metabolites. Cancer Res 1986; 46: 4858–61PubMed
120.
go back to reference Ehninger G, Proksch B, Heinzel G, et al. Clinical pharmacology of mitoxantrone. Cancer Treat Rep 1986; 70: 1373–8PubMed Ehninger G, Proksch B, Heinzel G, et al. Clinical pharmacology of mitoxantrone. Cancer Treat Rep 1986; 70: 1373–8PubMed
121.
go back to reference Blanz J, Mewes K, Ehninger G, et al. Evidence for oxidative activation of mitoxantrone in human, pig, and rat. Drug Metab Dispos 1991; 19: 871–80PubMed Blanz J, Mewes K, Ehninger G, et al. Evidence for oxidative activation of mitoxantrone in human, pig, and rat. Drug Metab Dispos 1991; 19: 871–80PubMed
122.
go back to reference Mewes K, Blanz J, Ehninger G, et al. Cytochrome P-450-induced cytotoxicity of mitoxantrone by formation of electrophilic intermediates. Cancer Res 1993; 53: 5135–42PubMed Mewes K, Blanz J, Ehninger G, et al. Cytochrome P-450-induced cytotoxicity of mitoxantrone by formation of electrophilic intermediates. Cancer Res 1993; 53: 5135–42PubMed
123.
go back to reference Duthie SJ, Grant MH. The role of reductive and oxidative metabolism in the toxicity of mitoxantrone, adriamycin and menadione in human liver derived HepG2 hepatoma cells. Br J Cancer 1989; 60: 566–71PubMed Duthie SJ, Grant MH. The role of reductive and oxidative metabolism in the toxicity of mitoxantrone, adriamycin and menadione in human liver derived HepG2 hepatoma cells. Br J Cancer 1989; 60: 566–71PubMed
124.
go back to reference Balis FM, Holcenberg JS, Bleyer WA. Clinical pharmacokinetics of commonly used anticancer drugs. Clin Pharmacokinet 1983; 8: 202–32PubMed Balis FM, Holcenberg JS, Bleyer WA. Clinical pharmacokinetics of commonly used anticancer drugs. Clin Pharmacokinet 1983; 8: 202–32PubMed
125.
go back to reference Montgomery MR, Furry J, Fernandez S. Bleomycin inhibits hepatic mixed function oxidation. Res Commun Chem Pathol Pharmacol 1981; 34: 287–93PubMed Montgomery MR, Furry J, Fernandez S. Bleomycin inhibits hepatic mixed function oxidation. Res Commun Chem Pathol Pharmacol 1981; 34: 287–93PubMed
126.
go back to reference Crewe HK, Ellis SW, Lennard MS, et al. Variable contribution of cytochromes P450 2D6, 2C9 and 3A4 to the 4-hydroxylation of tamoxifen by human liver microsomes. Biochem Pharmacol 1997; 53: 171–8PubMed Crewe HK, Ellis SW, Lennard MS, et al. Variable contribution of cytochromes P450 2D6, 2C9 and 3A4 to the 4-hydroxylation of tamoxifen by human liver microsomes. Biochem Pharmacol 1997; 53: 171–8PubMed
127.
go back to reference Jacolot F, Simon I, Dreano I, et al. Identification of the cytochrome P450 IIIA family as the enzymes involved in the N-demethylation of tamoxifen in human liver microsomes. Biochem Pharmacol 1991; 41: 1911–9PubMed Jacolot F, Simon I, Dreano I, et al. Identification of the cytochrome P450 IIIA family as the enzymes involved in the N-demethylation of tamoxifen in human liver microsomes. Biochem Pharmacol 1991; 41: 1911–9PubMed
128.
go back to reference Crewe HK, Notley LM, Wunsch RM, et al. Metabolism of tamoxifen by recombinant human cytochrome P450 enzymes: formation of the 4-hydroxy, 4′-hydroxy and N-desmethyl metabolites and isomerization of trans-4-hydroxytamoxifen. Drug Metab Dispos 2002; 30: 869–74PubMed Crewe HK, Notley LM, Wunsch RM, et al. Metabolism of tamoxifen by recombinant human cytochrome P450 enzymes: formation of the 4-hydroxy, 4′-hydroxy and N-desmethyl metabolites and isomerization of trans-4-hydroxytamoxifen. Drug Metab Dispos 2002; 30: 869–74PubMed
129.
go back to reference Fabian C, Tilzer L, Sternson L. Comparative binding affinities of tamoxifen, 4-hydroxytamoxifen, and desmethyltamoxifen for estrogen receptors isolated from human breast carcinoma: correlation with blood levels in patients with metastatic breast cancer. Biopharm Drug Dispos 1981; 2: 381–90PubMed Fabian C, Tilzer L, Sternson L. Comparative binding affinities of tamoxifen, 4-hydroxytamoxifen, and desmethyltamoxifen for estrogen receptors isolated from human breast carcinoma: correlation with blood levels in patients with metastatic breast cancer. Biopharm Drug Dispos 1981; 2: 381–90PubMed
130.
go back to reference Coezy E, Borgna JL, Rochefort H. Tamoxifen and metabolites in MCF7 cells: correlation between binding to estrogen receptor and inhibition of cell growth. Cancer Res 1982; 42: 317–23PubMed Coezy E, Borgna JL, Rochefort H. Tamoxifen and metabolites in MCF7 cells: correlation between binding to estrogen receptor and inhibition of cell growth. Cancer Res 1982; 42: 317–23PubMed
131.
go back to reference Mandlekar S, Hebbar V, Christov K, et al. Pharmacodynamics of tamoxifen and its 4-hydroxy and N-desmethyl metabolites: activation of caspases and induction of apoptosis in rat mammary tumors and in human breast cancer cell lines. Cancer Res 2000; 60: 6601–6PubMed Mandlekar S, Hebbar V, Christov K, et al. Pharmacodynamics of tamoxifen and its 4-hydroxy and N-desmethyl metabolites: activation of caspases and induction of apoptosis in rat mammary tumors and in human breast cancer cell lines. Cancer Res 2000; 60: 6601–6PubMed
132.
go back to reference Jordan VC, Collins MM, Rowsby L, et al. A monohydroxylated metabolite of tamoxifen with potent antiestrogenic activity. J Endocrinol 1977; 75: 305–16PubMed Jordan VC, Collins MM, Rowsby L, et al. A monohydroxylated metabolite of tamoxifen with potent antiestrogenic activity. J Endocrinol 1977; 75: 305–16PubMed
133.
go back to reference Boocock DJ, Brown K, Gibbs AH, et al. Identification of human CYP forms involved in the activation of tamoxifen and irreversible binding to DNA. Carcinogenesis 2002; 23: 1897–901PubMed Boocock DJ, Brown K, Gibbs AH, et al. Identification of human CYP forms involved in the activation of tamoxifen and irreversible binding to DNA. Carcinogenesis 2002; 23: 1897–901PubMed
134.
go back to reference Shibutani S, Ravindernath A, Suzuki N, et al. Identification of tamoxifen-DNA adducts in the endometrium of women treated with tamoxifen. Carcinogenesis 2000; 21: 1461–7PubMed Shibutani S, Ravindernath A, Suzuki N, et al. Identification of tamoxifen-DNA adducts in the endometrium of women treated with tamoxifen. Carcinogenesis 2000; 21: 1461–7PubMed
135.
go back to reference White IN, De Matteis F, Gibbs AH, et al. Species differences in the covalent binding of [14C]tamoxifen to liver microsomes and the forms of cytochrome P450 involved. Biochem Pharmacol 1995; 49: 1035–42PubMed White IN, De Matteis F, Gibbs AH, et al. Species differences in the covalent binding of [14C]tamoxifen to liver microsomes and the forms of cytochrome P450 involved. Biochem Pharmacol 1995; 49: 1035–42PubMed
136.
go back to reference Stiborova M, Borek-Dohalska L, Hodek P, et al. New selective inhibitors of cytochromes P450 2B and their application to antimutagenesis of tamoxifen. Arch Biochem Biophys 2002; 403: 41–9PubMed Stiborova M, Borek-Dohalska L, Hodek P, et al. New selective inhibitors of cytochromes P450 2B and their application to antimutagenesis of tamoxifen. Arch Biochem Biophys 2002; 403: 41–9PubMed
137.
go back to reference Kivisto KT, Villikka K, Nyman L, et al. Tamoxifen and toremifene concentrations in plasma are greatly decreased by rifampin. Clin Pharmacol Ther 1998; 64: 648–54PubMed Kivisto KT, Villikka K, Nyman L, et al. Tamoxifen and toremifene concentrations in plasma are greatly decreased by rifampin. Clin Pharmacol Ther 1998; 64: 648–54PubMed
138.
go back to reference Daniel PC, Gaskell J, Bishop H, et al. Determination of tamoxifen and biologically active metabolites in human breast tumour and plasma. Eur J Cancer Clin Oncol 1981; 17: 1183–9PubMed Daniel PC, Gaskell J, Bishop H, et al. Determination of tamoxifen and biologically active metabolites in human breast tumour and plasma. Eur J Cancer Clin Oncol 1981; 17: 1183–9PubMed
139.
go back to reference Desai PB, Nallani SC, Sane RS, et al. Induction of cytochrome P450 3A4 in primary human hepatocytes and activation of the human pregnane X receptor by tamoxifen and 4-hydroxytamoxifen. Drug Metab Dispos 2002; 30: 608–12PubMed Desai PB, Nallani SC, Sane RS, et al. Induction of cytochrome P450 3A4 in primary human hepatocytes and activation of the human pregnane X receptor by tamoxifen and 4-hydroxytamoxifen. Drug Metab Dispos 2002; 30: 608–12PubMed
140.
go back to reference Cotreau MM, von Moltke LL, Harmatz JS, et al. Molecular and pharmacokinetic evaluation of rat hepatic and gastrointestinal cytochrome p450 induction by tamoxifen. Pharmacology 2001; 63: 210–9PubMed Cotreau MM, von Moltke LL, Harmatz JS, et al. Molecular and pharmacokinetic evaluation of rat hepatic and gastrointestinal cytochrome p450 induction by tamoxifen. Pharmacology 2001; 63: 210–9PubMed
141.
go back to reference Buzdar AU, Robertson JFR, Eirmann W, et al. An overview of the pharmacology and pharmacokinetics of the newer generation aromatase inhibitors anastrozole, letrozole and exemestane. Cancer 2002; 95: 2006–16PubMed Buzdar AU, Robertson JFR, Eirmann W, et al. An overview of the pharmacology and pharmacokinetics of the newer generation aromatase inhibitors anastrozole, letrozole and exemestane. Cancer 2002; 95: 2006–16PubMed
142.
go back to reference Lamb HM, Adkins JC. Letrozole: a review of its use in postmenopausal women with advanced breast cancer. Drugs 1998; 56: 1125–40PubMed Lamb HM, Adkins JC. Letrozole: a review of its use in postmenopausal women with advanced breast cancer. Drugs 1998; 56: 1125–40PubMed
143.
go back to reference ATAC Trialists’ Group. Pharmacokinetics of anastrozole and tamoxifen alone, and in combination, during adjuvant endocrine therapy for early breast cancer in postmenopausal women: a sub-protocol of the ‘Arimidex™ and tamoxifen alone or in combination’ (ATAC) trial. Br J Cancer 2001; 85: 317–24 ATAC Trialists’ Group. Pharmacokinetics of anastrozole and tamoxifen alone, and in combination, during adjuvant endocrine therapy for early breast cancer in postmenopausal women: a sub-protocol of the ‘Arimidex™ and tamoxifen alone or in combination’ (ATAC) trial. Br J Cancer 2001; 85: 317–24
144.
go back to reference Berthou F, Dreano Y, Belloc C, et al. Involvement of cytochrome P450 3enzyme family in the major metabolic pathways of toremifene in human liver microsomes. Biochem Pharmacol 1994; 47: 1883–95PubMed Berthou F, Dreano Y, Belloc C, et al. Involvement of cytochrome P450 3enzyme family in the major metabolic pathways of toremifene in human liver microsomes. Biochem Pharmacol 1994; 47: 1883–95PubMed
145.
go back to reference Grill HJ, Pollow K. Pharmacokinetics of droloxifene and its metabolites in breast cancer patients. Am J Clin Oncol 1991; 14 Suppl. 2: S21–9PubMed Grill HJ, Pollow K. Pharmacokinetics of droloxifene and its metabolites in breast cancer patients. Am J Clin Oncol 1991; 14 Suppl. 2: S21–9PubMed
146.
go back to reference Bruning PF. Droloxifene, a new anti-oestrogen in postmenopausal advanced breast cancer: preliminary results of a double-blind dose-finding phase II trial. Eur J Cancer 1992; 28A: 1404–7PubMed Bruning PF. Droloxifene, a new anti-oestrogen in postmenopausal advanced breast cancer: preliminary results of a double-blind dose-finding phase II trial. Eur J Cancer 1992; 28A: 1404–7PubMed
147.
go back to reference John BA, Brodie RR, Baldock GA, et al. Pharmacokinetics and metabolism of the anti-oestrogen droloxifene in female human subjects. Xenobiotica 2002; 32: 699–713PubMed John BA, Brodie RR, Baldock GA, et al. Pharmacokinetics and metabolism of the anti-oestrogen droloxifene in female human subjects. Xenobiotica 2002; 32: 699–713PubMed
148.
go back to reference Lonning PE, Geisler J, Johannessen DC, et al. Pharmacokinetics and metabolism of formestane in breast cancer patients. J Steroid Biochem Mol Biol 2001; 77: 39–47PubMed Lonning PE, Geisler J, Johannessen DC, et al. Pharmacokinetics and metabolism of formestane in breast cancer patients. J Steroid Biochem Mol Biol 2001; 77: 39–47PubMed
149.
go back to reference Gentile DM, Tomlinson ES, Maggs JL, et al. Dexamethasone metabolism by human liver in vitro. Metabolite identification and inhibition of 6-hydroxylation. J Pharmacol Exp Ther 1996; 277: 105–12PubMed Gentile DM, Tomlinson ES, Maggs JL, et al. Dexamethasone metabolism by human liver in vitro. Metabolite identification and inhibition of 6-hydroxylation. J Pharmacol Exp Ther 1996; 277: 105–12PubMed
150.
go back to reference Tomlinson ES, Lewis DFV, Maggs JL, et al. In vitro metabolism of dexamethasone (DEX) in human liver and kidney: the involvement of CYP3A4 and CYP17 (17,20 LYASE) and molecular modelling studies. Biochem Pharmacol 1997; 54: 605–11PubMed Tomlinson ES, Lewis DFV, Maggs JL, et al. In vitro metabolism of dexamethasone (DEX) in human liver and kidney: the involvement of CYP3A4 and CYP17 (17,20 LYASE) and molecular modelling studies. Biochem Pharmacol 1997; 54: 605–11PubMed
151.
go back to reference Varis T, Kivisto KT, Backman JT, et al. Itraconazole decreases the clearance and enhances the effects of intravenously administered methylprednisolone in healthy volunteers. Pharmacol Toxicol 1999; 85: 29–32PubMed Varis T, Kivisto KT, Backman JT, et al. Itraconazole decreases the clearance and enhances the effects of intravenously administered methylprednisolone in healthy volunteers. Pharmacol Toxicol 1999; 85: 29–32PubMed
152.
go back to reference Varis T, Kaukonen KM, Kivisto KT, et al. Plasma concentrations and effects of oral methylprednisolone are considerably increased by itraconazole. Clin Pharmacol Ther 1998; 64: 363–8PubMed Varis T, Kaukonen KM, Kivisto KT, et al. Plasma concentrations and effects of oral methylprednisolone are considerably increased by itraconazole. Clin Pharmacol Ther 1998; 64: 363–8PubMed
153.
go back to reference Lebrun-Vignes B, Archer VC, Diquet B, et al. Effect of itraconazole on the pharmacokinetics of prednisolone and methylprednisolone and Cortisol secretion in healthy subjects. Br J Clin Pharmacol 2001; 51: 443–50PubMed Lebrun-Vignes B, Archer VC, Diquet B, et al. Effect of itraconazole on the pharmacokinetics of prednisolone and methylprednisolone and Cortisol secretion in healthy subjects. Br J Clin Pharmacol 2001; 51: 443–50PubMed
154.
go back to reference Varis T, Kivisto KT, Neuvonen PJ. Grapefruit juice can increase the plasma concentrations of oral methylprednisolone. Eur J Clin Pharmacol 2000; 56: 489–93PubMed Varis T, Kivisto KT, Neuvonen PJ. Grapefruit juice can increase the plasma concentrations of oral methylprednisolone. Eur J Clin Pharmacol 2000; 56: 489–93PubMed
155.
go back to reference Varis T, Kivisto KT, Backman JT, et al. The cytochrome P450 3A4 inhibitor itraconazole markedly increases the plasma concentrations of dexamethasone and enhances its adrenal-suppressant effect. Clin Pharmacol Ther 2000; 68: 487–94PubMed Varis T, Kivisto KT, Backman JT, et al. The cytochrome P450 3A4 inhibitor itraconazole markedly increases the plasma concentrations of dexamethasone and enhances its adrenal-suppressant effect. Clin Pharmacol Ther 2000; 68: 487–94PubMed
156.
go back to reference Ogg MS, Williams JM, Tarbit M, et al. A receptor gene assay to assess the molecular mechanisms of xenobiotic-dependent induction of the human CYP3A4 gene in vitro. Xenobiotica 1999; 29: 269–79PubMed Ogg MS, Williams JM, Tarbit M, et al. A receptor gene assay to assess the molecular mechanisms of xenobiotic-dependent induction of the human CYP3A4 gene in vitro. Xenobiotica 1999; 29: 269–79PubMed
157.
go back to reference McCune JS, Hawke RL, LeCluyse EL, et al. In vivo and in vitro induction of human cytochrome P450 3A4 by dexamethasone. Clin Pharmacol Ther 2000; 68: 356–66PubMed McCune JS, Hawke RL, LeCluyse EL, et al. In vivo and in vitro induction of human cytochrome P450 3A4 by dexamethasone. Clin Pharmacol Ther 2000; 68: 356–66PubMed
158.
go back to reference Yamashita SK, Ludwig EA, Middleton Jr E, et al. Lack of pharmacokinetic and pharmacodynamic interactions between ketoconazole and prednisolone. Clin Pharmacol Ther 1991; 49: 558–70PubMed Yamashita SK, Ludwig EA, Middleton Jr E, et al. Lack of pharmacokinetic and pharmacodynamic interactions between ketoconazole and prednisolone. Clin Pharmacol Ther 1991; 49: 558–70PubMed
159.
go back to reference Hollander AA, van Rooij J, Lentjes GW, et al. The effect of grapefruit juice on cyclosporine and prednisone metabolism in transplant patients. Clin Pharmacol Ther 1995; 57: 318–24PubMed Hollander AA, van Rooij J, Lentjes GW, et al. The effect of grapefruit juice on cyclosporine and prednisone metabolism in transplant patients. Clin Pharmacol Ther 1995; 57: 318–24PubMed
160.
go back to reference Varis T, Kivisto KT, Neuvonen PJ. The effect of itraconazole on the pharmacokinetics and pharmacodynamics of oral prednisolone. Eur J Clin Pharmacol 2000; 56: 57–60PubMed Varis T, Kivisto KT, Neuvonen PJ. The effect of itraconazole on the pharmacokinetics and pharmacodynamics of oral prednisolone. Eur J Clin Pharmacol 2000; 56: 57–60PubMed
161.
go back to reference Harris RZ, Tsunoda SM, Mroczkowski P, et al. The effects of menopause and hormone replacement therapies on prednisolone and erythromycin pharmacokinetics. Clin Pharmacol Ther 1996; 59: 429–35PubMed Harris RZ, Tsunoda SM, Mroczkowski P, et al. The effects of menopause and hormone replacement therapies on prednisolone and erythromycin pharmacokinetics. Clin Pharmacol Ther 1996; 59: 429–35PubMed
162.
go back to reference Bundow D, Aboulafia DM. Potential drug interaction with paclitaxel and highly active antiretroviral therapy in two patients with AIDS-associated Kaposi Sarcoma. Am J Clin Oncol 2004; 27: 81–4PubMed Bundow D, Aboulafia DM. Potential drug interaction with paclitaxel and highly active antiretroviral therapy in two patients with AIDS-associated Kaposi Sarcoma. Am J Clin Oncol 2004; 27: 81–4PubMed
163.
go back to reference Wen X, Wang JS, Backman JT, et al. Trimethoprim and sulfamethoxazole are selective inhibitors of CYP2C8 and CYP2C9, respectively. Drug Metab Dispos 2002; 30: 631–5PubMed Wen X, Wang JS, Backman JT, et al. Trimethoprim and sulfamethoxazole are selective inhibitors of CYP2C8 and CYP2C9, respectively. Drug Metab Dispos 2002; 30: 631–5PubMed
164.
go back to reference Klecker RW, Jamis-Dow CA, Egorin MJ, et al. Effect of Cimetidine, probenecid, and ketoconazole on the distribution, biliary secretion, and metabolism of [3H]taxol in the Sprague-Dawley rat. Drug Metab Dispos 1994; 22: 254–8PubMed Klecker RW, Jamis-Dow CA, Egorin MJ, et al. Effect of Cimetidine, probenecid, and ketoconazole on the distribution, biliary secretion, and metabolism of [3H]taxol in the Sprague-Dawley rat. Drug Metab Dispos 1994; 22: 254–8PubMed
165.
go back to reference Jamis-Dow CA, Klecker RW, Katki AG, et al. Metabolism of taxol by human and rat liver in vitro: a screen for drug interactions and interspecies differences. Cancer Chemother Pharmacol 1995; 36: 107–14PubMed Jamis-Dow CA, Klecker RW, Katki AG, et al. Metabolism of taxol by human and rat liver in vitro: a screen for drug interactions and interspecies differences. Cancer Chemother Pharmacol 1995; 36: 107–14PubMed
166.
go back to reference Slichenmeyer WJ, Donehower RC, Chen TL, et al. Pretreatment H2 receptor antagonists that differ in P450 modulation activity: comparative effects on paclitaxel clearance rates and neutropenia. Cancer Chemother Pharmacol 1995; 36: 227–32 Slichenmeyer WJ, Donehower RC, Chen TL, et al. Pretreatment H2 receptor antagonists that differ in P450 modulation activity: comparative effects on paclitaxel clearance rates and neutropenia. Cancer Chemother Pharmacol 1995; 36: 227–32
167.
go back to reference Jamis-Dow CA, Pearl ML, Watkins PB, et al. Predicting drug interactions in vivo from experiments in vitro: human studies with paclitaxel and ketoconazole. Am J Clin Oncol 1997; 20: 592–9PubMed Jamis-Dow CA, Pearl ML, Watkins PB, et al. Predicting drug interactions in vivo from experiments in vitro: human studies with paclitaxel and ketoconazole. Am J Clin Oncol 1997; 20: 592–9PubMed
168.
go back to reference Sonnichsen DS, Liu Q, Schuetz EG, et al. Variability in human cytochrome P450 paclitaxel metabolism. J Pharmacol Exp Ther 1995; 275: 566–75PubMed Sonnichsen DS, Liu Q, Schuetz EG, et al. Variability in human cytochrome P450 paclitaxel metabolism. J Pharmacol Exp Ther 1995; 275: 566–75PubMed
169.
go back to reference Dai D, Zeldin DC, Blaisdell JA, et al. Polymorphisms in human CYP2C8 decrease metabolism of the anticancer drug paclitaxel and arachidonic acid. Pharmacogenetics 2001; 11: 597–607PubMed Dai D, Zeldin DC, Blaisdell JA, et al. Polymorphisms in human CYP2C8 decrease metabolism of the anticancer drug paclitaxel and arachidonic acid. Pharmacogenetics 2001; 11: 597–607PubMed
170.
go back to reference Bahadur N, Leathart JBS, Mutch E, et al. CYP2C8 polymorphisms in Caucasians and their relationship with paclitaxel 6α-hydroxylase activity in human liver microsomes. Biochem Pharmacol 2002; 64: 1579–89PubMed Bahadur N, Leathart JBS, Mutch E, et al. CYP2C8 polymorphisms in Caucasians and their relationship with paclitaxel 6α-hydroxylase activity in human liver microsomes. Biochem Pharmacol 2002; 64: 1579–89PubMed
171.
go back to reference Kearns CM, Gianni L, Egorin MJ. Paclitaxel pharmacokinetics and pharmacodynamics. Semin Oncol 1995; 22 Suppl. 6: 16–23PubMed Kearns CM, Gianni L, Egorin MJ. Paclitaxel pharmacokinetics and pharmacodynamics. Semin Oncol 1995; 22 Suppl. 6: 16–23PubMed
172.
go back to reference Engels FK, Ten Tije AJ, Baker SD, et al. Effect of cytochrome P450 3A4 inhibition on the pharamcokinetics of docetaxel. Clin Pharmacol Ther 2004; 75: 448–54PubMed Engels FK, Ten Tije AJ, Baker SD, et al. Effect of cytochrome P450 3A4 inhibition on the pharamcokinetics of docetaxel. Clin Pharmacol Ther 2004; 75: 448–54PubMed
173.
go back to reference Bruno R, Hille D, Riva A, et al. Population pharmacokinetics/pharmacodynamics of docetaxel in phase II studies in patients with cancers. J Clin Oncol 1998; 16: 187–96PubMed Bruno R, Hille D, Riva A, et al. Population pharmacokinetics/pharmacodynamics of docetaxel in phase II studies in patients with cancers. J Clin Oncol 1998; 16: 187–96PubMed
175.
go back to reference Chang SM, Kuhn JG, Rizzo J, et al. Phase I study of paclitaxel in patients with recurrent malignant glioma: a North American Brain Tumor Consortium report. J Clin Oncol 1998; 16: 2188–94PubMed Chang SM, Kuhn JG, Rizzo J, et al. Phase I study of paclitaxel in patients with recurrent malignant glioma: a North American Brain Tumor Consortium report. J Clin Oncol 1998; 16: 2188–94PubMed
176.
go back to reference Kostrubsky VE, Lewis LD, Wood SG, et al. Effect of taxol on cytochrome P450 3and acetaminophen toxicity in cultured rat hepatocytes: comparison to dexamethasone. Toxicol Appl Pharmacol 1997; 142: 79–86PubMed Kostrubsky VE, Lewis LD, Wood SG, et al. Effect of taxol on cytochrome P450 3and acetaminophen toxicity in cultured rat hepatocytes: comparison to dexamethasone. Toxicol Appl Pharmacol 1997; 142: 79–86PubMed
177.
go back to reference Kostrubsky VE, Lewis LD, Strom SC, et al. Induction of cytochrome P450 3by Taxol in primary cultures of human hepatocytes. Arch Biochem Biophys 1998; 355: 131–6PubMed Kostrubsky VE, Lewis LD, Strom SC, et al. Induction of cytochrome P450 3by Taxol in primary cultures of human hepatocytes. Arch Biochem Biophys 1998; 355: 131–6PubMed
178.
go back to reference Nallani SC, Genter MB, Desai PB. Increased activity of CYP3A enzyme in primary cultures of rat hepatocytes treated with docetaxel: comparative evaluation with paclitaxel. Cancer Chemother Pharmacol 2001; 48: 115–22PubMed Nallani SC, Genter MB, Desai PB. Increased activity of CYP3A enzyme in primary cultures of rat hepatocytes treated with docetaxel: comparative evaluation with paclitaxel. Cancer Chemother Pharmacol 2001; 48: 115–22PubMed
179.
go back to reference Nallani SC, Goodwin B, Buckley AR, et al. Differences in the induction of cytochrome P450 3A4 by taxane anticancer drugs, docetaxel and paclitaxel, assessed employing primary human hepatocytes. Cancer Chemother Pharmacol 2004; 54: 219–29PubMed Nallani SC, Goodwin B, Buckley AR, et al. Differences in the induction of cytochrome P450 3A4 by taxane anticancer drugs, docetaxel and paclitaxel, assessed employing primary human hepatocytes. Cancer Chemother Pharmacol 2004; 54: 219–29PubMed
180.
go back to reference Baumhakel M, Kasel D, Rao-Schymanski RA, et al. Screening for inhibitory effects of antineoplastic agents on CYP3A4 in human liver microsomes. Int J Clin Pharmacol Ther 2001; 39: 517–28PubMed Baumhakel M, Kasel D, Rao-Schymanski RA, et al. Screening for inhibitory effects of antineoplastic agents on CYP3A4 in human liver microsomes. Int J Clin Pharmacol Ther 2001; 39: 517–28PubMed
181.
go back to reference Nasti G, Errante D, Talamini R, et al. Vinorelbine is an effective and safe drug for AIDS-related Kaposi’s sarcoma: results of a phase II study. J Clin Oncol 2000; 18: 1550–7PubMed Nasti G, Errante D, Talamini R, et al. Vinorelbine is an effective and safe drug for AIDS-related Kaposi’s sarcoma: results of a phase II study. J Clin Oncol 2000; 18: 1550–7PubMed
182.
go back to reference Spina M, Gabarre J, Rossi G, et al. Stanford V regimen and concomitant HAART in 59 patients with Hodgkin disease and HIV infection. Blood 2002; 100: 1984–8PubMed Spina M, Gabarre J, Rossi G, et al. Stanford V regimen and concomitant HAART in 59 patients with Hodgkin disease and HIV infection. Blood 2002; 100: 1984–8PubMed
183.
go back to reference Errante D, Tirelli U, Gastaldi R, et al. Combined antineoplastic and antiretroviral therapy for patients with Hodgkin’s disease and human immunodeficiency virus infection: a prospective study of 17 patients. Cancer 1994; 73: 437–44PubMed Errante D, Tirelli U, Gastaldi R, et al. Combined antineoplastic and antiretroviral therapy for patients with Hodgkin’s disease and human immunodeficiency virus infection: a prospective study of 17 patients. Cancer 1994; 73: 437–44PubMed
184.
go back to reference Kaplan LD, Straus DJ, Testa MA, et al. Low-dose compared with standard-dose m-BACOD chemotherapy for non-Hodgkin’s lymphoma associated with human immunodeficiency virus infection. N Engl J Med 1997; 336: 1641–8PubMed Kaplan LD, Straus DJ, Testa MA, et al. Low-dose compared with standard-dose m-BACOD chemotherapy for non-Hodgkin’s lymphoma associated with human immunodeficiency virus infection. N Engl J Med 1997; 336: 1641–8PubMed
185.
go back to reference Vaccher E, Spina M, di Gennaro G, et al. Concomitant cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy plus highly active antiretroviral therapy in patients with human immunodeficiency virus-related, non-Hodgkin lymphoma. Cancer 2001; 91: 155–63PubMed Vaccher E, Spina M, di Gennaro G, et al. Concomitant cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy plus highly active antiretroviral therapy in patients with human immunodeficiency virus-related, non-Hodgkin lymphoma. Cancer 2001; 91: 155–63PubMed
186.
go back to reference Baiocchi OCG, Colleoni GWB, Navajas EV, et al. Impact of highly active antiretroviral therapy in the treatment of HIV-infected patients with systemic non-Hodgkin’s lymphoma. Acta Oncol 2002; 41: 192–6PubMed Baiocchi OCG, Colleoni GWB, Navajas EV, et al. Impact of highly active antiretroviral therapy in the treatment of HIV-infected patients with systemic non-Hodgkin’s lymphoma. Acta Oncol 2002; 41: 192–6PubMed
187.
go back to reference Navarro JT, Ribera JM, Oriol A, et al. Influence of highly active anti-retroviral therapy on response to treatment and survival in patients with acquired immunodeficiency syndrome-related non-Hodgkin’s lymphoma treated with cyclophosphamide, hydroxydoxorubicin, vincristine and prednisone. Br J Haematol 2001; 112: 909–15PubMed Navarro JT, Ribera JM, Oriol A, et al. Influence of highly active anti-retroviral therapy on response to treatment and survival in patients with acquired immunodeficiency syndrome-related non-Hodgkin’s lymphoma treated with cyclophosphamide, hydroxydoxorubicin, vincristine and prednisone. Br J Haematol 2001; 112: 909–15PubMed
188.
go back to reference Owellen RJ, Hartke CA, Hains FO. Pharmacokinetics and metabolism of vinblastine in humans. Cancer Res 1977; 37: 2597–602PubMed Owellen RJ, Hartke CA, Hains FO. Pharmacokinetics and metabolism of vinblastine in humans. Cancer Res 1977; 37: 2597–602PubMed
189.
go back to reference Toffoli G, Tolusso Vaccher E, et al. Saquinavir and antineoplastic chemotherapy interactions in in vitro human cancer cell lines and in HIV-associated non-Hodgkin’s lymphoma (HIV-NHL) patients [abstract no. 2153]. 37th Annual Meeting of the American Society of Clinical Oncology; 2001 May 12–15; San Francisco Toffoli G, Tolusso Vaccher E, et al. Saquinavir and antineoplastic chemotherapy interactions in in vitro human cancer cell lines and in HIV-associated non-Hodgkin’s lymphoma (HIV-NHL) patients [abstract no. 2153]. 37th Annual Meeting of the American Society of Clinical Oncology; 2001 May 12–15; San Francisco
190.
go back to reference Cortes J, Thomas D, Rios A, et al. Hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone and highly active antiretroviral therapy for patients with acquired immunodeficiency syndrome-related Burkitt lymphoma/leukemia. Cancer 2002; 94: 1492–9PubMed Cortes J, Thomas D, Rios A, et al. Hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone and highly active antiretroviral therapy for patients with acquired immunodeficiency syndrome-related Burkitt lymphoma/leukemia. Cancer 2002; 94: 1492–9PubMed
191.
go back to reference Horning SJ, Hoppe RT, Breslin S, et al. Stanford V and radiotherapy for locally extensive and advanced Hodgkin’s disease: mature results of a prospective clinical trial. J Clin Oncol 2002; 20: 630–7PubMed Horning SJ, Hoppe RT, Breslin S, et al. Stanford V and radiotherapy for locally extensive and advanced Hodgkin’s disease: mature results of a prospective clinical trial. J Clin Oncol 2002; 20: 630–7PubMed
192.
go back to reference Sparano JA, Wiernik PH, Strack M, et al. Infusional cyclophosphamide, doxorubicin, and etoposide in HIV-related non-Hodgkin’s lymphoma: a follow-up report to a highly active regimen. Leuk Lymphoma 1994; 14: 263–71PubMed Sparano JA, Wiernik PH, Strack M, et al. Infusional cyclophosphamide, doxorubicin, and etoposide in HIV-related non-Hodgkin’s lymphoma: a follow-up report to a highly active regimen. Leuk Lymphoma 1994; 14: 263–71PubMed
193.
go back to reference Sparano JA, Wienik PH, Hu X, et al. Pilot trial of infusional cyclophosphamide, doxorubicin, and etoposide plus didanosine and filgrastim in patients with human immunodeficiency virus-associated non-Hodgkin’s lymphoma. J Clin Oncol 1996; 14: 3026–35PubMed Sparano JA, Wienik PH, Hu X, et al. Pilot trial of infusional cyclophosphamide, doxorubicin, and etoposide plus didanosine and filgrastim in patients with human immunodeficiency virus-associated non-Hodgkin’s lymphoma. J Clin Oncol 1996; 14: 3026–35PubMed
194.
go back to reference Spina M, Vaccher E, Juzbasic S, et al. Human immuno-deficiency virus-related non-Hodgkin lymphoma: activity of infusional cyclophosphamide, doxorubicin, and etoposide as second-line chemotherapy in 40 patients. Cancer 2001; 92: 200–6PubMed Spina M, Vaccher E, Juzbasic S, et al. Human immuno-deficiency virus-related non-Hodgkin lymphoma: activity of infusional cyclophosphamide, doxorubicin, and etoposide as second-line chemotherapy in 40 patients. Cancer 2001; 92: 200–6PubMed
195.
go back to reference van Maanen JMS, de Vries J, Pappie D, et al. Cytochrome P-450-mediated O-demethylation: a route in the metabolic activation of etoposide. Cancer Res 1987; 47: 4658–62PubMed van Maanen JMS, de Vries J, Pappie D, et al. Cytochrome P-450-mediated O-demethylation: a route in the metabolic activation of etoposide. Cancer Res 1987; 47: 4658–62PubMed
196.
go back to reference Mans DR, Retel J, van Maanen JM, et al. Role of the semiquinone free radical of the anti-tumour agent etoposide (VP-16-213) in the inactivation of single- and double-stranded phi X174 DNA. Br J Cancer 1990; 62: 54–60PubMed Mans DR, Retel J, van Maanen JM, et al. Role of the semiquinone free radical of the anti-tumour agent etoposide (VP-16-213) in the inactivation of single- and double-stranded phi X174 DNA. Br J Cancer 1990; 62: 54–60PubMed
197.
go back to reference Haim N, Nemec J, Roman J, et al. Peroxidase-catalyzed metabolism of etoposide (VP-16-213) and covalent binding of reactive intermediates to cellular macromolecules. Cancer Res 1987; 47: 5835–40PubMed Haim N, Nemec J, Roman J, et al. Peroxidase-catalyzed metabolism of etoposide (VP-16-213) and covalent binding of reactive intermediates to cellular macromolecules. Cancer Res 1987; 47: 5835–40PubMed
198.
go back to reference Usui N, Sinha BK. Tyrosinase-induced free radical formation from VP-16,213: relationship to cytotoxicity. Free Radic Res Commun 1990; 10: 287–93PubMed Usui N, Sinha BK. Tyrosinase-induced free radical formation from VP-16,213: relationship to cytotoxicity. Free Radic Res Commun 1990; 10: 287–93PubMed
199.
go back to reference Felix CA, Walker AH, Lange BJ, et al. Association of CYP3A4 genotype with treatment-related leukemia. Proc Natl Acad Sci U S A 1998; 95: 13176–81PubMed Felix CA, Walker AH, Lange BJ, et al. Association of CYP3A4 genotype with treatment-related leukemia. Proc Natl Acad Sci U S A 1998; 95: 13176–81PubMed
200.
go back to reference Relling MV, Yanishevski Y, Nemec J, et al. Etoposide and antimetabolite pharmacology in patients who develop secondary acute myeloid leukemia. Leukemia 1998; 12: 346–52PubMed Relling MV, Yanishevski Y, Nemec J, et al. Etoposide and antimetabolite pharmacology in patients who develop secondary acute myeloid leukemia. Leukemia 1998; 12: 346–52PubMed
201.
go back to reference Sparano JA, Wiernik PH, Hu X, et al. Saquinavir enhances the mucosal toxicity of infusional cyclophosphamide, doxorubicin, and etoposide in patients with HIV-associated non-Hodgkin’s lymphoma. Med Oncol 1998; 15: 50–7PubMed Sparano JA, Wiernik PH, Hu X, et al. Saquinavir enhances the mucosal toxicity of infusional cyclophosphamide, doxorubicin, and etoposide in patients with HIV-associated non-Hodgkin’s lymphoma. Med Oncol 1998; 15: 50–7PubMed
202.
go back to reference Sparano JA, Lee S, Chen MG, et al. Phase II trial of infusional cyclophosphamide, doxorubicin and etoposide in patients with HIV-associated non-Hodgkin’s lymphoma: an Eastern Cooperative Oncology Group trial (E1494). J Clin Oncol 2004; 22: 1491–500PubMed Sparano JA, Lee S, Chen MG, et al. Phase II trial of infusional cyclophosphamide, doxorubicin and etoposide in patients with HIV-associated non-Hodgkin’s lymphoma: an Eastern Cooperative Oncology Group trial (E1494). J Clin Oncol 2004; 22: 1491–500PubMed
203.
go back to reference Bower M, McCall-Peat N, Ryan N, et al. Protease inhibitors potentiate chemotherapy-induced neutropenia. Blood 2004; 104: 2943–6PubMed Bower M, McCall-Peat N, Ryan N, et al. Protease inhibitors potentiate chemotherapy-induced neutropenia. Blood 2004; 104: 2943–6PubMed
204.
go back to reference Baker DK, Relling MV, Pui CH, et al. Increased teniposide clearance with concomitant anticonvulsant therapy. J Clin Oncol 1992; 10: 311–5PubMed Baker DK, Relling MV, Pui CH, et al. Increased teniposide clearance with concomitant anticonvulsant therapy. J Clin Oncol 1992; 10: 311–5PubMed
205.
go back to reference Kehrer DFS, Mathijssen RHJ, Verweij J, et al. Modulation of irinotecan metabolism by ketoconazole. J Clin Oncol 2002; 20: 3122–9PubMed Kehrer DFS, Mathijssen RHJ, Verweij J, et al. Modulation of irinotecan metabolism by ketoconazole. J Clin Oncol 2002; 20: 3122–9PubMed
206.
go back to reference Mathijssen RHJ, Verweij J, de Bruijn P, et al. Effects of St John’s wort on irinotecan metabolism. J Natl Cancer Inst 2002; 94: 1247–9PubMed Mathijssen RHJ, Verweij J, de Bruijn P, et al. Effects of St John’s wort on irinotecan metabolism. J Natl Cancer Inst 2002; 94: 1247–9PubMed
207.
go back to reference Murry DJ, Cherrick I, Salama V, et al. Influence of phenytoin on the disposition of irinotecan: a case report. J Pediatr Hematol Oncol 2002; 24: 130–3PubMed Murry DJ, Cherrick I, Salama V, et al. Influence of phenytoin on the disposition of irinotecan: a case report. J Pediatr Hematol Oncol 2002; 24: 130–3PubMed
208.
go back to reference Gajjar A, Radomski K, Bowers D, et al. Pharmacokinetics of irinotecan (IRN) and metabolites in pediatric high-grade glioma patients with and without co-administration of enzyme-inducing anticonvulsants [abstract no. 626]. 36th Annual Meeting of the American Society of Clinical Oncology; 2000 May 19–23; New Orleans Gajjar A, Radomski K, Bowers D, et al. Pharmacokinetics of irinotecan (IRN) and metabolites in pediatric high-grade glioma patients with and without co-administration of enzyme-inducing anticonvulsants [abstract no. 626]. 36th Annual Meeting of the American Society of Clinical Oncology; 2000 May 19–23; New Orleans
209.
go back to reference Gupta E, Wang X, Ramirez J, et al. Modulation of glucuronidation of SN-38, the active metabolite of irinotecan, by valproic acid and phenobarbital. Cancer Chemother Pharmacol 1997; 40: 440–4 Gupta E, Wang X, Ramirez J, et al. Modulation of glucuronidation of SN-38, the active metabolite of irinotecan, by valproic acid and phenobarbital. Cancer Chemother Pharmacol 1997; 40: 440–4
210.
go back to reference Rosing H, Herben VMM, van Gortel-van Zomeren EH, et al. Isolation and structural confirmation of N-desmethyl topotecan, a metabolite of topotecan. Cancer Chemother Pharmacol 1997; 39: 498–504PubMed Rosing H, Herben VMM, van Gortel-van Zomeren EH, et al. Isolation and structural confirmation of N-desmethyl topotecan, a metabolite of topotecan. Cancer Chemother Pharmacol 1997; 39: 498–504PubMed
211.
go back to reference Rosing H, van Zomeren DM, Doyle E, et al. O-glucuronidation, a newly identified metabolic pathway for topotecan and N-desmethyl topotecan. Anticancer Drugs 1998; 9: 587–92PubMed Rosing H, van Zomeren DM, Doyle E, et al. O-glucuronidation, a newly identified metabolic pathway for topotecan and N-desmethyl topotecan. Anticancer Drugs 1998; 9: 587–92PubMed
212.
go back to reference Zamboni WC, Gajjar AJ, Heideman RL, et al. Phenytoin alters the disposition of topotecan and N-desmethyl topotecan in a patient with medulloblastoma. Clin Cancer Res 1998; 4: 783–9PubMed Zamboni WC, Gajjar AJ, Heideman RL, et al. Phenytoin alters the disposition of topotecan and N-desmethyl topotecan in a patient with medulloblastoma. Clin Cancer Res 1998; 4: 783–9PubMed
213.
go back to reference Kaijser GP, Korst A, Beijnen JH, et al. The analysis of ifosfamide and its metabolites. Anticancer Res 1993; 13: 1311–24PubMed Kaijser GP, Korst A, Beijnen JH, et al. The analysis of ifosfamide and its metabolites. Anticancer Res 1993; 13: 1311–24PubMed
214.
go back to reference Boddy AV, Furtun Y, Sardas S, et al. Individual variation in the activation and inactivation of metabolic pathways of cyclophosphamide. J Natl Cancer Inst 1992; 84: 1744–8PubMed Boddy AV, Furtun Y, Sardas S, et al. Individual variation in the activation and inactivation of metabolic pathways of cyclophosphamide. J Natl Cancer Inst 1992; 84: 1744–8PubMed
215.
go back to reference Wainer IW, Ducharme J, Granvil CP, et al. Ifosfamide stereoselective dechloroethylation and neurotoxicity. Lancet 1994; 383: 982–3 Wainer IW, Ducharme J, Granvil CP, et al. Ifosfamide stereoselective dechloroethylation and neurotoxicity. Lancet 1994; 383: 982–3
216.
go back to reference Ratner L, Lee J, Tang S, et al. Chemotherapy for human immunodeficiency virus-associated non-Hodgkin’s lymphoma in combination with highly active antiretroviral therapy. J Clin Oncol 2001; 19(8): 2171–8PubMed Ratner L, Lee J, Tang S, et al. Chemotherapy for human immunodeficiency virus-associated non-Hodgkin’s lymphoma in combination with highly active antiretroviral therapy. J Clin Oncol 2001; 19(8): 2171–8PubMed
217.
go back to reference Hesse LM, von Moltke LL, Shader RI, et al. Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Drug Metab Dispos 2001; 29: 100–2PubMed Hesse LM, von Moltke LL, Shader RI, et al. Ritonavir, efavirenz, and nelfinavir inhibit CYP2B6 activity in vitro: potential drug interactions with bupropion. Drug Metab Dispos 2001; 29: 100–2PubMed
218.
go back to reference Kumar GN, Dykstra J, Roberts EM, et al. Potent inhibition of the cytochrome P-450 3A-mediated human liver microsomal metabolism of a novel HIV protease inhibitor by ritonavir: a positive drug-drug interaction. Drug Metab Dispos 1999; 27: 902–8PubMed Kumar GN, Dykstra J, Roberts EM, et al. Potent inhibition of the cytochrome P-450 3A-mediated human liver microsomal metabolism of a novel HIV protease inhibitor by ritonavir: a positive drug-drug interaction. Drug Metab Dispos 1999; 27: 902–8PubMed
219.
go back to reference Yu LJ, Drewes P, Gustafsson KG, et al. In vivo modulation of alternative pathways of P-450-catalyzed cyclophosphamide metabolism: impact on pharmacokinetics and antitumor activity. J Pharmacol Exp Ther 1999; 288: 928–37PubMed Yu LJ, Drewes P, Gustafsson KG, et al. In vivo modulation of alternative pathways of P-450-catalyzed cyclophosphamide metabolism: impact on pharmacokinetics and antitumor activity. J Pharmacol Exp Ther 1999; 288: 928–37PubMed
220.
go back to reference Granvil CP, Wang T, Batist G, et al. Influence of phenobarbital induction on the enantioselective N-dechloroethylation of ifosfamide enantiomers in the rat. Drug Metab Dispos 1994; 22: 165–7PubMed Granvil CP, Wang T, Batist G, et al. Influence of phenobarbital induction on the enantioselective N-dechloroethylation of ifosfamide enantiomers in the rat. Drug Metab Dispos 1994; 22: 165–7PubMed
221.
go back to reference Villikka K, Kivisto KT, Neuvonen PJ. The effect of dexamethasone on the pharmacokinetics of triazolam. Pharmacol Toxicol 1998; 83: 135–8PubMed Villikka K, Kivisto KT, Neuvonen PJ. The effect of dexamethasone on the pharmacokinetics of triazolam. Pharmacol Toxicol 1998; 83: 135–8PubMed
222.
go back to reference Kerbusch T, Jansen RLH, Mathot RAA, et al. Modulation of the cytochrome P450-mediated metabolism of ifosfamide by ketoconazole and rifampin. Clin Pharmacol Ther 2001; 70: 132–41PubMed Kerbusch T, Jansen RLH, Mathot RAA, et al. Modulation of the cytochrome P450-mediated metabolism of ifosfamide by ketoconazole and rifampin. Clin Pharmacol Ther 2001; 70: 132–41PubMed
223.
go back to reference Lindley C, Hamilton G, McCune JS, et al. The effect of cyclophosphamide with and without dexamethasone on cytochrome P450 3A4 and 2B6 in human hepatocytes. Drug Metab Dispos 2002; 30: 814–22PubMed Lindley C, Hamilton G, McCune JS, et al. The effect of cyclophosphamide with and without dexamethasone on cytochrome P450 3A4 and 2B6 in human hepatocytes. Drug Metab Dispos 2002; 30: 814–22PubMed
224.
go back to reference Chang TK, Yu L, Maurel P, et al. Enhanced cyclophosphamide and ifosfamide activation in primary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res 1997; 15: 1946–54 Chang TK, Yu L, Maurel P, et al. Enhanced cyclophosphamide and ifosfamide activation in primary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res 1997; 15: 1946–54
225.
go back to reference Boddy AV, Yule SM. Metabolism and pharmacokinetics of oxazaphosphorines. Clin Pharmacokinet 2000; 38: 291–304PubMed Boddy AV, Yule SM. Metabolism and pharmacokinetics of oxazaphosphorines. Clin Pharmacokinet 2000; 38: 291–304PubMed
226.
go back to reference Gatti G, De Pascalis CR, Toffoli G, et al. Case control study of indinavir pharmacokinetics in patients undergoing chemotherapy for AIDS related neoplasia [abstract no. 1.05]. 2nd International Workshop on Clinical Pharmacology of HIV Therapy; 2001 Apr 2–4; Noordwijk Gatti G, De Pascalis CR, Toffoli G, et al. Case control study of indinavir pharmacokinetics in patients undergoing chemotherapy for AIDS related neoplasia [abstract no. 1.05]. 2nd International Workshop on Clinical Pharmacology of HIV Therapy; 2001 Apr 2–4; Noordwijk
227.
go back to reference Rosenthal E, Poizot-Martin I, Saint-Marc T, et al. Phase IV study of liposomal daunorubicin (Daunoxome) in AIDS-related Kaposi Sarcoma. Am J Clin Oncol 2002; 25(1): 57–9PubMed Rosenthal E, Poizot-Martin I, Saint-Marc T, et al. Phase IV study of liposomal daunorubicin (Daunoxome) in AIDS-related Kaposi Sarcoma. Am J Clin Oncol 2002; 25(1): 57–9PubMed
228.
go back to reference Stewart S, Jablonowski H, Goebel FD, et al. Randomized comparative trial of pegylated liposomal doxorubicin versus bleomycin and vincristine in the treatment of AIDS-related Kaposi’s sarcoma. J Clin Oncol 1998; 16: 683–91PubMed Stewart S, Jablonowski H, Goebel FD, et al. Randomized comparative trial of pegylated liposomal doxorubicin versus bleomycin and vincristine in the treatment of AIDS-related Kaposi’s sarcoma. J Clin Oncol 1998; 16: 683–91PubMed
229.
go back to reference Northfelt DW, Dezube BJ, Thommes JA, et al. Pegylatedliposomal doxorubicin versus doxorubicin, bleomycin, and vincristine in the treatment of AIDS-related Kaposi’s sarcoma: results of a randomized phase III clinical trial. J Clin Oncol 1998; 16: 2445–51PubMed Northfelt DW, Dezube BJ, Thommes JA, et al. Pegylatedliposomal doxorubicin versus doxorubicin, bleomycin, and vincristine in the treatment of AIDS-related Kaposi’s sarcoma: results of a randomized phase III clinical trial. J Clin Oncol 1998; 16: 2445–51PubMed
230.
go back to reference Nunez M, Saballs P, Valencia ME, et al. Response to liposomal doxorubicin and clinical outcome of HIV-1 infected patients with Kaposi’s sarcoma receiving highly active antiretroviral therapy. HIV Clin Trials 2001; 2: 429–37PubMed Nunez M, Saballs P, Valencia ME, et al. Response to liposomal doxorubicin and clinical outcome of HIV-1 infected patients with Kaposi’s sarcoma receiving highly active antiretroviral therapy. HIV Clin Trials 2001; 2: 429–37PubMed
231.
go back to reference Rajagopalan S, Politi P, Sinha BK, et al. Adriamycin induced free radical formation in the perfused rat heart: implications for cardiotoxicity. Cancer Res 1988; 48: 4766–9PubMed Rajagopalan S, Politi P, Sinha BK, et al. Adriamycin induced free radical formation in the perfused rat heart: implications for cardiotoxicity. Cancer Res 1988; 48: 4766–9PubMed
232.
go back to reference Myers CE, McGuire WP, Liss RH, et al. Adriamycin: the role of lipid peroxidation in cardiac toxicity and tumor response. Science 1977; 197: 165–7PubMed Myers CE, McGuire WP, Liss RH, et al. Adriamycin: the role of lipid peroxidation in cardiac toxicity and tumor response. Science 1977; 197: 165–7PubMed
233.
go back to reference Gerwitz DA. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem Pharmacol 1999; 57: 727–41 Gerwitz DA. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem Pharmacol 1999; 57: 727–41
234.
go back to reference Gabizon A, Catane R, Uziely B, et al. Prolonged circulation time and enhanced accumulation in malignant exudates of doxorubicin encapsulated in polyethylene-glycol coated liposomes. Cancer Res 1994; 54: 987–92PubMed Gabizon A, Catane R, Uziely B, et al. Prolonged circulation time and enhanced accumulation in malignant exudates of doxorubicin encapsulated in polyethylene-glycol coated liposomes. Cancer Res 1994; 54: 987–92PubMed
235.
go back to reference Toffoli G, Corona G, Cattarossi G, et al. Effect of highly active antiretroviral therapy (HAART) on pharmacokinetics and pharmacodynamics of doxorubicin in patients with HlV-associated non-Hodgkin’s lymphoma. Ann Oncol 2004; 15: 1805–9PubMed Toffoli G, Corona G, Cattarossi G, et al. Effect of highly active antiretroviral therapy (HAART) on pharmacokinetics and pharmacodynamics of doxorubicin in patients with HlV-associated non-Hodgkin’s lymphoma. Ann Oncol 2004; 15: 1805–9PubMed
236.
go back to reference Zhao XJ, Jones DR, Wang YH, et al. Reversible and irreversible inhibition of CYP3A enzymes by tamoxifen and metabolites. Xenobiotica 2002; 10: 863–78 Zhao XJ, Jones DR, Wang YH, et al. Reversible and irreversible inhibition of CYP3A enzymes by tamoxifen and metabolites. Xenobiotica 2002; 10: 863–78
237.
go back to reference Sridar C, Kent UM, Notley LM, et al. Effect of tamoxifen on the enzymatic activity of human cytochrome CYP2B6. J Pharmacol Exp Ther 2002; 301: 945–52PubMed Sridar C, Kent UM, Notley LM, et al. Effect of tamoxifen on the enzymatic activity of human cytochrome CYP2B6. J Pharmacol Exp Ther 2002; 301: 945–52PubMed
238.
go back to reference Dowsett M, Pfister C, Johnston SRD, et al. Impact of tamoxifen on the pharmacokinetics and endocrine effects of the aromatase inhibitor letrozole in postmenopausal women with breast cancer. Clin Cancer Res 1999; 5: 2338–43PubMed Dowsett M, Pfister C, Johnston SRD, et al. Impact of tamoxifen on the pharmacokinetics and endocrine effects of the aromatase inhibitor letrozole in postmenopausal women with breast cancer. Clin Cancer Res 1999; 5: 2338–43PubMed
239.
go back to reference Grimm SW, Dyroff MC. Inhibition of human drug metabolizing cytochromes P450 by anastrozole, a potent and selective inhibitor of aromatase. Drug Metab Dispos 1997; 25: 598–602PubMed Grimm SW, Dyroff MC. Inhibition of human drug metabolizing cytochromes P450 by anastrozole, a potent and selective inhibitor of aromatase. Drug Metab Dispos 1997; 25: 598–602PubMed
240.
go back to reference Lin FI. Rifampin-induced deterioration in steroid-dependent asthma. J Allergy Clin Immunol 1996; 98: 1125PubMed Lin FI. Rifampin-induced deterioration in steroid-dependent asthma. J Allergy Clin Immunol 1996; 98: 1125PubMed
241.
go back to reference Bartoszek M, Brenner AM, Szefler SJ. Prednisolone and methylprednisolone kinetics in children receiving anticonvulsant therapy. Clin Pharmacol Ther 1987; 42: 424–32PubMed Bartoszek M, Brenner AM, Szefler SJ. Prednisolone and methylprednisolone kinetics in children receiving anticonvulsant therapy. Clin Pharmacol Ther 1987; 42: 424–32PubMed
242.
go back to reference Carrie F, Roblot P, Bouquet S, et al. Rifampin-induced nonresponsiveness of giant cell arteritis to prednisone treatment. Arch Intern Med 1994; 154: 1521–4PubMed Carrie F, Roblot P, Bouquet S, et al. Rifampin-induced nonresponsiveness of giant cell arteritis to prednisone treatment. Arch Intern Med 1994; 154: 1521–4PubMed
243.
go back to reference Thiebaut F, Tsuruo T, Hamada H, et al. Cellular localization of the multidrug resistance gene product in normal human tissues. Proc Natl Acad Sci U S A 1987; 84: 7735–8PubMed Thiebaut F, Tsuruo T, Hamada H, et al. Cellular localization of the multidrug resistance gene product in normal human tissues. Proc Natl Acad Sci U S A 1987; 84: 7735–8PubMed
244.
go back to reference Cordon-Cardo C, O’Brien JP, Casals D, et al. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci U S A 1989; 86: 695–8PubMed Cordon-Cardo C, O’Brien JP, Casals D, et al. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci U S A 1989; 86: 695–8PubMed
245.
go back to reference Van Asperen J, Van Teilingen O, Beijnen JH. The pharmacological role of p-glycoprotein in the intestinal epithelium. Pharmacol Res 1998; 37: 429–35PubMed Van Asperen J, Van Teilingen O, Beijnen JH. The pharmacological role of p-glycoprotein in the intestinal epithelium. Pharmacol Res 1998; 37: 429–35PubMed
246.
go back to reference Schellens JHM, Malingre MM, Kruitjzer CMF, et al. Modulation of oral bioavailability of anticancer drugs: from mouse to man. Eur J Pharm Sci 2000; 12: 103–10PubMed Schellens JHM, Malingre MM, Kruitjzer CMF, et al. Modulation of oral bioavailability of anticancer drugs: from mouse to man. Eur J Pharm Sci 2000; 12: 103–10PubMed
247.
go back to reference Matheny CJ, Lamb MW, Brouwer KLR, et al. Pharmacokinetic and pharmacodynamic implications of p-glycoprotein modulation. Pharmacotherapy 2001; 21: 778–96PubMed Matheny CJ, Lamb MW, Brouwer KLR, et al. Pharmacokinetic and pharmacodynamic implications of p-glycoprotein modulation. Pharmacotherapy 2001; 21: 778–96PubMed
248.
go back to reference Stormer E, von Moltke LL, Perloff MD, et al. Differential modulation of P-glycoprotein expression and activity by non-nucleoside HIV-1 reverse transcriptase inhibitors in cell culture. Pharm Res 2002; 19: 1038–45PubMed Stormer E, von Moltke LL, Perloff MD, et al. Differential modulation of P-glycoprotein expression and activity by non-nucleoside HIV-1 reverse transcriptase inhibitors in cell culture. Pharm Res 2002; 19: 1038–45PubMed
249.
go back to reference Drewe J, Gutmann H, Fricker G, et al. HIV protease inhibitor ritonavir: a more potent inhibitor of p-glycoprotein than the cyclosporine analog SDZ PSC 833. Biochem Pharmacol 1999; 57: 1147–52PubMed Drewe J, Gutmann H, Fricker G, et al. HIV protease inhibitor ritonavir: a more potent inhibitor of p-glycoprotein than the cyclosporine analog SDZ PSC 833. Biochem Pharmacol 1999; 57: 1147–52PubMed
250.
go back to reference Washington CB, Duran GE, Man MC, et al. Interaction of anti-HIV protease inhibitors with the multidrug transporter p-glycoprotein (P-gp) in human cultured cells. J Acquir Immune Defic Syndr Hum Retroviral 1998; 19: 203–9 Washington CB, Duran GE, Man MC, et al. Interaction of anti-HIV protease inhibitors with the multidrug transporter p-glycoprotein (P-gp) in human cultured cells. J Acquir Immune Defic Syndr Hum Retroviral 1998; 19: 203–9
251.
go back to reference Srinivas RV, Middlemas D, Flynn P, et al. Human immunodeficiency virus protease inhibitors serve as substrates for multidrug transporter proteins MDR1 and MRP1 but retain antiviral efficacy in cell lines expressing these transporters. Antimicrob Agents Chemother 1998; 42: 3157–62PubMed Srinivas RV, Middlemas D, Flynn P, et al. Human immunodeficiency virus protease inhibitors serve as substrates for multidrug transporter proteins MDR1 and MRP1 but retain antiviral efficacy in cell lines expressing these transporters. Antimicrob Agents Chemother 1998; 42: 3157–62PubMed
252.
go back to reference Perloff MD, von Moltke LL, Marchand JE, et al. Ritonavir induces p-glycoprotein expression, mutidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line. J Pharm Sci 2001; 90: 1829–37PubMed Perloff MD, von Moltke LL, Marchand JE, et al. Ritonavir induces p-glycoprotein expression, mutidrug resistance-associated protein (MRP1) expression, and drug transporter-mediated activity in a human intestinal cell line. J Pharm Sci 2001; 90: 1829–37PubMed
253.
go back to reference Huang L, Wring SA, Woolley JL, et al. Induction of p-glycoprotein and cytochrome P-450 3A by HIV protease inhibitors. Drug Metab Dispos 2001; 29: 754–60PubMed Huang L, Wring SA, Woolley JL, et al. Induction of p-glycoprotein and cytochrome P-450 3A by HIV protease inhibitors. Drug Metab Dispos 2001; 29: 754–60PubMed
254.
go back to reference Washington CB, Wiltshire HR, Man M, et al. The disposition of saquinavir in normal and p-glycoprotein deficient mice, rats, and in cultured cells. Drug Metab Dispos 2000; 28: 1058–62PubMed Washington CB, Wiltshire HR, Man M, et al. The disposition of saquinavir in normal and p-glycoprotein deficient mice, rats, and in cultured cells. Drug Metab Dispos 2000; 28: 1058–62PubMed
255.
go back to reference Choo EF, Leake Wandele, et al. Pharmacological inhibition of p-glycoprotein transport enhances the distribution of HIV-1 protease inhibitors into brain and testes. Drug Metab Dispos 2000; 28: 655–60PubMed Choo EF, Leake Wandele, et al. Pharmacological inhibition of p-glycoprotein transport enhances the distribution of HIV-1 protease inhibitors into brain and testes. Drug Metab Dispos 2000; 28: 655–60PubMed
256.
go back to reference Kim RB, Fromm MF, Wandel C, et al. The drug transporter p-glycoprotein limits oral absorption and brain entry of HIV-1 protease inhibitors. J Clin Invest 1998; 101: 289–94PubMed Kim RB, Fromm MF, Wandel C, et al. The drug transporter p-glycoprotein limits oral absorption and brain entry of HIV-1 protease inhibitors. J Clin Invest 1998; 101: 289–94PubMed
257.
go back to reference Lucia MB, Rutella S, Leone G, et al. In vitro and in vivo modulation of MDR1/p-glycoprotein in HIV-infected patients administered highly active antiretroviral therapy and liposomal doxorubicin. J Acquir Immune Defic Syndr Hum Retroviral 2002; 30: 369–78 Lucia MB, Rutella S, Leone G, et al. In vitro and in vivo modulation of MDR1/p-glycoprotein in HIV-infected patients administered highly active antiretroviral therapy and liposomal doxorubicin. J Acquir Immune Defic Syndr Hum Retroviral 2002; 30: 369–78
258.
go back to reference Gill PS, Miles SA, Mitsuyasu RT, et al. Phase I AIDS Clinical Trials Group (075) study of adriamycin, bleomycin and vincristine chemotherapy with zidovudine in the treatment of AIDS-related Kaposi’s sarcoma. AIDS 1994; 8: 1695–9PubMed Gill PS, Miles SA, Mitsuyasu RT, et al. Phase I AIDS Clinical Trials Group (075) study of adriamycin, bleomycin and vincristine chemotherapy with zidovudine in the treatment of AIDS-related Kaposi’s sarcoma. AIDS 1994; 8: 1695–9PubMed
259.
go back to reference Walker RE, Parker RI, Kovacs JA, et al. Anemia and erythropoiesis in patients with the acquired immunodeficiency syndrome (AIDS) and Kaposi sarcoma treated with zidovudine. Ann Intern Med 1988; 108: 372–6PubMed Walker RE, Parker RI, Kovacs JA, et al. Anemia and erythropoiesis in patients with the acquired immunodeficiency syndrome (AIDS) and Kaposi sarcoma treated with zidovudine. Ann Intern Med 1988; 108: 372–6PubMed
260.
go back to reference Gill PS, Mitsuyasu RT, Montgomery T, et al. AIDS Clinical Trials Group Study 094: a phase I/II trial of ABV chemotherapy with zidovudine and recombinant human GM-CSF in AIDS-related Kaposi’s sarcoma. Cancer J Sci Am 1997; 3: 278–83PubMed Gill PS, Mitsuyasu RT, Montgomery T, et al. AIDS Clinical Trials Group Study 094: a phase I/II trial of ABV chemotherapy with zidovudine and recombinant human GM-CSF in AIDS-related Kaposi’s sarcoma. Cancer J Sci Am 1997; 3: 278–83PubMed
261.
go back to reference Moyle GJ, Sadler M. Peripheral neuropathy with nucleoside antiretrovirale: risk factors, incidence and management. Drug Saf 1998; 19: 481–94PubMed Moyle GJ, Sadler M. Peripheral neuropathy with nucleoside antiretrovirale: risk factors, incidence and management. Drug Saf 1998; 19: 481–94PubMed
Metadata
Title
Interactions Between Antiretrovirals and Antineoplastic Drug Therapy
Authors
Mr Tony Antoniou
Alice L. Tseng
Publication date
01-02-2005
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 2/2005
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.2165/00003088-200544020-00001

Other articles of this Issue 2/2005

Clinical Pharmacokinetics 2/2005 Go to the issue