Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Ann Oncol ; 20(8): 1344-51, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19468030

ABSTRACT

BACKGROUND: The role of adjuvant dose-intensive chemotherapy and its efficacy according to baseline features has not yet been established. PATIENTS AND METHODS: Three hundred and forty-four patients were randomized to receive seven courses of standard-dose chemotherapy (SD-CT) or three cycles of dose-intensive epirubicin and cyclophosphamide (epirubicin 200 mg/m(2) plus cyclophosphamide 4 mg/m(2) with filgrastim and progenitor cell support). All patients were assigned tamoxifen at the completion of chemotherapy. The primary end point was disease-free survival (DFS). This paper updates the results and explores patterns of recurrence according to predicting baseline features. RESULTS: At 8.3-years median follow-up, patients assigned DI-EC had a significantly better DFS compared with those assigned SD-CT [8-year DFS percent 47% and 37%, respectively, hazard ratio (HR) 0.76; 95% confidence interval 0.58-1.00; P = 0.05]. Only patients with estrogen receptor (ER)-positive disease benefited from the DI-EC (HR 0.61; 95% confidence interval 0.39, 0.95; P = 0.03). CONCLUSIONS: After prolonged follow-up, DI-EC significantly improved DFS, but the effect was observed only in patients with ER-positive disease, leading to the hypothesis that efficacy of DI-EC may relate to its endocrine effects. Further studies designed to confirm the importance of endocrine responsiveness in patients treated with dose-intensive chemotherapy are encouraged.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Adult , Aged , Amenorrhea , Breast Neoplasms/metabolism , Breast Neoplasms/surgery , Cyclophosphamide/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Epirubicin/administration & dosage , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Middle Aged , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/surgery , Receptors, Estrogen/biosynthesis , Recombinant Proteins , Stem Cell Transplantation , Survival Rate , Tamoxifen/administration & dosage , Treatment Outcome
2.
J Thromb Haemost ; 7(2): 347-54, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18983488

ABSTRACT

BACKGROUND: High-density lipoprotein (HDL) exerts a variety of anti-atherothrombotic functions, including a potent anti-inflammatory impact. In line, the direct pro-inflammatory effects of C-reactive protein (CRP) can be attenuated by HDL in vitro. OBJECTIVE: To evaluate whether this also holds true in humans, we assessed the ability of reconstituted HDL to neutralize CRP-mediated activation of coagulation and inflammation. METHODS: Fifteen healthy male volunteers received an infusion of recombinant human (rh)CRP (1.25 mg kg(-1) body weight). In eight of these volunteers, an infusion of human apoAI reconstituted with phosphatidylcholine (apoAI-PC; 80 mg kg(-1) body weight) preceded rhCRP infusion. RESULTS: Infusion of rhCRP alone elicited an inflammatory response and thrombin generation. In individuals who received apoAI-PC prior to rhCRP, these effects were abolished. Parallel tests in primary human endothelial cells showed that apoAI-PC preincubation with rhCRP abolished the CRP-mediated activation of inflammation as assessed by IL-6 release. Although we were able to show that rhCRP co-eluted with HDL after size-exclusion chromatography, plasmon surface resonance indicated the absence of a direct interaction between HDL and CRP. CONCLUSION: Infusion of apoAI-PC prior to rhCRP in humans completely prevents the direct atherothrombotic effects of rhCRP. These findings imply that administration of apoAI-PC may offer benefit in patients with increased CRP.


Subject(s)
Apolipoprotein A-I/pharmacology , C-Reactive Protein/pharmacology , Inflammation/prevention & control , Thrombosis/etiology , Adult , Apolipoprotein A-I/administration & dosage , Atherosclerosis , C-Reactive Protein/administration & dosage , Endothelial Cells , Humans , Inflammation/chemically induced , Male , Middle Aged , Phosphatidylcholines/administration & dosage , Recombinant Proteins
3.
Ann Oncol ; 13(5): 770-6, 2002 May.
Article in English | MEDLINE | ID: mdl-12075747

ABSTRACT

BACKGROUND: The novel molecule PI-88 is a highly sulfonated oligosaccharide which inhibits heparanase activity and competes with heparan sulfate binding of growth factors such as FGF and VEGF. Preclinical data demonstrates that PI-88 inhibits angiogenesis and has anti-metastatic effects. The aim of this phase I study was to determine the recommended dose and toxicity profile of PI-88. PATIENTS AND METHODS: PI-88 was given intravenously in increasing duration of administration (0.57 mg/kg for 2 h, 0.57 mg/kg/day for 1 day, 4, 7 and 14 consecutive days) and then increasing dose for 14 consecutive days (1.14 mg/kg/day and 2.28 mg/kg/day) in patients with advanced malignancies until dose-limiting toxicity (DLT) was observed. Fourteen assessable patients with advanced malignancies received PI-88 intravenously. RESULTS: DLT was thrombocytopenia. The thrombocytopenia appeared to be immunologically mediated with the development of anti-heparin platelet factor 4 complex antibodies. There were no other significant toxicities. At the final dose and schedule (2.28 mg/kg/day for 14 days), there was limited evidence of biological activity as measured by the surrogate marker activated partial thromboplastin time (APTT), although two patients had stabilisation of disease. CONCLUSIONS: In conclusion, PI-88 at a dose of 2.28 mg/kg/day for 14 days resulted in dose-limiting thrombocytopenia which appeared to be immune related. Limited evidence of biological activity was noted. Alternate scheduling and routes of administration are now being explored.


Subject(s)
Neoplasms/drug therapy , Oligosaccharides/administration & dosage , Oligosaccharides/adverse effects , Thrombocytopenia/chemically induced , Adult , Aged , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Incidence , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/mortality , Neoplasms/pathology , Risk Assessment , Survival Analysis , Thrombocytopenia/epidemiology , Treatment Outcome
5.
J Clin Oncol ; 19(2): 519-24, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11208846

ABSTRACT

PURPOSE: To determine the recommended dose, toxicity profile, and pharmacokinetics of a novel boronated porphyrin (BOPP) for photodynamic therapy (PDT) of intracranial tumors. PATIENTS AND METHODS: BOPP was administered alone in increasing doses (0.25, 0.5, 1.0, 2.0, 4.0, or 8.0 mg/kg) preoperatively in patients with intracranial tumors undergoing postresection PDT until dose-limiting toxicity (DLT) was observed. RESULTS: Twenty-nine assessable patients with intracranial tumors received BOPP intravenously 24 hours before surgery. The recommended dose was 4 mg/kg. Dose escalation was limited by thrombocytopenia. The most common nonhematologic toxicity was skin photosensitivity. Pharmacokinetic parameters showed increased area under the plasma concentration-time curve and maximum concentration with increased dose. Tumor BOPP concentrations also increased with increased dose. CONCLUSION: BOPP at a dose of 4 mg/kg was well tolerated. DLT was thrombocytopenia, and photosensitivity was the only other toxicity of note. The efficacy of PDT using BOPP requires further exploration.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Photochemotherapy , Protoporphyrins/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Adult , Aged , Area Under Curve , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Protoporphyrins/pharmacokinetics , Radiation-Sensitizing Agents/pharmacokinetics , Tissue Distribution
6.
Int J Hematol ; 74(4): 390-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11794693

ABSTRACT

A number of hematopoietic growth factors have been identified that are active on megakaryocytes and platelets, but only 2, interleukin-11 (IL-11) and thrombopoietin, are being actively pursued clinically, with IL-11 approved for treatment of thrombocytopenia. The development of these agents in general has been disappointing, and in part this reflects the inherent biology of these factors with a failure to match clinical need with physiological function. The delayed action of these factors is also a consequence of the intrinsic biology of megakaryocytes and platelets, and thus is likely to be limiting regardless of which factor is employed. In addition, the development of these agents has occurred at a time when there is something of a decreasing demand for platelets, at least in the context of chemotherapy-induced thrombocytopenia. This decrease is the result of increased use of blood stem cells to support intensive chemotherapy procedures, reduced thresholds for platelet transfusion, and a decreasing role for intensive chemotherapy. These issues are discussed.


Subject(s)
Growth Substances/physiology , Hematopoiesis/drug effects , Animals , Blood Platelets/cytology , Blood Platelets/drug effects , Growth Substances/pharmacology , Humans , Megakaryocytes/cytology , Megakaryocytes/drug effects , Thrombopoietin/pharmacology , Thrombopoietin/physiology
7.
Br J Haematol ; 109(4): 751-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10929025

ABSTRACT

Fifty-two patients with poor prognosis carcinoma of the breast underwent peripheral blood stem cell (PBSC) mobilization using five different regimens. The yields of primitive haemopoietic progenitors were quantified by a recently described pre-colony-forming unit (pre-CFU) assay using limiting dilution analysis (LDA). Results of days 14 and 35 pre-CFU were also correlated with conventional CD34+ cell enumeration, CFU-GM (granulocyte-macrophage) and long-term culture-initiating cell (LTCIC) assays. The yield of pre-CFUs with the combination of granulocyte colony-stimulating factor (G-CSF) and stem cell factor (SCF) was significantly higher than with G-CSF alone, cyclophosphamide (Cyclo) and granulocyte-monocyte colony-stimulating factor (GM-CSF), interleukin (IL)-3 and GM-CSF, or Cyclo alone. No significant correlation between neutrophil engraftment and pre-CFU could be demonstrated. Furthermore, CFU-GM was shown to bear a stronger correlation with pre-CFU and LTCIC than CD34+ cell measurement; thus, CFU-GM remains a useful biological tool for haemopoietic stem cell assay. We conclude that the combination of G-CSF and SCF mobilizes the highest number of pre-CFUs as measured by functional pre-CFU assay, which provides an alternative measurement of primitive haemopoietic progenitors to the LTCIC assay.


Subject(s)
Breast Neoplasms/therapy , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Stem Cell Factor/administration & dosage , Antigens, CD34 , Cells, Cultured , Colony-Forming Units Assay , Cyclophosphamide/administration & dosage , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppressive Agents/administration & dosage , Interleukin-3/administration & dosage , Interleukin-6/administration & dosage , Lymphocyte Count , Time Factors
8.
J Clin Oncol ; 18(15): 2852-61, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10920133

ABSTRACT

PURPOSE: To explore the influence of dose and schedule on the ability of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) to abrogate thrombocytopenia after multiple cycles of chemotherapy and to mobilize peripheral-blood progenitor cells (PBPC). PATIENTS AND METHODS: In this open-label study, 68 patients with advanced cancer were randomized to receive PEG-rHuMGDF subcutaneously at different doses and durations before administration of carboplatin 600 mg/m(2), cyclophosphamide 1,200 mg/m(2), and filgrastim 5 microgram/kg/d. PEG-rHuMGDF was not given after the first cycle of chemotherapy but was given after the second and subsequent cycles. Chemotherapy was given every 28 days for up to six cycles. RESULTS: In patients who received the same dose of chemotherapy for at least two cycles, the platelet nadir was significantly higher (47.5 x 10(9)/L v 35.5 x 10(9)/L; P =.003) and duration of grade 3 or 4 thrombocytopenia significantly shorter (0 v 3 days; P =.004) when PEG-rHuMGDF was administered after chemotherapy. There was no evidence of an effect of PEG-rHuMGDF when it was given before chemotherapy. Platelet recovery after the first cycle of chemotherapy was no different for different PEG-rHuMGDF regimens, and there was no difference between patients treated with PEG-rHuMGDF and historical controls treated with identical chemotherapy. There was a modest dose-related increase in progenitor cell levels after administration of PEG-rHuMGDF alone. Peak levels of PBPC occurred later in cycle 2 than in cycle 1 but were not different in magnitude. CONCLUSION: PEG-rHuMGDF abrogated severe thrombocytopenia after dose-intensive chemotherapy. However, it had only a modest effect on progenitor cell levels and did not enhance progenitor cell mobilization after chemotherapy and filgrastim.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Mobilization , Neoplasms/therapy , Polyethylene Glycols/pharmacology , Thrombocytopenia/drug therapy , Thrombopoietin/pharmacology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Cyclophosphamide/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Injections, Subcutaneous , Male , Middle Aged , Neoplasms/immunology , Polyethylene Glycols/therapeutic use , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombocytopenia/chemically induced , Thrombopoietin/therapeutic use
9.
Semin Hematol ; 37(2 Suppl 4): 19-27, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10831285

ABSTRACT

The search for a thrombopoietic agent has resulted in the identification of numerous cytokines and growth factors with thrombopoietic activity. However, with the exception of interleukin (IL)-11 and thrombopoietin (TPO), the megakaryopoietic activity of most of these molecules has not produced clearly identifiable clinical benefits. Despite the relatively modest effect of IL-11 on megakaryocyte and platelet production in vitro and in vivo, it does reduce the need for platelet transfusions in specialized clinical settings. In contrast, the c-Mpl ligand TPO has been shown to be a potent stimulator of megakaryocyte and platelet production both in vitro and in vivo. Clinical studies are being conducted with two different preparations of the c-Mpl ligand: recombinant human thrombopoietin (rhTPO) and pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF). A recombinant form of the complete human molecule, rhTPO is glycosylated and produced in mammalian cells. PEG-rHuMGDF consists of only the receptor-binding domain linked to a polyethylene glycol (PEG) moiety and is generated in Escherichia coil. Although c-Mpl ligands are still being evaluated, preliminary evidence indicates that these molecules can elevate platelet counts and may be useful in a range of clinical contexts. This report discusses aspects of the biology behind the clinical actions of IL-11 and the c-Mpl ligands.


Subject(s)
Blood Platelets/drug effects , Growth Substances/pharmacology , Megakaryocytes/drug effects , Animals , Blood Platelets/cytology , Cytokines/blood , Cytokines/pharmacology , Cytokines/physiology , Growth Substances/blood , Growth Substances/physiology , Hematopoiesis/drug effects , Humans , Interleukin-11/blood , Interleukin-11/pharmacology , Interleukin-11/physiology , Megakaryocytes/cytology , Thrombopoietin/blood , Thrombopoietin/pharmacology , Thrombopoietin/physiology
10.
Growth Factors ; 18(3): 215-26, 2000.
Article in English | MEDLINE | ID: mdl-11334057

ABSTRACT

Phase I studies with pegylated megakaryocyte growth and development factor (PEG-rHuMGDF), a c-Mpl ligand that stimulates megakaryopoiesis, have demonstrated that PEG-rHuMGDF is biologically active alone and causes a dose-related enhancement of platelet recovery when administered after chemotherapy. Here we report the dose-ranging pharmacokinetics of PEG-rHuMGDF. Pre-injection blood samples were drawn daily for pharmacokinetic studies on 43 patients. An ELISA, established using PEG-rHuMGDF as the standard, was able to quantitate Mpl ligand at concentrations > 0.02 ng/mL. Over the dose range 0.03 to 5.0 microg/kg/day, subcutaneous administration produced linear increases in steady-state serum levels. Maximum levels of PEG-rHuMGDF attained after 5.0 microg/kg/day were 5.88 to 10.9 ng/mL. After discontinuation of PEG-rHuMGDF, concentrations of Mpl ligand returned to baseline within 5 days. The pharmacokinetics were best described by a one-compartment model with first-order absorption, an absorption delay, and non linear clearance over the first 48 hours. The mean terminal half-life was 33.3 + 16.7 hours, and the average apparent at steady state was 27.7 + 14.0 mL/h/kg; both were independent of administered dose. The apparent clearance of PEG-rHuMGDF was not predicted by platelet count. Administration of chemotherapy and Filgrastim did not alter the pharmacokinetics of PEG-rHuMGDF.


Subject(s)
Polyethylene Glycols/pharmacokinetics , Recombinant Proteins/pharmacokinetics , Thrombopoietin/pharmacokinetics , Antineoplastic Agents/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay/methods , Humans , Injections, Subcutaneous , Neoplasms/blood , Neoplasms/drug therapy , Platelet Count , Polyethylene Glycols/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Thrombopoietin/administration & dosage , Thrombopoietin/blood
11.
J Clin Oncol ; 17(8): 2579-84, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10561325

ABSTRACT

PURPOSE: To assess the efficacy and toxicity of KRN8602 when administered as an intravenous bolus to patients with recurrent high-grade malignant glioma. PATIENTS AND METHODS: Patients with recurrent or persistent anaplastic astrocytoma or glioblastoma multiforme who had not received recent chemotherapy or radiotherapy and were of good performance status (Eastern Cooperative Oncology Group score < or = 2) were treated with an intravenous bolus of 40 mg/m(2) KRN8602 every 28 days. Tumor responses were assessed radiologically and clinically after every second cycle of therapy. Treatment was continued until documented progression or a total of six cycles. RESULTS: A median of three cycles (range, one to six cycles) of KRN8602 was administered to 55 patients, 49 of whom received at least two cycles and were, therefore, assessable for response. The overall response rate (disease stabilization or better) was 43% (95% confidence interval, 29% to 58%). There were three complete responses, one partial response, seven minor responses, and 10 patients with stable disease. The median time to progression was 2 months (range, 1.5 to 37 months) and overall survival was 11 months (range, 1.5 to 40 months). Neutropenia was the most common toxicity, although it was generally of brief duration, and there were only seven episodes of febrile neutropenia in 176 cycles delivered. Nonhematologic toxicity was mostly gastrointestinal (nausea and vomiting, diarrhea) and events were grade 2 or lower except for a single episode of grade 3 vomiting. CONCLUSION: KRN8602 is an active new agent with minimal toxicity in the treatment of relapsed or refractory high-grade glioma. Further studies with KRN8602 in combination with other cytotoxics and in adjuvant treatment of gliomas are warranted.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/therapeutic use , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Carubicin/analogs & derivatives , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Adolescent , Adult , Aged , Astrocytoma/mortality , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Carubicin/adverse effects , Carubicin/therapeutic use , Combined Modality Therapy , Female , Glioblastoma/mortality , Humans , Male , Middle Aged , Neutropenia/chemically induced , Survival Analysis
12.
J Clin Oncol ; 17(1): 82-92, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10458221

ABSTRACT

PURPOSE: To determine the safety and efficacy of multiple cycles of dose-intensive, nonablative chemotherapy in women with poor-prognosis breast cancer. PATIENTS AND METHODS: Women with stage II breast cancer and 10 or more involved nodes or four or more involved nodes and estrogen receptor-negative tumors and women with stage III disease received three cycles of epirubicin 200 mg/m2 and cyclophosphamide 4 g/m2, with progenitor cell and filgrastim support every 28 days (n = 79) or 21 days (n = 20). Patients were reviewed at least twice yearly thereafter. Twenty-six patients had bone marrow and apheresis collections assessed for the presence of micrometastatic tumor cells. RESULTS: Ninety-nine women (median age, 43 years; range, 24 to 60 years) were treated. Ninety-two completed all three cycles of chemotherapy. The major toxicity was severe, reversible myelosuppression that was more prolonged with successive cycles, and this did not differ between patients given treatment every 28 days and those treated every 21 days. Febrile neutropenia occurred in 176 (61%) of 287 cycles. Severe mucositis (grade 3 or 4) occurred in 23% of cycles but tended to be short-lived and was reversible. The cardiac ejection fraction fell by a median of 4% during treatment, and three patients developed evidence of cardiac failure after chemotherapy. Two patients (2%) died of acute toxicity. Three of 26 patients had evidence of circulating micrometastatic tumor cells. The actuarial distant disease-free and overall survival rates at 60-month follow-up were 64% (95% confidence interval [CI], 53% to 75%) and 67% (95% CI, 56% to 78%), respectively. CONCLUSION: Multiple cycles of dose-intensive, nonablative chemotherapy is a feasible and safe approach. Disease control and survival are similar to those in other studies of myeloablative chemotherapy in poor-prognosis breast cancer. The regimen is being evaluated in a randomized trial of the International Breast Cancer Study Group.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Epirubicin/administration & dosage , Epirubicin/adverse effects , Female , Filgrastim , Follow-Up Studies , Humans , Middle Aged , Neoplastic Cells, Circulating , Prognosis , Recombinant Proteins , Survival Rate
13.
Br J Haematol ; 104(4): 778-84, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10192440

ABSTRACT

Mobilization of haemopoietic precursor cells into the circulation by the combination of cytokines, stem cell factor (SCF) and G-CSF in previously untreated patients with carcinoma of the breast resulted in increased yield of collected peripheral blood precursor cells (PBPC). This mobilization of PBPC by SCF with G-CSF lasted several days after ceasing the cytokines in comparison to the rapid fall of PBPC after ceasing G-CSF. Possible mechanisms for this increased and prolonged mobilization were investigated. Immunological phenotyping with CD38, Thy-1 and MDR-1 of the CD34-positive mobilized PBPC detected no difference in maturity compared to PBPC mobilized by G-CSF alone. However, the down-regulation of c-kit, which is associated with the mechanism of mobilization, was much greater in the PBPC mobilized by SCF and G-CSF. The potential clinical implication of increased and prolonged mobilization is increased yield, allowing transplantation of heavily pre-treated patients, transplantation with PBPC from a single apheresis, or PBSC support for multiple courses of high-dose therapy from one mobilization procedure.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/therapeutic use , Antigens, CD34 , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cohort Studies , Drug Combinations , Female , Flow Cytometry , Humans , Immunophenotyping , Leukapheresis/methods , Phenotype
14.
Ann Oncol ; 10(1): 53-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10076722

ABSTRACT

PURPOSE: To prospectively evaluate the long term cardiac effects of high-dose epirubicin and cyclophosphamide given to women with early stage, poor prognosis breast cancer. PATIENTS AND METHODS: Women with stage 2 breast cancer and 10+ nodes or 4+ nodes and estrogen receptor negative tumor, or stage 3 breast cancer received three cycles of epirubicin 200 mg/m2 and cyclophosphamide 4 gm/m2 with peripheral blood progenitor cell and filgrastim support. Treatment was given every 28 days (n = 79) or 21 days (n = 20). Fifty patients received radiotherapy to the chest wall or breast, 25 of to the left side. Patients were assessed clinically regularly during chemotherapy and at least three times yearly after completion of treatment. Cardiac left ventricular ejection fraction (LVEF) was assessed by radionuclide scan before therapy, after each cycle of chemotherapy, three months and six months after completion of chemotherapy, and yearly thereafter until relapse. RESULTS: Ninety-nine women were treated, and 92 completed all three cycles of chemotherapy. The median age was 43 years (range 24 to 60 years). All patients were included in this analysis. The median relapse-free survival was 39 months (11 to 68 months). There was a significant fall in LVEF during chemotherapy. In general, there was no further deterioration in cardiac function from the third month after cessation of treatment, however there was substantial variation between individuals. 35 patients had at least one LVEF measure less than normal (< 50%), but the LVEF returned to normal in 20 of these with further follow-up. Cardiac dysfunction was not increased in women who received radiotherapy and was not different between cohorts given chemotherapy every three or every four weeks. One patient died of acute myocardial necrosis following the third cycle of chemotherapy. Two patients developed clinical evidence of cardiac failure, and another had radiological signs but was asymptomatic. One woman died of progressive cardiac failure, one recovered clinically but also developed recurrent breast cancer, while the third recovered after commencement of medical therapy. CONCLUSIONS: During follow-up after high-dose epirubicin and cyclophosphamide as delivered in this study, the LVEF fell to below normal in approximately one third of patients. However, in over half of these patients the LVEF subsequently recovered to the normal range, and the incidence of clinically evident chronic cardiac failure was low. Further follow-up is required to assess the long-term safety. A randomized comparison with standard-dose anthracycline-based chemotherapy is needed to determine whether this regimen is associated with an increased risk of clinical cardiac toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Heart Failure/chemically induced , Stroke Volume/drug effects , Adolescent , Adult , Antibiotics, Antineoplastic/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cyclophosphamide/administration & dosage , Dose-Response Relationship, Drug , Epirubicin/administration & dosage , Female , Humans , Middle Aged , Neoplasm Staging , Prognosis , Prospective Studies , Risk Assessment , Severity of Illness Index , Survival Rate
15.
BioDrugs ; 11(4): 261-76, 1999 Apr.
Article in English | MEDLINE | ID: mdl-18031136

ABSTRACT

Thrombopoietin (TPO), the central regulator of megakaryocytopoiesis, was first identified in 1994 and since then much has been learnt about megakaryocyte and platelet physiology. Two forms of TPO have entered clinical trials, the full length recombinant version (rhTPO) and a truncated form (megakaryocyte growth and development factor, MGDF). Both have equivalent biological activity in preclinical studies and both have been demonstrated to have potent biological activity in humans. TPO and MGDF administration cause a dose-dependent increase in platelet count with no effect on white cell count or haematocrit. These platelets are morphologically and functionally normal. When administered following myelosuppressive chemotherapy, MGDF and TPO significantly enhance platelet recovery, although scheduling in relation to chemotherapy may be important in optimising the full effects. TPO and MGDF mobilise progenitor cells of multiple haematopoietic lineages and may enhance the effects of filgrastim on peripheral blood progenitor cell levels after chemotherapy. Both forms of TPO are well tolerated and cause few acute toxicities. Numerous studies are underway to help determine the precise role of TPO in clinical practice.

16.
Aust N Z J Med ; 28(2): 190-6, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9612527

ABSTRACT

BACKGROUND: Much research has been conducted into the pathobiology, diagnosis, and management of acute myeloid leukaemia (AML) since 1980, with major contributions from Australian studies in this period. AIMS: To determine whether advances in basic and clinical research into AML have translated into improved survival for patients in the community. METHODS: A retrospective survey of records of all patients with AML presenting to the Royal Melbourne Hospital (RMH) over a 16 year period, analysed according to induction therapy and established prognostic factors. Between 1980 and December 1996 223 (98%) of 227 patients were evaluable. RESULTS: The probability of survival at five years for patients treated since 1990 has improved significantly compared to the cohort treated between 1980-89 (34 +/- 5% vs 4 +/- 2%; mean +/- standard error). This benefit is most evident in patients less than 60 years of age (50 +/- 7% vs 11 +/- 4%). Successive induction protocols in the context of clinical trials conducted since 1985 contributed to improved outcomes. The selective application of bone marrow transplantation, and use of retinoic acid as induction therapy for acute promyelocytic leukaemia has also improved survival. Despite increases in dose-intensity, early death rates for patients undergoing induction therapy fell during the study period. CONCLUSIONS: Participation in clinical and basic research with the development of more intense and specific treatments for patients with AML has contributed to better outcomes, underpinned by improvements in supportive care.


Subject(s)
Leukemia, Myeloid/mortality , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Australia/epidemiology , Bone Marrow Transplantation , Female , Humans , Leukemia, Myeloid/therapy , Male , Middle Aged , Prognosis , Remission Induction , Retrospective Studies , Survival Analysis
17.
J Clin Oncol ; 16(6): 2181-7, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626219

ABSTRACT

PURPOSE: To determine the recommended dose, toxicity profile, and pharmacokinetics of KRN8602 (MX2-hydrochloride), a novel morpholino anthracycline with potent cytotoxicity against anthracycline-sensitive and resistant experimental tumors in vitro and in vivo. PATIENTS AND METHODS: KRN8602 was administered alone in increasing doses to patients with advanced cancer or high-grade gliomas until dose-limiting toxicity (DLT) was observed in three or more of five patients treated in a dose level. Because neutropenia was dose limiting, further escalation was investigated with filgrastim support. RESULTS: Fifty-six assessable patients completed at least one cycle of chemotherapy. The recommended dose of KRN8602 alone was 40 mg/m2. Dose escalation was limited by neutropenia. The recommended dose of KRN8602 with filgrastim was 70 mg/m2, and limiting toxicities were neutropenia, diarrhea, and vomiting. The most commonly experienced nonhematologic toxicity was nausea and vomiting. Alopecia and mucositis were infrequent and mild. Pharmacokinetic parameters showed substantial variation, although the area under the plasma concentration-time curve (AUC) and maximum concentration both increased with dose. There was no relationship between pharmacokinetic parameters and toxicity. CONCLUSION: KRN8602 at doses of 40 mg/m2 when administered alone and 70 mg/m2 when administered with filgrastim appeared to be manageable. The major DLTs were neutropenia and, at higher doses, diarrhea and vomiting. The efficacy of this drug is currently being tested in phase II studies.


Subject(s)
Carubicin/analogs & derivatives , Granulocyte Colony-Stimulating Factor/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Carubicin/administration & dosage , Carubicin/adverse effects , Carubicin/pharmacokinetics , Dose-Response Relationship, Drug , Female , Filgrastim , Humans , Male , Middle Aged , Neutropenia/chemically induced , Recombinant Proteins , Treatment Outcome
18.
J Clin Oncol ; 16(5): 1899-908, 1998 May.
Article in English | MEDLINE | ID: mdl-9586908

ABSTRACT

PURPOSE: To assess the mobilization potential and safety of recombinant human stem-cell factor (SCF) when coadministered with filgrastim to untreated women with poor-prognosis breast cancer. PATIENTS AND METHODS: Eligible women had breast cancer with 10 or more positive axillary nodes, or estrogen receptor-negative tumor with 4 positive nodes, or stage III disease. Patients were randomized to receive SCF plus filgrastim or filgrastim alone. Filgrastim 12 microg/kg daily was administered for 6 days by continuous subcutaneous infusion. SCF was administered by daily subcutaneous injection at 5, 10, or 15 microg/kg concurrent with filgrastim for 7 days, or 10 microg/kg daily starting 3 days before filgrastim for a total of 10 days (SCF pretreatment). Apheresis was performed on days 5, 6, and 7 of filgrastim administration. Patients then had three cycles of epirubicin 200 mg/m2 and cyclophosphamide 4 g/m2 every 28 days, each supported by one third of the apheresis product. RESULTS: Sixty-two women were treated. Greater yields occurred in patients who received SCF 10 microg/kg daily plus filgastim than those who received filgrastim alone (P=.013 for CD34+ cells; P=.07 for granulocyte-macrophage colony-forming cells [GM-CFCs]). The difference was more marked with SCF-pretreatment than concurrent SCF. Fewer aphereses were required to reach the predetermined target of peripheral-blood progenitor/stem cells (PBPCs) in women who received SCF. SCF was generally well tolerated. Hematologic recovery was rapid after each of the three cycles of chemotherapy. There was no difference in recovery between the different treatment groups. CONCLUSION: Mobilization of PBPCs by filgrastim is significantly enhanced by coadministration of SCF, and commencing SCF before filgrastim can optimize this effect. SCF has the potential to reduce the number of aphereses required to collect a target number of PBPCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/drug therapy , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoiesis , Hematopoietic Stem Cell Mobilization , Stem Cell Factor/administration & dosage , Adult , Antigens, CD34/analysis , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Blood Cell Count , Blood Component Removal , Breast Neoplasms/blood , Colony-Forming Units Assay , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Epirubicin/administration & dosage , Epirubicin/adverse effects , Erythroid Precursor Cells , Female , Filgrastim , Hematopoiesis/drug effects , Hematopoietic Stem Cell Mobilization/methods , Hemoglobins/analysis , Humans , Lymphocyte Subsets , Middle Aged , Recombinant Proteins/administration & dosage , Stem Cell Factor/adverse effects
19.
Diagn Cytopathol ; 18(3): 204-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9523139

ABSTRACT

A 43-yr-old woman with recently diagnosed breast carcinoma presented with a right pleural effusion after a cycle of adjuvant, high-dose chemotherapy supported by peripheral blood progenitor cells (PBPC) and granulocyte-colony-stimulating factor (G-CSF, Filgrastim). Cytologic examination of the pleural aspirate yielded highly cellular material composed predominantly of cells of myeloid and macrophage/monocytic lineages. Despite clinical concern of a malignant effusion, the combination of cytologic and immunophenotypic examination yielded the correct diagnosis of a nonneoplastic effusion related to underlying pleural inflammation and possibly the administration of G-CSF.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/therapy , Granulocyte Colony-Stimulating Factor/adverse effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Pleural Effusion/chemically induced , Adult , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Cyclophosphamide/administration & dosage , Cytodiagnosis , Epirubicin/administration & dosage , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cells/pathology , Humans , Immunohistochemistry , Macrophages/immunology , Macrophages/pathology , Monocytes/immunology , Monocytes/pathology , Neoplasm Staging , Pleural Effusion/pathology , Recombinant Proteins
20.
Cancer Chemother Pharmacol ; 41(5): 423-6, 1998.
Article in English | MEDLINE | ID: mdl-9523740

ABSTRACT

PURPOSE: To investigate the effect of granulocyte colony-stimulating factor (G-CSF) on the pharmacokinetics and pharmacodynamics of the new morpholino anthracycline drug MX2. METHODS: A total of 25 patients with advanced malignant disease participated in a dose-escalation study in the first cycle of treatment given i.v. at doses of 50-80 mg/m2 (74-152 mg) with concomitant filgrastim (G-CSF, 5 microg/kg) given daily beginning at 24 h after the dose of MX2. RESULTS: The mean fast distribution half-life (1.5 +/- 1.0 min) and the mean plasma clearance (2.18 +/- 0.95 l/min) were significantly lower than the respective mean values found in a previous study in which 27 patients had received MX2 (16.8-107.5 mg) alone (3.3 +/- 2.2 min and 2.98 +/- 1.68 l/min, respectively; P < 0.05). There was no correlation between plasma clearance and the delivered dose for the combined MX2-alone and MX2-filgrastim groups, indicating that the lower clearance observed in the G-CSF group was probably not due to the higher dose. Elimination half-lives of the metabolites M1 and M4 were significantly greater in the filgrastim group (19.8 +/- 14.7 and 11.8 +/- 5.0 h for M1 and 14.8 +/- 4.1 and 12.3 +/- 6.3 h for M2, respectively). Unlike the MX2-alone group, there was no relationship in the MX2-filgrastim group between the relative nadir neutrophil count and the dose or between the duration of grade IV neutropenia and the dose of MX2. CONCLUSIONS: This study shows that filgrastim decreased the plasma clearance of MX2 by approximately 25%, possibly by inhibition of metabolism.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Carubicin/analogs & derivatives , Granulocyte Colony-Stimulating Factor/pharmacology , Neoplasms/metabolism , Antibiotics, Antineoplastic/administration & dosage , Area Under Curve , Carubicin/administration & dosage , Carubicin/pharmacokinetics , Dose-Response Relationship, Drug , Filgrastim , Humans , Metabolic Clearance Rate/drug effects , Neoplasms/drug therapy , Neutrophils/drug effects , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...