1
|
Hasegawa K, Shimada M, Takeuchi S, Fujiwara H, Imai Y, Iwasa N, Wada S, Eguchi H, Oishi T, Sugiyama T, Suzuki M, Nishiyama M, Fujiwara K. A phase 2 study of intraperitoneal carboplatin plus intravenous dose-dense paclitaxel in front-line treatment of suboptimal residual ovarian cancer. Br J Cancer 2020; 122:766-770. [PMID: 32001833 PMCID: PMC7078205 DOI: 10.1038/s41416-020-0734-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/23/2019] [Accepted: 01/17/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND We evaluated the efficacy of intraperitoneal (IP) carboplatin in combination with dose-dense paclitaxel (ddTCip) for suboptimal residual ovarian cancer. METHODS This was a phase 2 study to evaluate ddTCip. Patients with stage II-IV ovarian carcinoma, who underwent primary cytoreductive surgery and had radiologically evaluable disease after surgery, were eligible to participate in this study. IP carboplatin (AUC = 6) was administered on day 1, and intravenous paclitaxel (80 mg/m2) was administered on days 1, 8 and 15. The primary endpoint was response rate. Secondary endpoints included progression-free survival (PFS), overall survival (OS) and safety. Interval- debulking surgery followed by the same regimen was allowed when indicated. RESULTS A total of 117 patients were considered eligible for this study prior to surgery and temporarily registered. Of the 117 patients, 76 patients met the inclusion criteria and were enrolled in this study. Fifty-nine (83.1%) patients had objective clinical responses. Median PFS and OS were 18.3 and 55.5 months, respectively. Sixty-four (84.2%) patients had grade 3/4 neutropenia, 43 (56.5%) patients had anaemia and 17 (22.4%) patients had thrombocytopenia. Port-related adverse events occurred in nine (11.8%) patients. CONCLUSIONS Front-line chemotherapy with ddTCip therapy appears safe and effective, even for patients with suboptimal residual ovarian cancer. TRIAL REGISTRATION UMIN Clinical Trials Registry (ID: UMIN000001713) on February 16th, 2009.
Collapse
Affiliation(s)
- Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan.
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan.
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
| | - Satoshi Takeuchi
- Department of Obstetrics and Gynecology, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yuichi Imai
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
| | - Norihiro Iwasa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Satoru Wada
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Hidetaka Eguchi
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| | - Tetsuro Oishi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Tottori, 683-8504, Japan
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, Iwate Medical University, Morioka, Iwate, 020-8505, Japan
| | - Mitsuaki Suzuki
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Masahiko Nishiyama
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
- Division of Translational Research, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
- Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, 350-1298, Japan
- Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, 350-1241, Japan
| |
Collapse
|
2
|
Regimens with intraperitoneal cisplatin plus intravenuous cyclophosphamide and intraperitoneal carboplatin plus intravenuous cyclophosphamide are equally effective in second line intraperitoneal chemotherapy for advanced ovarian cancer. Adv Med Sci 2012; 57:46-50. [PMID: 22430042 DOI: 10.2478/v10039-012-0002-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE We compared response, survival and side effects of regiments with intravenous cyclophosphamide followed by intraperitoneal cisplatin versus intravenous cyclophosphamide followed by intraperitoneal carboplatin as second line treatment in one center retrospective study. MATERIAL AND METHODS Inclusion criteria were: relapse or recurrence of the disease after surgery and first line treatment; stage III histologicaly documented serous epithelial ovarian cancer after one or more prior regiments of chemotherapy. Recurrence were confirmed throughout restaging laparotomy or second look laparotomy. Patients from one of the groups received 90 mg/m(2) cisplatin on the first day and 750 mg/m(2) cyclophosphamide intravenously, while the second group members AUC 6 carboplatin intraperitoneally and 750 mg/m(2) cyclophosphamide intravenously. Four courses were administrated for each patient. RESULTS Of the 49 patients in the cisplatin group the response rates were 21 (43%), 10 (20%) and 18 (37%) in the groups of pathologic complete response, pathologic partial response and progressive disease, respectively. The median survival from the initiation of intraperitoneal chemotherapy was 59 months. Of the 25 patients in the carboplatin group the response rates were 10 (40%), 4 (16%) and 11 (44%) respectively. The median survival -51 months. The differences between the groups were not statistically significant p>0.05 either in response or in toxicity. CONCLUSIONS The results of our research including relatively long survival from intraperotoneal chemotherapy initiation confirm that carboplatin treatment is as good as cisplatin in second line intraperitoneal chemotherapy for ovarian cancer.
Collapse
|
3
|
Duiker EW, de Vries EG, Mahalingam D, Meersma GJ, Boersma-van Ek W, Hollema H, Lub-de Hooge MN, van Dam GM, Cool RH, Quax WJ, Samali A, van der Zee AG, de Jong S. Enhanced Antitumor Efficacy of a DR5-Specific TRAIL Variant over Recombinant Human TRAIL in a Bioluminescent Ovarian Cancer Xenograft Model. Clin Cancer Res 2009; 15:2048-57. [DOI: 10.1158/1078-0432.ccr-08-1535] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
4
|
Muntz HG, Malpass TW, McGonigle KF, Robertson MD, Weiden PL. Phase 2 study of intraperitoneal topotecan as consolidation chemotherapy in ovarian and primary peritoneal carcinoma. Cancer 2008; 113:490-6. [DOI: 10.1002/cncr.23576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
5
|
Bae JH, Lee JM, Ryu KS, Lee YS, Park YG, Hur SY, Ahn WS, Namkoong SE. Treatment of ovarian cancer with paclitaxel- or carboplatin-based intraperitoneal hyperthermic chemotherapy during secondary surgery. Gynecol Oncol 2007; 106:193-200. [PMID: 17466362 DOI: 10.1016/j.ygyno.2007.03.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 03/02/2007] [Accepted: 03/09/2007] [Indexed: 02/08/2023]
Abstract
OBJECTIVE We aimed to evaluate the efficacy and feasibility of treating advanced ovarian cancer with paclitaxel or carboplatin in intraperitoneal hyperthermic chemotherapy (IPHC) during secondary surgery. METHODS We reviewed clinical data of 96 eligible patients with stage Ic-IIIc epithelial ovarian cancer. After primary staging operation and 6-12 cycles of adjuvant chemotherapy, 22 patients were treated with IPHC-paclitaxel (175 mg/m(2)) and 45 patients were treated with IPHC-carboplatin (350 mg/m(2)) during secondary surgery. Survival rates were compared with those of 29 patients treated with only conventional therapy (control group). RESULTS In stage III diseases, 5-year survival rates were 84.6% in IPHC-paclitaxel, 63.0% in IPHC-carboplatin (P=0.4098) and 32.8% in control group (vs. IPHC, P=0.0003). Three-year progression-free survival rates in stage III diseases were both 56.3% in IPHC-paclitaxel and IPHC-carboplatin (P=0.8911) and 16.7% in control group (vs. IPHC, P=0.0028). For the relative risk of disease progression yielded from multivariate analyses, hazard ratio of IPHC-paclitaxel was 0.281 (P=0.0039) and that of IPHC-carboplatin was 0.443 (P=0.0083). Like carboplatin (hazard ratio: 0.396, P=0.0004), IPHC-paclitaxel considerably decreased the risk of death (hazard ratio: 0.197, P=0.0253). CONCLUSION In advanced ovarian cancer, IPHC using paclitaxel or carboplatin during secondary surgery could be a candidate for regional consolidation therapy to prolong survival and hinder disease progression.
Collapse
Affiliation(s)
- Jeong Hoon Bae
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Kangnam St. Mary's Hospital, Catholic University of Korea, Seocho-gu, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Molucon-Chabrot C, Isambert N, Benoit L, Zanetta S, Fraisse J, Guilland JC, Royer B, Monin-Baroille P, Flesch M, Fargeot P, Coudert B, Mayer F, Fumoleau P, Chauffert B. Feasibility of using intraperitoneal epinephrine and cisplatin in patients with advanced peritoneal carcinomatosis. Anticancer Drugs 2007; 17:1211-7. [PMID: 17075321 DOI: 10.1097/01.cad.0000236309.66080.3b] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Intraperitoneal epinephrine above 1 mg/l concentration has been shown to enhance the intratumoral accumulation and antitumor activity of intraperitoneal cisplatin in rats with advanced peritoneal carcinomatosis. The aim of this study was to determine the tolerance of intraperitoneal epinephrine combined with intraperitoneal cisplatin in patients with advanced peritoneal carcinomatosis (17 ovarian cancers, one peritoneal mesothelioma). Intraperitoneal epinephrine (1-5 mg/l) and cisplatin (50 mg/l; 100 mg total dose) were infused in 2 l of saline solution over 2 h. The maximal tolerated concentration of intraperitoneal epinephrine was not reached at 5 mg/l. Cardiovascular symptoms were infrequent and not strictly related to the epinephrine concentration. Tumor responses were obtained in some patients with disease resistant to intravenous platinum compounds. This work demonstrates for the first time that intraperitoneal epinephrine at sufficient concentration enhances the cisplatin effect and can be safely infused into the peritoneal cavity of patients with peritoneal carcinomatosis. The greatest limitation was abdominal pain and limited intraperitoneal distribution of the peritoneal fluid in this closed-abdomen procedure.
Collapse
|
7
|
Michalakis J, Georgatos SD, de Bree E, Polioudaki H, Romanos J, Georgoulias V, Tsiftsis DD, Theodoropoulos PA. Short-term exposure of cancer cells to micromolar doses of paclitaxel, with or without hyperthermia, induces long-term inhibition of cell proliferation and cell death in vitro. Ann Surg Oncol 2007; 14:1220-8. [PMID: 17206477 DOI: 10.1245/s10434-006-9305-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 07/17/2006] [Accepted: 08/08/2006] [Indexed: 01/07/2023]
Abstract
BACKGROUND During intraoperative hyperthermic intraperitoneal chemotherapy for primary or secondary peritoneal malignancies, tumor cells are exposed to high drug concentrations for a relatively short period of time. We investigated in vitro the effect of paclitaxel and hyperthermia on cell proliferation, cell cycle kinetics and cell death under conditions resembling those during intraoperative hyperthermic intraperitoneal chemotherapy. METHODS Human breast MCF-7, ovarian SKOV-3 and hepatocarcinoma HEpG2 cells were exposed to 10 and 20 microM paclitaxel at 37, 41.5 or 43 degrees C for 2 h. Cell proliferation, cell cycle kinetics, necrosis and apoptosis were evaluated. RESULTS Hyperthermia exerted a cytostatic effect to all cell lines and at 43 degrees C a cytotoxic effect on MCF-7 cells. MCF-7 and SKOV-3 cells treated under normothermic conditions with paclitaxel were arrested at G2/M or M phase for at least 3 days. Most of MCF-7 cells and approximately half of SKOV-3 cells were in interphase and became multinucleated without properly completing cytokinesis. Hyperthermia at 41.5 degrees C altered cell cycle distribution and affected the paclitaxel-related effect on cell cycle kinetics of MCF-7 and SKOV-3 cells. Analysis of the mode of cell death showed that cell necrosis prevailed over apoptosis. Hyperthermia at 43 degrees C increased paclitaxel-mediated cytotoxicity in MCF-7 cells and to a lesser extent in SKOV-3 and HEpG2 cells. CONCLUSIONS Short-time treatment of carcinoma cells with high (micromolar) concentrations of paclitaxel in normothermic and hyperthermic conditions is highly efficient for cell growth arrest and could be of clinical relevance in locoregional chemotherapy.
Collapse
Affiliation(s)
- John Michalakis
- Department of Biochemistry, School of Medicine, University of Crete, P.O. Box 2208, 71003, Heraklion, Greece
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Petignat P, du Bois A, Bruchim I, Fink D, Provencher DM. Should intraperitoneal chemotherapy be considered as standard first-line treatment in advanced stage ovarian cancer? Crit Rev Oncol Hematol 2006; 62:137-47. [PMID: 17188887 DOI: 10.1016/j.critrevonc.2006.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Accepted: 11/22/2006] [Indexed: 11/22/2022] Open
Abstract
Current standard therapy for patients with advanced stage epithelial ovarian cancer is cytoreductive surgery followed by combination chemotherapy with paclitaxel and carboplatin. Intraperitoneal (IP) chemotherapy has demonstrated improved outcome compared to standard intravenous treatment in three large randomized phase III trials and confirmed by Cochrane meta-analysis. Although compelling evidence suggests that IP therapy provides survival benefit in a selected group of ovarian cancer patients, it remains unclear which group of patients will really benefit from IP therapy, which is the optimal drug, dose and combination, and what is the real benefit of IP treatment alone. Other concerns about IP therapy are difficulties in completing the assigned treatment and management of its pattern of toxic side-effects. Today, IP chemotherapy has yet to gain a role as standard first-line treatment in advanced stage ovarian cancer. In the near future, efforts should aim at developing an effective IP regimen and research undertaken for a better understanding of the peritoneal environment.
Collapse
Affiliation(s)
- Patrick Petignat
- Gynecologic Oncology Service, Centre Hospitalier de l'Université de Montréal (CHUM)-Hôpital Notre-Dame, Montréal, Quebec, Canada.
| | | | | | | | | |
Collapse
|
9
|
de Bree E, Theodoropoulos PA, Rosing H, Michalakis J, Romanos J, Beijnen JH, Tsiftsis DD. Treatment of ovarian cancer using intraperitoneal chemotherapy with taxanes: from laboratory bench to bedside. Cancer Treat Rev 2006; 32:471-82. [PMID: 16942841 DOI: 10.1016/j.ctrv.2006.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2006] [Revised: 07/02/2006] [Accepted: 07/04/2006] [Indexed: 01/20/2023]
Abstract
The combination of a taxane, paclitaxel or docetaxel, and a platinum compound has become the systemic chemotherapy of choice for primary ovarian cancer and has demonstrated high efficacy. However, ultimately most patients will die from this disease. Hence, there is a need for even more effective systemic chemotherapy or different treatment strategies. Intraperitoneal chemotherapy with taxanes is such an alternative treatment option. Ovarian cancer is theoretically an attractive malignancy for this regional treatment, because the disease remains largely confined to the peritoneal cavity. The choice of taxanes for this kind of chemotherapy is rational, because of its high activity against ovarian cancer cells and expected favourable pharmacokinetics because of limited absorption from the peritoneal cavity due to their large molecular weight and first-pass effect in the liver. In animal model and human pharmacokinetic studies, very high intraperitoneal drug concentrations and exposure and high peritoneal tumour concentrations were achieved, while systemic drug levels were low. The combination of intraperitoneal chemotherapy with hyperthermia enhances the penetration and cytotoxic activity of many drugs. Although data concerning thermal enhancement of taxane cytotoxicity are inconsistent, experimental studies show that at high locoregional concentrations there seems to be such an effect. Recently, feasibility and efficacy of this treatment have evidently been demonstrated in various clinical studies. A large randomized trial revealed improvement of outcome by intraperitoneal instillation chemotherapy with paclitaxel and cisplatin as first-line treatment. Moreover, promising results have been observed after intraoperative hyperthermic intraperitoneal chemotherapy with docetaxel for recurrent disease.
Collapse
Affiliation(s)
- Eelco de Bree
- Department of Surgical Oncology, Medical School of Crete University Hospital, P.O. Box 1352, 71110 Herakleion, Greece.
| | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
UNLABELLED A large, recent study has shown significantly improved survival in women with epithelial ovarian cancer treated with intraperitoneal chemotherapy. This review is intended for all clinicians caring for women with ovarian cancer, including family physicians, general gynecologists, and oncologists. The subset of patients most likely to derive a survival benefit from intraperitoneal chemotherapy should be distinguished. Because effective surgical debulking is critical to long-term survival for ovarian cancer, it is important that women known or suspected to have ovarian cancer should be referred to centers with the surgical expertise and resources necessary for aggressive tumor debulking and safe delivery of intraperitoneal chemotherapy. TARGET AUDIENCE Obstetricians & Gynecologists, Family Physicians. LEARNING OBJECTIVES After completion of this article, the reader should be able to state that there is a subset of women with widespread intraabdominal ovarian cancer who will have improved survival after a debulking procedure and intraperitoneal (IP) chemotherapy and explain that there needs to be the expertise and resources necessary to follow these patients.
Collapse
Affiliation(s)
- Maryam Alhayki
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology University of Ottawa, Ottawa, Canada.
| | | | | |
Collapse
|
11
|
Lardinois D, Jung FJ, Opitz I, Rentsch K, Latkoczy C, Vuong V, Varga Z, Rousson V, Günther D, Bodis S, Stahel R, Weder W. Intrapleural topical application of cisplatin with the surgical carrier Vivostat increases the local drug concentration in an immune-competent rat model with malignant pleuromesothelioma. J Thorac Cardiovasc Surg 2006; 131:697-703. [PMID: 16515926 DOI: 10.1016/j.jtcvs.2005.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Revised: 08/05/2005] [Accepted: 08/17/2005] [Indexed: 11/29/2022]
Abstract
OBJECTIVE We sought to investigate whether intrapleural topical application of cisplatin with a surgical carrier has a prolonged local tissue level in comparison with cisplatin solution while reducing systemic toxicity. METHODS Forty immune-competent Fischer rats were inoculated with 10(6) mesothelioma cells. Ten days later, left pneumonectomy with tumor debulking was performed. Twenty animals underwent local application of cisplatin solution (100 mg/m2), whereas the same quantity of cisplatin was topically applied as a gel with the Vivostat (Vivolution) system in 20 other animals. In each group 5 subgroups of 4 animals were defined according to the harvesting time of blood and tissue samples (2, 4, 24, and 72 hours and 1 week) after local therapy. Platinum concentrations in serum and tissue and systemic toxicity were analyzed. RESULTS Platinum concentrations in tissue were significantly higher in the gel group (group 1) than in the solution group (group 2) at 1, 3, and 7 days after therapy (1510, 1224, and 1069 pg/mg for group 1 vs 598, 382, and 287 pg/mg for group 2; P = .007, P = .005, and P = .0002, respectively). Laboratory findings showed renal insufficiency in the animals of the solution group at 1 week, with values of 98 mmol/L versus 7.7 mmol/L for urea and 410 mumol/L versus 43 mumol/L for creatinine (P = .02 and P = .01, respectively), which was confirmed by means of pathologic analysis. CONCLUSIONS Intrapleural administration of cisplatin with the carrier Vivostat significantly provides sustained higher platinum concentrations up to 1 week in tissue in comparison with application of cisplatin solution without conferring systemic toxicity in this model.
Collapse
Affiliation(s)
- D Lardinois
- Division of Thoracic Surgery, University Hospital, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
de Bree E, Rosing H, Michalakis J, Romanos J, Relakis K, Theodoropoulos PA, Beijnen JH, Georgoulias V, Tsiftsis DD. Intraperitoneal chemotherapy with taxanes for ovarian cancer with peritoneal dissemination. Eur J Surg Oncol 2006; 32:666-70. [PMID: 16618534 DOI: 10.1016/j.ejso.2006.03.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Accepted: 03/02/2006] [Indexed: 12/17/2022] Open
Abstract
Paclitaxel and docetaxel are currently the two clinically available taxanes. The combination of a taxane and a platinum compound has become the systemic chemotherapy of choice for primary ovarian cancer. Despite the high activity of these drugs in systemic chemotherapy, the majority of patients with advanced ovarian cancer will develop recurrent disease and ultimately decease of this disease. Therefore, more effective systemic chemotherapy regimens or alternative treatment modalities are warranted. Intraperitoneal chemotherapy is such an alternative treatment option. Pharmacokinetic studies on intraperitoneal administration of paclitaxel and docetaxel demonstrated very high locoregional drug concentrations and exposure. Their activity and response seem to be dose-dependent and hence higher efficacy with limited systemic toxicity is to be expected. Intraperitoneal chemotherapy may be combined intraoperatively with hyperthermia, which enhances tissue penetration and cytotoxic activity of many drugs. The data concerning thermal enhancement of taxanes are inconsistent, but at the high locoregional concentrations provided by intraperitoneal drug administration such a thermal enhancement seems to exist. Clinical studies have clearly demonstrated the feasibility and efficacy of intraperitoneal instillation chemotherapy with taxanes in patients with ovarian cancer. Preliminary results of a phase III study demonstrated improved outcome with the addition of intraperitoneal instillation chemotherapy to systemic chemotherapy after optimal primary cytoreductive surgery. Intraoperative hyperthermic intraperitoneal chemotherapy with docetaxel has been performed in a single study, in which promising results were observed. Further clinical investigations with an adequate follow-up period are needed to confirm the promising initial results and to determine the exact efficacy of intraperitoneal chemotherapy with these drugs.
Collapse
Affiliation(s)
- E de Bree
- Department of Surgical Oncology, Medical School of Crete University Hospital, Herakleion, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstracts from the Chemotherapy Foundation Symposium XXIV: Innovative Cancer Therapy for Tomorrow, November 8-11, 2006, New York, New York, USA. Cancer Invest 2006; 24 Suppl 1:1-59. [PMID: 16546846 DOI: 10.1080/07357900600560838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
14
|
Gadducci A, Cosio S, Conte PF, Genazzani AR. Consolidation and maintenance treatments for patients with advanced epithelial ovarian cancer in complete response after first-line chemotherapy: A review of the literature. Crit Rev Oncol Hematol 2005; 55:153-66. [PMID: 15890524 DOI: 10.1016/j.critrevonc.2005.03.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Accepted: 03/16/2005] [Indexed: 02/01/2023] Open
Abstract
Most patients with advanced epithelial ovarian cancer experience objective responses to paclitaxel/platinum-based chemotherapy, but responses are generally short-lived and the clinical outcome is still unsatisfactory. Therefore, the strategy to consolidate and to prolong the duration of response is very attractive. Different consolidation or maintenance treatments have been attempted, such as whole abdomen radiotherapy, intraperitoneal chromic phosphate, radioimmunotherapy, intraperitoneal chemotherapy, high-dose chemotherapy with haematopoietic support, prolonged administration of the first-line regimen, second-line single-agent chemotherapy, and biological agents. Clinical studies have given conflicting, inconclusive, and generally disappointing results. A recent US randomised trial appeared to show that the prolonged administration of single-agent paclitaxel (175 mg/m2 every 3 weeks) significantly improved the progression-free survival of complete responders to paclitaxel/platinum-based chemotherapy. Alternative less toxic, and probably more effective schedules of administration of chemotherapy (i.e. weekly paclitaxel) might assure a better balance between quality of life and anti-tumor activity in patients previously exposed to chemotherapy.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Procreative Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 56, Pisa 56127, Italy.
| | | | | | | |
Collapse
|
15
|
Bos AME, De Vos FYFL, de Vries EGE, Beijnen JH, Rosing H, Mourits MJE, van der Zee AGJ, Gietema JA, Willemse PHB. A phase I study of intraperitoneal topotecan in combination with intravenous carboplatin and paclitaxel in advanced ovarian cancer. Eur J Cancer 2005; 41:539-48. [PMID: 15737558 DOI: 10.1016/j.ejca.2004.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Revised: 10/25/2004] [Accepted: 12/03/2004] [Indexed: 11/16/2022]
Abstract
The aim of this study was to determine the maximum tolerated dose (MTD) of intraperitoneal (i.p.) topotecan combined with standard doses of intravenous (i.v.) carboplatin and paclitaxel and to investigate its pharmacokinetics. Women with primary ovarian cancer stage IIb - IV received six cycles of i.v. carboplatin and paclitaxel with escalating topotecan doses i.p. of 10, 15, 20 and 25 mg/m(2). Twenty-one patients entered this trial. Febrile neutropenia, thrombocytopenia requiring platelet transfusion and fatigue grade 3 were dose-limiting toxicities (DLT) at 25 mg/m(2) i.p. and 20 mg/m(2) i.p. of topotecan was considered to be the MTD. The mean plasma t(1/2) was 3.8 +/- 2.3 h for total topotecan and 4.4 +/- 3.9 h for active lactone. The area under the curve (AUC) was proportional with dose, R = 0.54, p < 0.05 for total topotecan and the peritoneal / plasma AUC ratio was 46 +/- 30. Fifteen patients who completed treatment had a median progression-free survival (PFS) of 27 months. In this setting the MTD of topotecan is 20 mg/m(2) i.p. The efficacy of this regimen should be explored further in a formal phase III study.
Collapse
Affiliation(s)
- A M E Bos
- Department of Medical Oncology, University Hospital Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Adam RA, Adam YG. Malignant ascites: past, present, and future. J Am Coll Surg 2004; 198:999-1011. [PMID: 15194082 DOI: 10.1016/j.jamcollsurg.2004.01.035] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Revised: 01/13/2004] [Accepted: 01/16/2004] [Indexed: 02/07/2023]
Affiliation(s)
- Rony A Adam
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA 30303, USA
| | | |
Collapse
|
17
|
Snyder EL, Meade BR, Saenz CC, Dowdy SF. Treatment of terminal peritoneal carcinomatosis by a transducible p53-activating peptide. PLoS Biol 2004; 2:E36. [PMID: 14966535 PMCID: PMC340944 DOI: 10.1371/journal.pbio.0020036] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Accepted: 12/03/2003] [Indexed: 12/04/2022] Open
Abstract
Advanced-stage peritoneal carcinomatosis is resistant to current chemotherapy treatment and, in the case of metastatic ovarian cancer, results in a devastating 15%–20% survival rate. Therapeutics that restore genes inactivated during oncogenesis are predicted to be more potent and specific than current therapies. Experiments with viral vectors have demonstrated the theoretical utility of expressing the p53 tumor suppressor gene in cancer cells. However, clinically useful alternative approaches for introducing p53 activity into cancer cells are clearly needed. It has been hypothesized that direct reactivation of endogenous p53 protein in cancer cells will be therapeutically beneficial, but few tests of this hypothesis have been carried out in vivo. We report that a transducible D-isomer RI-TATp53C′ peptide activates the p53 protein in cancer cells, but not normal cells. RI-TATp53C′ peptide treatment of preclinical terminal peritoneal carcinomatosis and peritoneal lymphoma models results in significant increases in lifespan (greater than 6-fold) and the generation of disease-free animals. These proof-of-concept observations show that specific activation of endogenous p53 activity by a macromolecular agent is therapeutically effective in preclinical models of terminal human malignancy. Our results suggest that TAT-mediated transduction may be a useful strategy for the therapeutic delivery of large tumor suppressor molecules to malignant cells in vivo. Specific activiation of the tumor suppressor protein p53, using a transducible p53 C-terminal peptide, dramatically increases survival in a mouse model of peritoneal carcinomatosis. This peptide offers therapeutic potential for tumors in which p53 is mutated
Collapse
Affiliation(s)
- Eric L Snyder
- 1Howard Hughes Medical Institute, Chevy ChaseMarylandUnited States of America
- 2Washington University School of Medicine, St. LouisMissouriUnited States of America
| | - Bryan R Meade
- 3Department of Cellular and Molecular Medicine, School of MedicineUniversity of California, San Diego, La Jolla, CaliforniaUnited States of America
| | - Cheryl C Saenz
- 1Howard Hughes Medical Institute, Chevy ChaseMarylandUnited States of America
- 4Department of Reproductive Medicine, School of MedicineUniversity of California, San Diego, La Jolla, CaliforniaUnited States of America
| | - Steven F Dowdy
- 1Howard Hughes Medical Institute, Chevy ChaseMarylandUnited States of America
- 3Department of Cellular and Molecular Medicine, School of MedicineUniversity of California, San Diego, La Jolla, CaliforniaUnited States of America
| |
Collapse
|
18
|
Katz MH, Barone RM. The rationale of perioperative intraperitoneal chemotherapy in the treatment of peritoneal surface malignancies. Surg Oncol Clin N Am 2003; 12:673-88. [PMID: 14567024 DOI: 10.1016/s1055-3207(03)00034-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In summary, the use of perioperative intraperitoneal chemotherapy is a rational and scientifically sound treatment option for patients with peritoneal carcinomatosis. By delivering chemotherapeutic agents directly into the peritoneal cavity in the perioperative period, after cytoreductive procedures resulting in minimal residual tumor load, the cytotoxicity, efficacy, and safety of these agents can be maximized. The use of this treatment strategy in the intraoperative or perioperative period ensures that the efficacy of the chemotherapeutic agents is not reduced by limitations of abdominal compartmentalization and scarring. Treating patients under hyperthermic conditions may confer an additional benefit. Although the use of perioperative chemotherapy or hyperthermic intraperitoneal chemotherapy is not yet part of the standard of care for the treatment of advanced abdominal malignancies, both basic science and clinical investigations have confirmed the validity of these regimens. Further clinical studies in a cooperative group setting are necessary to prove the efficacy of perioperative intraperitoneal chemotherapy in both the treatment and prevention of peritoneal surface malignancy.
Collapse
Affiliation(s)
- Matthew H Katz
- Department of Surgery, University of California at San Diego, Medical Center, 200 West Arbor Drive, San Diego, CA 92103, USA
| | | |
Collapse
|
19
|
Abstract
Intraperitoneal chemotherapy provides a means by which high concentrations of drugs and long durations of tissue exposure can be attained at the peritoneal surface. It has been studied widely in ovarian cancer, a disease in which intra-abdominal progression remains the major source of morbidity and mortality. Three large randomized trials have shown improved survival in optimally debulked patients who were treated with intraperitoneal chemotherapy as part of a front-line regimen, yet it has not become part of usual therapy. Several factors have contributed to the reluctance to adopt intraperitoneal therapy, including technical issues related to drug delivery and the fact that all of the large randomized trials employed intraperitoneal cisplatin, which has more toxicity than intravenous carboplatin, the current standard of care. Future research is needed for further definition of the clinical benefit of intraperitoneal chemotherapy, modification of existing regimens to minimize side effects, and exploration of intraperitoneal biologic, immunologic, and gene therapy techniques.
Collapse
Affiliation(s)
- Gregory Friberg
- The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 2115, Chicago, IL 60637-1470, USA
| | | |
Collapse
|
20
|
Link KH, Roitman M, Holtappels M, Runnebaum I, Urbanzyk H, Leder G, Staib L. Intraperitoneal chemotherapy with mitoxantrone in malignant ascites. Surg Oncol Clin N Am 2003; 12:865-72, xvi-xvii. [PMID: 14567037 DOI: 10.1016/s1055-3207(03)00050-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A retrospective analysis of intraperitoneal mitoxantrone instillation therapy for malignant ascites in advanced breast and gynecologic pelvic cancers was performed to confirm the efficacy and safety of this therapy. Several smaller phase II trials had suggested good palliative effects. In 143 patients (37 breast cancer and 106 gynecologic cancers), 257 instillations were registered. Response in breast cancer was induced in 49% and in 63% with gynecologic cancer. Severe or life-threatening clinical or laboratory side effects related to intraperitoneal mitoxantrone occurred in 2.7% (clinical) or 1.9% (laboratory) of the 257 instillations. Induction of adverse side effect was dose dependent. Intraperitoneal chemotherapy with mitoxantrone for treatment of malignant ascites in breast cancer and gynecologic malignancy is effective and well tolerated. For this treatment 30 mg mitoxantrone in > or = 1000 mL carrier solution (e.g., saline) is recommended. A minimal concentration of at least 10 micrograms/mL should be achieved.
Collapse
Affiliation(s)
- K H Link
- Department of Visceral and Oncologic Surgery, Asklepios Paulinen Klinik and Asklepios, Tumor Center Rhein-Main (ATC), Geisenheimer Str. 10, D 65197 Wiesbaden, Germany. k-h.link.@asklepios.com
| | | | | | | | | | | | | |
Collapse
|
21
|
Rothenberg ML, Liu PY, Braly PS, Wilczynski SP, Hannigan EV, Wadler S, Stuart G, Jiang C, Markman M, Alberts DS. Combined intraperitoneal and intravenous chemotherapy for women with optimally debulked ovarian cancer: results from an intergroup phase II trial. J Clin Oncol 2003; 21:1313-9. [PMID: 12663720 DOI: 10.1200/jco.2003.07.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE The median survival time for women with optimally debulked adenocarcinoma of the ovary treated with intravenous (IV) chemotherapy is 41 to 52 months, and the 2-year survival rate is 65% to 70%. Recent studies evaluating intraperitoneal (IP) chemotherapy have reported a median survival time of 49 to 63 months and 2-year survival rates of 70% to 80%. This phase II trial was undertaken to evaluate the feasibility of and 2-year survival rate achieved by the combination of IP paclitaxel, IP cisplatin, and IV paclitaxel in women with optimally debulked, stage III ovarian cancer. PATIENTS AND METHODS Treatment consisted of paclitaxel 135 mg/m(2) IV over 24 hours on days 1 to 2, cisplatin 100 mg/m(2) IP on day 2, and paclitaxel 60 mg/m(2) IP on day 8 administered every 21 days for six cycles. RESULTS In 68 assessable women with optimal stage III ovarian cancer, the 2-year survival rate was 91%, and the median survival time was 51 months. The 2-year disease-free survival rate was 66%, and median disease-free survival time was 33 months. Ninety-six percent of all patients experienced at least one grade 3 to 4 adverse event during therapy, with the most common events being neutropenia (79%), nausea (50%), vomiting (34%), and fatigue/malaise/lethargy (24%). Seventy-one percent of patients completed all six cycles of IV/IP therapy as planned. CONCLUSION Combined IV and IP chemotherapy with cisplatin and paclitaxel is associated with a very promising 2-year survival rate in women with optimally debulked ovarian cancer. The ultimate impact of this approach on overall survival requires further evaluation in a randomized trial setting.
Collapse
|
22
|
de Bree E, Rosing H, Beijnen JH, Romanos J, Michalakis J, Georgoulias V, Tsiftsis DD. Pharmacokinetic study of docetaxel in intraoperative hyperthermic i.p. chemotherapy for ovarian cancer. Anticancer Drugs 2003; 14:103-10. [PMID: 12569296 DOI: 10.1097/00001813-200302000-00003] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The purpose of this study was to evaluate the pharmacokinetics and toxicity of docetaxel in continuous hyperthermic perfusion peritoneal chemotherapy (CHPPC) after cytoreductive surgery for peritoneal involvement of gynecological malignancies, mainly ovarian cancer. Eighteen patients, with a mean age of 64 years (range 51-80), underwent cytoreductive surgery and subsequent CHPPC with 75 mg/m2 docetaxel at 41-43 degrees C. One patient was treated twice. In eight cases, peritoneal fluid and blood samples were obtained for pharmacokinetic analysis. Death occurred in two heavily pretreated elderly patients with a high volume i.p. tumor recurrence, probably reflecting poor patient selection (mortality rate 10.5%). Other complications, mainly minor, were recorded after 63% of the procedures. Hematological docetaxel-induced toxicity was limited, while the incidence of wound complications was relatively high and probably caused by the direct exposure of the wound to docetaxel during CHPPC. The maximal i.p. versus plasma concentration ratio ranged from 17 to 95 (average 45), while the i.p. versus systemic exposure ratio varied between 105 and 555 (average 207). We conclude that the use of docetaxel in CHPPC following cytoreductive surgery seems feasible and results in a high i.p. versus systemic exposure ratio. The AUC for the peritoneal cavity is on average 13-27 times higher after i.p. administration of 75 mg/m2 during CHPPC than the AUC achieved in the systemic compartment after i.v. administration of the recommended dose of 100 mg/m2, while docetaxel-induced systemic toxicity is highly limited.
Collapse
Affiliation(s)
- Eelco de Bree
- Department of Surgical Oncology, University Hospital, Herakleion, Greece.
| | | | | | | | | | | | | |
Collapse
|
23
|
Anaf V, Gangji D, Simon P, Saylam K. Laparoscopical insertion of intraperitoneal catheters for intraperitoneal chemotherapy. Acta Obstet Gynecol Scand 2003; 82:1140-5. [PMID: 14616261 DOI: 10.1046/j.1600-0412.2003.00210.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND METHODS Eight patients (seven clinically negative stage Ill ovarian cancer and one peritoneal mesothelioma) respectively underwent second-look laparoscopy for staging, adhesiolysis and insertion of an intraperitoneal catheter and fixation of a portal. All patients had received six courses of cisplatin-paclitaxel-based chemotherapy intravenously. At the end of the laparoscopic staging, a 5-mm catheter was inserted under direct vision through a 5-mm trocar in the abdomen. A preaponevrotic forceps was used to grasp the catheter and bring it to the portal, which is located on the intercostal aponevrosis 2 or 3 cm above the laparoscope entry. RESULTS Although previously operated, laparoscopy was possible in all patients and the catheters were easily inserted. All patients received intraperitoneal chemotherapy on the second postoperative day. We did not observe any complication after a mean follow-up of 12 months. CONCLUSIONS Laparoscopic insertion of intraperitoneal catheters is a feasible and safe procedure but requires experience in laparoscopic surgery. In many cases it might preclude from performing non-useful laparotomies. It might help to reduce the hospital stay and the morbidity in relation to second-look laparotomies. Compared with the blind surgical technique of insertion of intraperitoneal catheters, this technique also allows intraabdominal staging. Nevertheless, further studies are necessary to confirm our results.
Collapse
Affiliation(s)
- Vincent Anaf
- Department of Gynecology, Free University of Brussels, Academic Hospital Erasme, Brussels, Belgium
| | | | | | | |
Collapse
|
24
|
Lee MJ, Cho SS, You JR, Lee Y, Kang BD, Choi JS, Park JW, Suh YL, Kim JA, Kim DK, Park JS. Intraperitoneal gene delivery mediated by a novel cationic liposome in a peritoneal disseminated ovarian cancer model. Gene Ther 2002; 9:859-66. [PMID: 12080380 DOI: 10.1038/sj.gt.3301704] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2001] [Accepted: 02/19/2002] [Indexed: 01/22/2023]
Abstract
We have previously synthesized a new cationic liposome that displays high efficiency and low toxicity, 3 beta[l-ornithinamide-carbamoyl] cholesterol (O-Chol), using solid-phase synthesis. In this study, O-Chol was applied to in vitro and in vivo models of ovarian cancer. Intraperitoneal gene delivery for peritoneal disseminated ovarian cancer in nude mice was achieved using a stable chloramphenicol acetyl transferase (CAT)-expressing ovarian cancer cell line (OV-CA-2774/CAT), which allowed us to quantify the exact tumor burden of organs. When luciferase and beta-galactosidase genes were used as reporter genes, O-Chol showed better efficiency than other commercial transfection reagents such as lipofectin, lipofectAMINE, DC-Chol, and FuGENE 6, both in vitro and in vivo. Moreover, the transfection efficiency of this new cationic lipid reagent remained high in serum-containing medium and under serum-free conditions. Furthermore, in vivo transfection with O-Chol showed high levels of gene expression specific to peritoneal tumor cells. Consequently, the O-Chol:DNA lipoplex appears to offer potential advantages over other commercial transfection reagents because of (1) its higher level of gene expression in vitro and in vivo; (2) its reduced susceptibility to serum inhibition; and (3) its highly selective transfection into tumor cells. These results suggest that the O-Chol:DNA lipoplex is a promising tool in gene therapy for patients with peritoneal disseminated ovarian cancer.
Collapse
Affiliation(s)
- M-J Lee
- School of Chemistry, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bonhomme-Faivre L, Pelloquin A, Tardivel S, Urien S, Mathieu MC, Castagne V, Lacour B, Farinotti R. Recombinant interleukin-2 treatment decreases P-glycoprotein activity and paclitaxel metabolism in mice. Anticancer Drugs 2002; 13:51-7. [PMID: 11914641 DOI: 10.1097/00001813-200201000-00006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Recombinant rIL-2 was reported to be able to decrease P-glycoprotein (P-gp) expression in cultured cells from human colon carcinoma. P-gp is considered an important factor in the control of Taxol efflux from tumor cells. Based on the premise that Taxol pharmacokinetic parameters could be modified as a result of diminished P-gp expression induced by recombinant interleukin (rIL)-2 and that this might elicit an interaction between the two drugs, we evaluated the pharmacokinetics of a novel strategy combining i.p. immunotherapy with rIL-2 and a cytotoxic agent, Taxol. Mice were allocated to two groups treated with rIL-2 (15 microg x 2/day from day 1 to 4) then Taxol (10 mg/kg i.p. day 5) or Taxol (10 mg/kg i.p.) alone (control group). The Taxol + rIL-2 combination provoked the development of ascites, presumably due to the presence of Cremophor EL in the Taxol preparation. Paclitaxel was measured in plasma and ascites by HPLC with UV detection. Paclitaxel pharmacokinetics were strongly modified by rIL-2 pretreatment. Compared to that observed in control mice, the apparent volume of distribution increased dramatically (Vd/F = 18.2 versus 4.1 l/kg) and the apparent plasma clearance decreased (Cl/F = 1.12 versus 1.66 l/h/kg). P-gp expression was determined in the liver, lung, intestine, brain and kidney in the two groups by immunodetection with the C219 anti-P-gp monoclonal antibody. A significant decrease in P-gp expression was observed in the intestine and in the brain in the rIL-2-pretreated mice as compared to controls. To study the functionality of P-gp, we compared digoxin (a model P-gp substrate) pharmacokinetics before and after pretreatment with rIL-2 (10 microg x 2/day from day 1 to 4), after a single 1 microg oral dose of digoxin used to quantify P-gp activity. Results showed a decrease in oral digoxin clearance after rIL-2 pretreatment indicating modified P-gp activity. We conclude that rIL-2 pretreatment is able to decrease P-gp activity and paclitaxel metabolism in vivo. This is the first study to demonstrate a decrease in P-gp activity and expression in organs such as the brain in vivo. A novel strategy combining immunotherapy with rIL-2 and a cytotoxic agent could potentially improve clinical results, particularly in brain cancer.
Collapse
|
26
|
Hofstra LS, Bos AM, de Vries EG, van der Zee AG, Beijnen JH, Rosing H, Mulder NH, Aalders JG, Willemse PH. A phase I and pharmacokinetic study of intraperitoneal topotecan. Br J Cancer 2001; 85:1627-33. [PMID: 11742479 PMCID: PMC2363984 DOI: 10.1054/bjoc.2001.2161] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To evaluate the feasibility and pharmacology of intraperitoneal (IP) topotecan. PATIENTS AND METHODS Fifteen patients with recurrent ovarian cancer in a phase I trial were treated with escalating IP topotecan doses (5-30 mg/m(2)) for pharmacokinetic analysis. RESULTS Dose limiting toxicity (DLT) was acute hypotension, chills and fever at the 30 mg/m(2) dose level. Haematological toxicity and abdominal pain were mild for all dose levels studied. PHARMACOKINETICS Peak plasma levels of total topotecan were reached at 2.7 +/- 1.1 h after IP instillation. The apparent V(ss) was 69.9 +/- 25.4 L/m(2), plasma clearance 13.4 +/- 2.5 L/h/m(2) and plasma T1/2 3.7 +/- 1.3 h. The plasma AUC was correlated with the dose (R = 0.95, P < 0.01). The plasma AUC ratio of lactone versus total topotecan (lactone + carboxy-forms) increased with the dose from 16% to 55%, (R = 0.84, P < 0.01). Peritoneal total topotecan was cleared from the peritoneal cavity at 0.4 +/- 0.3 L/h.m(2) with a T1/2 = 2.7 +/- 1.7 h. The mean peritoneal/plasma AUC ratio for total topotecan was 54 +/- 34. CONCLUSION A substantial dose of topotecan can be delivered by the IP route, achieving cytotoxic plasma levels of topotecan, with acceptable toxicity. The recommended dose for further phase II trials is 20 mg/m(2) IP, which enables combination with active doses of other cytotoxic drugs, in view of its limited myelotoxicity when given by this route.
Collapse
Affiliation(s)
- L S Hofstra
- Department of Medical, Gynaecologic Oncology, University Hospital Groningen, PO Box 30.001, Groningen, RB, 9700, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|