Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Toxicol Appl Pharmacol ; 239(3): 241-50, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19523970

ABSTRACT

Cyclosporin A (CsA) has nephrotoxic effects known to involve reactive oxygen species (ROS), since antioxidants prevent the kidney damage induced by this drug. Given that mitochondria are among the main sources of intracellular ROS, the aims of our study were to examine the mitochondrial effects of CsA in the porcine renal endothelial cell line LLC-PK1 and the influence of the antioxidant Vitamin E (Vit E). Following the treatment of LLC-PK1 cells with CsA, we assessed the mitochondrial synthesis of superoxide anion, permeability transition pore opening, mitochondrial membrane potential, cardiolipin peroxidation, cytochrome c release and cellular apoptosis, using flow cytometry and confocal microscopy procedures. Similar experiments were done after Vit E preincubation of cells. CsA treatment increased superoxide anion in a dose-dependent way. CsA opened the permeability transition pores, caused Bax migration to mitochondria, and decreased mitochondrial membrane potential and cardiolipin content. Also CsA released cytochrome c into cytosol and provoked cellular apoptosis. Vit E pretreatment inhibited the effects that CsA induced on mitochondrial structure and function in LLC-PK1 cells and avoided apoptosis. CsA modifies mitochondrial LLC-PK1 cell physiology with loss of negative electrochemical gradient across the inner mitochondrial membrane and increased lipid peroxidation. These features are related to apoptosis and can explain the cellular damage that CsA induces. As Vit E inhibited these effects, our results suggest that they were mediated by an increase in ROS production by mitochondria.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Cyclosporine/toxicity , Endothelial Cells/drug effects , Mitochondria/drug effects , Vitamin E/pharmacology , Animals , Blotting, Western , Cardiolipins/metabolism , Caspase 6/metabolism , Cell Culture Techniques , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/metabolism , Endothelial Cells/metabolism , Flow Cytometry , LLC-PK1 Cells , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Swine , bcl-2-Associated X Protein/metabolism
2.
Gynecol Oncol ; 110(3 Suppl 2): S45-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18678399

ABSTRACT

Locally advanced cervical carcinoma had been treated with radiation therapy until 1999, when five different large clinical trials showed an overall survival benefit when chemotherapy was administered concomitantly with radiotherapy. The chemotherapy agents used in these trials were cisplatin, cisplatin combined with fluorouracil or hydroxyurea. Weekly cisplatin (40 mg/m(2)) achieved the best responses, even when compared with the combination with fluorouracil. These results led the United States National Cancer Institute (NCI) to recommend platinum-based chemotherapy for the treatment of locally advanced cervical carcinoma. Other cytotoxic agents have been tried in combination with radiotherapy for the management of the disease, including carboplatin, paclitaxel, gemcitabine and even topotecan. Gemcitabine has shown promising results and the combination of paclitaxel and carboplatin has proved safe and effective. However, to date, there has been no agent or combination of agents to have shown superiority over weekly cisplatin. Biologic agents such as bevacizumab, cetuximab, sorafenib and erlotinib are currently being tried in different trials in combination with radiotherapy and cisplatin. Celecoxib, a COX-2 inhibitor was evaluated in an RTOG study in combination with cisplatin and flourouracil with radiation therapy with no apparent effect on DFS and poor rates of locoregional control. Chemoradiation is the current standard therapy in locally advanced cervical carcinoma. The integration of novel agents will be established by the ongoing clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/radiotherapy , Combined Modality Therapy , Female , Humans
3.
Nefrologia ; 27(5): 565-73, 2007.
Article in Spanish | MEDLINE | ID: mdl-18045032

ABSTRACT

Reactive oxygen species (ROS) have been implicated in cyclosporin A (CsA) nephrotoxicity. As mitochondria are one of the main sources of ROS in cells, we evaluated the role of CsA in mitochondrial structure and function in LLC-PK1 cells. We incubated cells with CsA 1 microM for 24 hours and studies were performed with flow citometry and confocal microscopy. We studied mitochondrial NAD(P)H content, superoxide anion (O2.-) production (MitoSOX Red), oxidation of cardiolipin of inner mitochondrial membrane (NAO) and mitochondrial membrane potential (DIOC2(3)). Also we analyzed the intracellular ROS synthesis (H2DCF-DA) and reduced glutation (GSH) of cells. Our results showed that CsA decreased NAD(P)H and membrane potential, and increased O2.- in mitochondria. CsA also provoked oxidation of cardiolipin. Furthermore, CsA increased intracellular ROS production and decreased GSH content. These results suggest that CsA has crucial effects in mitochondria. CsA modified mitochondrial physiology through the decrease of antioxidant mitochondrial compounds as NAD(P)H and the dissipation of mitochondrial membrane potential and increase of oxidants as O2.-. Also, CsA alters lipidic structure of inner mitochondrial membrane through the oxidation of cardiolipin. These effects trigger a chain of events that favour intracellular synthesis of ROS and depletion of GSH that can compromise cellular viability. Nephrotoxic cellular effects of CsA can be explained, at least in part, through its influence on mitochondrial functionalism.


Subject(s)
Cyclosporine/adverse effects , Kidney Tubules/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Animals , Cells, Cultured , Kidney Tubules/metabolism , Kidney Tubules/ultrastructure , Swine
4.
Nefrología (Madr.) ; 27(5): 565-573, sept.-oct. 2007. ilus
Article in Es | IBECS | ID: ibc-057269

ABSTRACT

Estudiamos el efecto de la ciclosporina A (CsA) sobre la estructura y función mitocondrial en células LLC-PK1. Las células se incubaron durante 24 horas con CsA 1 mM y se analizó la producción de anión superóxido, contenido de NAD(P)H, oxidación de cardiolipina y potencial de membrana mitocondrial; además se estudió la formación de radicales libres y el contenido de glutatión reducido intracelular. Nuestros resultados demuestran que la CsA provocó un aumento del anión superóxido mitocondrial de modo paralelo al descenso de NAD(P)H; además, se produjo oxidación de la cardiolipina de la membrana interna y un descenso del potencial de membrana mitocondrial. Finalmente, observamos un aumento de la producción de radicales libres intracelulares y un descenso del glutatión reducido. En conclusión, la CsA produce modificaciones importantes en la fisiología y estructura mitocondrial con aumento de la síntesis de especies reactivas de oxígeno y descenso de la capacidad antioxidante, hechos que podrían justificar la toxicidad celular de la droga


Reactive oxygen species (ROS) have been implicated in cyclosporin A (CsA) nephrotoxicity. As mitochondria are one of the main sources of ROS in cells, we evaluated the role of CsA in mitochondrial structure and function in LLC-PK1 cells. We incubated cells with CsA 1 mM for 24 hours and studies were performed by flow cytometry and confocal microscopy.We studied mitochondrial NAD(P)H content, superoxide anion (O2.-) production (MitoSOX Red), oxidation of cardiolipin of inner mitochondrial membrane (NAO) and mitochondrial membrane potential [DIOC2(3)]. We also analyzed the intracellular ROS synthesis (H2DCF-DA) and reduced glutation (GSH) of cells. Our results showed that CsA decreased NAD(P)H and membrane potential, and increased O2.- in mitochondria. CsA also provoked oxidation of cardiolipin. Furthermore, CsA increased intracellular ROS production and decreased GSH content. These results suggest that CsA has crucial effects in mitochondria. CsA modified mitochondrial physiology through the decrease of antioxidant mitochondrial compounds as NAD(P)H and the dissipation of mitochondrial membrane potential and increase of oxidants such O2.-. Also, CsA alters lipidic structure of inner mitochondrial membrane through the oxidation of cardiolipin. These effects trigger a chain of events that favour intracellular synthesis of ROS and depletion of GSH that can compromise cellular viability. Nephrotoxic cellular effects of CsA can be explained, at least in part, through its influence on mitochondrial functionalism


Subject(s)
Cyclosporine/adverse effects , Mitochondria , Kidney Tubules , Oxidative Stress , Free Radicals/analysis , Cardiolipins/analysis , Superoxides/analysis , Reactive Oxygen Species/analysis , NADP/analysis , LLC-PK1 Cells
5.
Clin Transl Oncol ; 8(12): 889-95, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17169762

ABSTRACT

OBJECTIVE: This randomized clinical trial evaluated the efficacy and safety of monotherapy with cefepime for patients with solid tumors treated with high dose chemotherapy (HDC) and peripheral blood stem cell support (PBSCS) with febrile neutropenia. SUBJECTS: Patients with solid tumors treated with HDC and PBSCS, that developed fever and neutropenia (absolute neutrophil count < 500 cells/microL) were eligible, and randomly assigned to receive ceftazidime plus amikacin or cefepime. RESULTS: Fifty-one episodes were randomized, and all were evaluable (27 received ceftazidime plus amikacin arm, and 24 cefepime). Major efficacy endpoints did not show significant differences, with success rates of 44.4% and 54.2% (p = 0.481) for the combination arm and the monotherapy arm, respectively. The proportion of patients that became afebrile in the first 24 hours was significantly higher in the cefepime group (41.7% vs 11.1%, respectively; p = 0.012). However, due to its premature closure and small sample size, this study lacks the adequate power to definitely address this question. CONCLUSIONS: Cefepime monotherapy appeared to have an equivalent efficacy and safety as empiric treatment in febrile neutropenia episodes in a highrisk population compared with ceftazidime and amikacin. Nevertheless, this study is not adequately powered to answer this question. Given the small number of patients randomized and the single-center nature of this study, these results must be cautiously interpreted.


Subject(s)
Amikacin/administration & dosage , Anti-Bacterial Agents/therapeutic use , Ceftazidime/administration & dosage , Cephalosporins/therapeutic use , Neoplasms/drug therapy , Neutropenia/drug therapy , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cefepime , Drug Therapy, Combination , Female , Fever/drug therapy , Humans , Male , Middle Aged , Neutropenia/chemically induced , Stem Cell Transplantation , Treatment Outcome
6.
Clin Transl Oncol ; 8(12): 896-902, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17169763

ABSTRACT

Purpose. To assess the toxicity and efficacy of biweekly gemcitabine plus vinorelbine in first-line advanced breast cancer, and to establish whether circulating HER2 ECD levels correlate with the efficacy of the combination. Patients and methods. 52 patients were treated with gemcitabine 2500 mg/m(2) plus vinorelbine 30 mg/m(2), both on day 1 of 14-day cycles, for a maximum of 10 cycles. Baseline serum levels of HER2 ECD were assessed with an ELISA. Results. All patients were evaluable for toxicity, and 50 for efficacy. Overall toxicity was moderate. Grade 3 neutropenia occurred in 35% of patients and grade 4 in 19%. Other grade 3 toxicities were observed in less than 6%. There was one episode of febrile neutropenia, and one death after cycle three. Overall response rate was 52% (95% CI: 38% to 66%), with 2 patients achieving a CR (4%). Response rate did not correlate with HER2 ECD, with 50% of HER2 ECD positive patients responding, vs 48.5% of the HER2 ECD negative. Median overall survival was 24.6 months. Conclusion. Gemcitabine plus vinorelbine, given as an every-two-week schedule, is an active regimen in advanced breast carcinoma. This combination can be an option when anthracyclines and taxanes are not preferred. HER2 ECD has no predictive value in this non-taxane combination.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Receptor, ErbB-2/blood , Adenocarcinoma/blood , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Breast Neoplasms/blood , Breast Neoplasms/mortality , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Enzyme-Linked Immunosorbent Assay , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinorelbine , Gemcitabine
7.
Clin. transl. oncol. (Print) ; 8(12): 889-895, dic. 2006. tab
Article in English | IBECS | ID: ibc-126348

ABSTRACT

OBJECTIVE: This randomized clinical trial evaluated the efficacy and safety of monotherapy with cefepime for patients with solid tumors treated with high dose chemotherapy (HDC) and peripheral blood stem cell support (PBSCS) with febrile neutropenia. SUBJECTS: Patients with solid tumors treated with HDC and PBSCS, that developed fever and neutropenia (absolute neutrophil count < 500 cells/microL) were eligible, and randomly assigned to receive ceftazidime plus amikacin or cefepime. RESULTS: Fifty-one episodes were randomized, and all were evaluable (27 received ceftazidime plus amikacin arm, and 24 cefepime). Major efficacy endpoints did not show significant differences, with success rates of 44.4% and 54.2% (p = 0.481) for the combination arm and the monotherapy arm, respectively. The proportion of patients that became afebrile in the first 24 hours was significantly higher in the cefepime group (41.7% vs 11.1%, respectively; p = 0.012). However, due to its premature closure and small sample size, this study lacks the adequate power to definitely address this question. CONCLUSIONS: Cefepime monotherapy appeared to have an equivalent efficacy and safety as empiric treatment in febrile neutropenia episodes in a highrisk population compared with ceftazidime and amikacin. Nevertheless, this study is not adequately powered to answer this question. Given the small number of patients randomized and the single-center nature of this study, these results must be cautiously interpreted (AU)


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Anti-Bacterial Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cephalosporins/therapeutic use , Neoplasms/drug therapy , Neutropenia/chemically induced , Neutropenia/drug therapy , Ceftazidime/administration & dosage , Amikacin/administration & dosage , Treatment Outcome , Stem Cell Transplantation , Fever/drug therapy
8.
Clin. transl. oncol. (Print) ; 8(12): 896-902, dic. 2006. tab, ilus
Article in English | IBECS | ID: ibc-126349

ABSTRACT

Purpose. To assess the toxicity and efficacy of biweekly gemcitabine plus vinorelbine in first-line advanced breast cancer, and to establish whether circulating HER2 ECD levels correlate with the efficacy of the combination. Patients and methods. 52 patients were treated with gemcitabine 2500 mg/m(2) plus vinorelbine 30 mg/m(2), both on day 1 of 14-day cycles, for a maximum of 10 cycles. Baseline serum levels of HER2 ECD were assessed with an ELISA. Results. All patients were evaluable for toxicity, and 50 for efficacy. Overall toxicity was moderate. Grade 3 neutropenia occurred in 35% of patients and grade 4 in 19%. Other grade 3 toxicities were observed in less than 6%. There was one episode of febrile neutropenia, and one death after cycle three. Overall response rate was 52% (95% CI: 38% to 66%), with 2 patients achieving a CR (4%). Response rate did not correlate with HER2 ECD, with 50% of HER2 ECD positive patients responding, vs 48.5% of the HER2 ECD negative. Median overall survival was 24.6 months. Conclusion. Gemcitabine plus vinorelbine, given as an every-two-week schedule, is an active regimen in advanced breast carcinoma. This combination can be an option when anthracyclines and taxanes are not preferred. HER2 ECD has no predictive value in this non-taxane combination (AU)


Subject(s)
Humans , Female , Adult , Middle Aged , Aged , Aged, 80 and over , Adenocarcinoma/blood , Adenocarcinoma/drug therapy , Adenocarcinoma/mortality , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Enzyme-Linked Immunosorbent Assay , Kaplan-Meier Estimate , /blood , Vinblastine/analogs & derivatives , Vinblastine/administration & dosage , Vinblastine/adverse effects
9.
Cytometry A ; 69(3): 161-4, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16479613

ABSTRACT

BACKGROUND: Cyclosporine A increases oxidative stress in kidney and we hypothesized that cyclooxygenase (COX) may be involved in this effect. MATERIAL AND METHODS: Mesangial cells of Cyclosporine A-treated (4, 7 or 10 days) rats were obtained to evaluate mRNA expression of COX-isoforms (COX-1, constitutive and COX-2, inducible) by "in situ" hybridization. Probes were labelled using "Gene Image Random Prime Labelling Protocol" and COX expression was measured by flow cytometry. RESULTS AND DISCUSSION: "In situ" hybridization by flow cytometry is an useful method to detect mRNA. We observed an increased COX-2 expression in a time-dependent manner in parallel with Reactive Oxygen Species synthesis. COX-1 expression increased only at 10 days.


Subject(s)
Cyclosporine/pharmacology , Flow Cytometry/methods , In Situ Hybridization, Fluorescence/methods , Mesangial Cells/drug effects , Prostaglandin-Endoperoxide Synthases/genetics , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Gene Expression/drug effects , Gene Expression/genetics , Immunosuppressive Agents/pharmacology , Isoenzymes/genetics , Male , Mesangial Cells/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
11.
Nefrologia ; 25(2): 131-6, 138, 140, 2005.
Article in Spanish | MEDLINE | ID: mdl-15912649

ABSTRACT

All-trans retinoic acid (AR-t) is used for treating acute promyelocytic leukemia and renal cell carcinoma and it also has therapeutic value in several animal models of renal disease. Among its renal targets, mesangial cells have been widely studied: they have both retinoic acid receptors (RAR) and retinoid X receptors (RXR) and the cell growth is inhibited when human mesangial cells are incubated with 1-10 microM AR-t. Although his effect has been related with the antiproliferative action of AR-t, there are no studies on the involvement of apoptosis in AR-t induced cell growth when higher concentrations of retinoid are used. Our studies show that 25 microM AR-t triggers mesangial cell apoptosis assessed by light and fluorescence microscopy (Giemsa stain and acridine orange stain, respectively), DNA electrophoresis, flow cytometry (annexin-V) and immunocytochemistry (TUNEL). AR-t induced apoptosis was not inhibited by preincubation with the RXR pan-antagonist HX531 nor with the RAR pan-antagonist AGN 193109, this suggesting RAR and RXIR are not involved in AR-t induced cell death. Previous results of our group showed that ERK (extracellular regulated kinase) and INK (c-Jun kinase), two members of the MAP (mitogen activated protein) kinase family, are involved in non apoptotic effects of AR-t on mesangial cells. Therefore we focussed on the stress activated p38 kinase, the third member of the MAPK family, to investigate its involvement in AR-t induced apoptosis. The results confirmed a role of p38 since: 1) preincubation with B5203589, a p38 inhibitor, inhibited ARA induced apoptosis; 2) incubation with AR-t induced p38 phosphorilation after few minutes and p38 remained phosphorilated for at least 8 hours and 3) AR-t induced p38 phosphorilation was inhibited by SB203589. These data suggest that AR-t might have toxic side effects on the kidney but also suggest that AR-t could be an useful inhibitor of pathological mesangial cell expansion.


Subject(s)
Apoptosis/drug effects , Glomerular Mesangium/cytology , Tretinoin/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology , Cells, Cultured , Humans
12.
Nefrología (Madr.) ; 25(2): 131-140, mar. 2005. ilus, graf
Article in Es | IBECS | ID: ibc-042540

ABSTRACT

El ácido retinoico todo-trans (AR-t) se utiliza en clínica en el tratamiento de la leucemiapromielocítica aguda y el cáncer renal. También presenta efecto terapéutico endiversas formas de enfermedad renal experimental. Las células mesangiales son una delas dianas farmacológicas de AR-t mejor estudiadas: presentan receptores de ácido retinoico(RAR) y receptores X de retinoides (RXR) y el AR-t, a concentraciones entre 1 y 10µM, inhibe su crecimiento. Este efecto se ha relacionado con la acción antiproliferativadel AR-t, aunque no se ha estudiado la participación de mecanismos apoptóticos cuandose utilizan mayores concentraciones de AR-t. El presente trabajo demuestra que AR-t25 µM induce apoptosis de células mesangiales humanas en cultivo, caracterizada porestudios de microscopía óptica y de fluorescencia (tinciones de Giemsa y naranja deacridina, respectivamente), electroforesis del ADN fragmentado, citometría de flujo(anexina-V/ioduro de propidio) e inmunocitoquímica (TUNEL). Ni HX531 (pan-antagonistaRXR), ni AGN193109 (pan-antagonista RAR) redujeron el grado de muerte celularinducido por el AR-t, lo que sugiere un mecanismo independiente de receptores. Resultadosprevios de nuestro grupo indican que dos de los tres miembros de las quinasasactivadas por mitógenos (MAP), ERK (quinasa regulada por estímulos extracelulares) yJNK (quinasa de c-Jun), están implicados en efectos no apoptóticos del AR-t en célulasmesangiales. Nos centramos, pues, en el potencial pro-apoptótico del tercer miembro,la quinasa activada por estrés p38. Confirmamos su implicación en la apoptosis inducidapor el AR-t porque: 1) su inhibidor farmacológico, SB203580, previno dicha apoptosis2) El AR-t indujo en pocos minutos la fosforilación de p38, manteniéndose fosforiladadurante las 8 horas posteriores; y 3) dicha fosforilación se inhibió por preincubación conSB203580. Estos datos sugieren una posible toxicidad renal del AR-t, pero también suutilidad para controlar la proliferación patológica de células mesangiales


All-trans retinoic acid (AR-t) is used for treating acute promyelocytic leukemia andrenal cell carcinoma and it also has therapeutic value in several animal models of renaldisease. Among its renal targets, mesangial cells have been widely studied: they haveboth retinoic acid receptors (RAR) and retinoid X receptors (RXR) and the cell growthis inhibited when human mesangial cells are incubated with 1-10 µM AR-t. Althoughhis effect has been related with the antiproliferative action of AR-t, there are no studieson the involvement of apoptosis in AR-t induced cell growth when higher concentrationsof retinoid are used. Our studies show that 25 µM AR-t triggers mesangial cellapoptosis assessed by light and fluorescence microscopy (Giemsa stain and acridineorange stain, respectively), DNA electrophoresis, flow cytometry (annexin-V) andimmunocytochemistry (TUNEL). AR-t induced apoptosis was not inhibited by preincubationwith the RXR pan-antagonist HX531 nor with the RAR pan-antagonist AGN193109, this suggesting RAR and RXIR are not involved in AR-t induced cell death.Previous results of our group showed that ERK (extracellular regulated kinase) andJNK (c-Jun kinase), two members of the MAP (mitogen activated protein) kinasefamily, are involved in non apoptotic effects of AR-t on mesangial cells. Therefore wefocussed on the stress activated p38 kinase, the third member of the MAPK family, toinvestigate its involvement in AR-t induced apoptosis. The results confirmed a role ofp38 since: 1) preincubation with SB203589, a p38 inhibitor, inhibited ARA inducedapoptosis; 2) incubation with AR-t induced p38 phosphorilation after few minutesand p38 remained phosphorilated for at least 8 hours and 3) AR-t induced p38 phosphorilationwas inhibited by SB203589. These data suggest that AR-t migth have toxicside effects on the kidney but also suggest that AR-t could be an useful inhibitor of pathologicalmesangial cell expansion


Subject(s)
Humans , Apoptosis , Glomerular Mesangium/cytology , Tretinoin/pharmacology , Cells, Cultured
13.
Ann Oncol ; 15(2): 201-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14760109

ABSTRACT

BACKGROUND: We wanted to assess the toxicity and efficacy of paclitaxel plus gemcitabine in advanced breast cancer and to confirm whether circulating HER2 extracellular domain (ECD) correlates with treatment response. PATIENTS AND METHODS: Forty-three patients received paclitaxel 150 mg/m2 followed by gemcitabine 2500 mg/m2, both on day 1 of 14-day cycles, with a maximum of eight cycles. Serum levels of HER2 ECD were assessed by ELISA. RESULTS: All patients were evaluable for toxicity and 42 for efficacy. Overall toxicity was low. Grade 3 neutropenia occurred in 12% of patients and grade 4 in 17%, and other grade 3 toxicities in <5%. One patient had an allergic infusion reaction. Overall response rate was 71% [95% confidence interval (CI) 62% to 81%], with 11 patients achieving a complete response (26%). With a median follow-up of 26 months, the median time to progression was 16.6 months. Response rate correlated significantly with HER2 ECD, with 42% of HER2 ECD-positive patients responding versus 83% of HER2 ECD-negative patients (P = 0.02). Furthermore, response duration was shorter in patients with positive HER2 ECD levels (7.9 versus 14.4 months; P = 0.04). CONCLUSIONS: Paclitaxel plus gemcitabine given as an every 2-weeks schedule is a well tolerated and active regimen in advanced breast carcinoma. This is an attractive combination to use when anthracyclines are not indicated, such as in HER2 positive cases that receive trastuzumab. In addition, elevated levels of HER2 ECD adversely affect the efficacy of treatment.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Deoxycytidine/analogs & derivatives , Genes, erbB-2 , Adenocarcinoma/pathology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , DNA, Neoplasm/analysis , Deoxycytidine/administration & dosage , Disease Progression , Disease-Free Survival , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infusions, Intravenous , Middle Aged , Paclitaxel/administration & dosage , Predictive Value of Tests , Prognosis , Treatment Outcome , Gemcitabine
14.
Leukemia ; 17(2): 437-41, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12592344

ABSTRACT

The objective of our study was to determine the effect of adding r-metHuSCF to Filgrastim and cyclophosphamide for mobilization of peripheral blood progenitor cells (PBPC), on collection of CD34(+) cells and engraftment after autologous stem cell transplant. Twenty-three patients with previously treated stage II-IV breast cancer received cyclophosphamide (3 g/m(2)), Filgrastim 5 microg/kg daily and r-metHuSCF 20 microg/kg daily. Two PBPC collections were performed on consecutive days starting the day the WBC count was above 7.5 x 10(3)/microl. Collection was performed between days +9 and +12 and the median number of CD34(+) cells collected was 9.9 x 10(6)/kg (1.1-53.1) and 6.6 x 10(6)/kg (1.4-33.8) for the first and second apheresis, respectively. Despite being previously treated patients, the target CD34(+) cell dose required for SCT was obtained in all patients. SCT was associated with rapid neutrophil and platelet engraftment and a highly significant correlation was observed between the number of CD34(+) cells infused and engraftment. Treatment with SCF plus filgrastim was well tolerated, with mild to moderate local skin rash being the most frequently reported adverse event. In conclusion, addition of r-metHuSCF induces mobilization of a large number of CD34(+) cells which results in shortening of time to engraftment and hospitalization.


Subject(s)
Breast Neoplasms/therapy , Cyclophosphamide/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/pathology , Stem Cell Factor/analogs & derivatives , Stem Cell Factor/therapeutic use , Stem Cell Transplantation/methods , Adult , Aged , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Filgrastim , Hematopoietic Stem Cells/drug effects , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Recombinant Proteins/therapeutic use , Stem Cell Transplantation/adverse effects , Transplantation, Autologous , Treatment Outcome
15.
Bone Marrow Transplant ; 29(9): 737-43, 2002 May.
Article in English | MEDLINE | ID: mdl-12040470

ABSTRACT

The administration of G-CSF post transplant has been shown to accelerate the time to neutrophil engraftment. However, this does not necessarily translate into a meaningful clinical benefit to the patient. This randomized study was designed to determine the role of G-CSF following transplantation in patients with breast cancer (BC). A total of 241 evaluable patients with BC were included. There were 200 patients with high-risk BC, and 41 had disseminated BC in complete remission. All patients received conventional dose chemotherapy prior to transplantation. Patients were mobilized with G-CSF, received the STAMP V regimen, were transplanted with > or = 2.5 x 10(6) of CD34(+) cells/kg and were then randomized to receive 5 microg/kg of G-CSF starting on the day of infusion (arm A), five days later (arm B), or no G-CSF (arm C). The need for transfusion support, infectious complications and length of hospitalization were the variables chosen to demonstrate clinical benefit. Patients receiving G-CSF reached 500 and 1000 neutrophils significantly faster (P = 0.001) than patients with no G-CSF. This translated into a significantly (P < 0.05) shorter hospitalization time for patients receiving G-CSF. Arm C was closed and, after recruiting 110 patients in arm A, and 106 in arm B, the significant difference in neutrophil recovery persisted with no difference in the time of hospitalization between arms A and B. Therefore, G-CSF significantly accelerates the time to neutrophil engraftment. This translates into a shorter time of hospitalization. There is no difference in this variable regarding the time of administering the G-CSF: day 0 vs day +5. Therefore, G-CSF on day +5 should be the standard in this setting.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Peripheral Blood Stem Cell Transplantation/methods , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/therapy , Female , Graft Survival/drug effects , Hematopoiesis/drug effects , Humans , Middle Aged , Neutrophils/cytology , Neutrophils/drug effects , Transplantation, Autologous/methods
16.
Cancer ; 92(10): 2508-16, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11745183

ABSTRACT

BACKGROUND: Currently employed high-dose regimens for patients with breast carcinoma consist mainly of single-cycle combinations of alkylating agents. In a previous Phase I trial, the authors developed a tandem high-dose combination of two cycles of mitoxantrone and cyclophosphamide for the treatment of patients with metastatic breast carcinoma (MBC) and high-risk breast carcinoma (HRBC). Treatment was delivered with granulocyte-colony stimulating factor (G-CSF) but without stem cell support to avoid potential tumor cell reinfusion. The objective was to validate the safety and obtain preliminary efficacy assessment of this combination in a Phase II trial. METHODS: Fifty-three patients were included: 27 patients with MBC and 26 patients with HRBC. After standard induction treatment, patients received two cycles of mitoxantrone 25 mg/m2 and cyclophosphamide 4000 mg/m2 separated by a 4-week interval. Patients received G-CSF and ciprofloxacin until hematologic recovery. Follow-up was performed in an outpatient setting. RESULTS: One hundred one of 106 projected cycles (95%) were delivered. The mean dose intensities achieved were mitoxantrone 5.8 mg/m2 per week and cyclophosphamide 933 mg/m2 per week. Infection developed in 46% of the cycles, and platelet transfusions were required in 42%. Nonhematologic toxicity was mainly Grade 3 emesis. There were no toxic deaths. In 17 evaluable patients with MBC, 13 patients (77%) had response improvements, including 7 complete responses (41%). CONCLUSIONS: Treatment with two cycles of mitoxantrone 25 mg/m2 and cyclophosphamide 4000 mg/m2 with G-CSF but without stem cell support was well tolerated. The dose intensities achieved approach those obtained with conventional high-dose therapy. This combination warrants further investigation as an alternative to conventional high-dose regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Hematopoietic Stem Cell Transplantation , Adult , Breast Neoplasms/pathology , Cyclophosphamide/administration & dosage , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Infections/chemically induced , Infusions, Intravenous , Middle Aged , Mitoxantrone/administration & dosage , Neoplasm Metastasis , Risk Factors , Thrombocytopenia/chemically induced , Treatment Outcome
17.
Bone Marrow Transplant ; 27(2): 117-23, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11281378

ABSTRACT

This phase I study was designed to develop a high-dose combination of two cycles of mitoxantrone and cyclophosphamide in patients with solid tumors, as an alternative to single-cycle high-dose regimens that use only alkylating agents. Treatment was delivered with granulocyte colony-stimulating factor (G-CSF), but without stem cell support, in order to avoid potential tumor cell reinfusion. Thirty-one patients with advanced solid tumors received two cycles of high-dose mitoxantrone (20-30 mg/m2) plus high-dose cyclophosphamide (3000-4000 mg/m2). All patients received G-CSF until hematologic recovery. Dose-escalation was performed when less than 50% of cycles per level had dose-limiting toxicity (DLT). The maximum tolerated dose (MTD) achieved was mitoxantrone 25 mg/m2 and cyclophosphamide 4000 mg/m2. Main dose-limiting toxicities (DLTs) were hematological: grade IV neutropenia lasting more than 7 days and thrombopenia below 20 x 10(9)/l requiring more than one platelet transfusion. Non-hematological DLT consisted predominantly of grade III emesis and asthenia. Follow-up after each cycle was performed in an outpatient setting and there were no toxic deaths. In conclusion, the administration of two cycles of high-dose mitoxantrone and cyclophosphamide with G-CSF support is safe and feasible. MTD was mitoxantrone 25 mg/m2 and cyclophosphamide 4000 mg/m2. Evaluation of this regimen is being done in a phase II trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cyclophosphamide/administration & dosage , Mitoxantrone/administration & dosage , Neoplasms/drug therapy , Adolescent , Adult , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Transplantation , Humans , Middle Aged , Neoplasms/pathology , Treatment Outcome
18.
Leukemia ; 15(3): 430-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11237067

ABSTRACT

Administration of stem cell factor (SCF) has been proven to enhance cytokine-induced mobilization of CD34+ hematopoietic progenitor cells (HPC) into the peripheral blood (PB). The aim of the present study was to explore in a homogeneous group of 22 uniformly treated breast cancer patients: (1) the kinetics of mobilization into PB of both CD34+ and CD34- cell subsets, including dendritic cells, in sequential samples obtained from day +7 up to day +12 after mobilization; and (2) the composition of the CD34+ and CD34- cell subsets present in the two leukapheresis products obtained for each patient. The following CD34+ and CD34- subsets were analyzed: early CD34+ HPC, erythroid-, myeloid- and B-lymphoid-committed CD34+ precursor cells, mature T, B and NK cells, monocytes, neutrophils, eosinophils, basophils, and dendritic cells (DC) including three subsets of lin-/HLADR+DC (CD16+, CD33high and CD123high). Our results show that the absolute number of PB CD34+ HPC progressively increases from day +7 onwards. As far as the CD34- PB leukocyte subsets are concerned, monocytes (CD14+) displayed the earliest recovery after mobilization predicting neutrophil recovery 1 day in advance. The number of CD34+ HPC collected in a single leukapheresis product was always > or = 1.4 x 10(6) cells/kg body weight. No significant changes were observed between the two leukapheresis sessions either as regards their composition in CD34+ HPC subsets or their CD34- leukocyte populations except for a higher ratio of both CD34+ erythroid/CD34+ myeloid HPC (0.35 +/- 0.13 vs 0.30 +/- 0.13; P = 0.04) and neutrophils/monocytes (1.58 +/- 2.1 vs 0.69 +/- 0.27; P = 0.009) found for the first leukapheresis. Interestingly, the overall number of dendritic cells (DC) was higher in the second leukapheresis (1.06 +/- 0.56 vs 1.9 +/- 0.46; P = 0.02) due to a selective increase of the CD16+ antigen-presenting cells. In summary, our results show that the combination of cyclophosphamide, G-CSF and SCF is highly effective for stem cell mobilization, with differences observed in the mobilization kinetics of the different hematopoietic cell subsets analyzed.


Subject(s)
Antigens, CD34/blood , Breast Neoplasms/immunology , Cyclophosphamide/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization , Stem Cell Factor/administration & dosage , Adolescent , Adult , Aged , Breast Neoplasms/drug therapy , Female , Humans , Immunophenotyping , Leukapheresis , Middle Aged , Recombinant Proteins
19.
J Natl Cancer Inst ; 93(1): 31-8, 2001 Jan 03.
Article in English | MEDLINE | ID: mdl-11136839

ABSTRACT

BACKGROUND: Granulocyte colony-stimulating factors (G-CSFs) have been shown to help prevent febrile neutropenia in certain subgroups of cancer patients undergoing chemotherapy, but their role in treating febrile neutropenia is controversial. The purpose of our study was to evaluate-in a prospective multicenter randomized clinical trial-the efficacy of adding G-CSF to broad-spectrum antibiotic treatment of patients with solid tumors and high-risk febrile neutropenia. METHODS: A total of 210 patients with solid tumors treated with conventional-dose chemotherapy who presented with fever and grade IV neutropenia were considered to be eligible for the trial. They met at least one of the following high-risk criteria: profound neutropenia (absolute neutrophil count <100/mm(3)), short latency from previous chemotherapy cycle (<10 days), sepsis or clinically documented infection at presentation, severe comorbidity, performance status of 3-4 (Eastern Cooperative Oncology Group scale), or prior inpatient status. Eligible patients were randomly assigned to receive the antibiotics ceftazidime and amikacin, with or without G-CSF (5 microg/kg per day). The primary study end point was the duration of hospitalization. All P values were two-sided. RESULTS: Patients randomly assigned to receive G-CSF had a significantly shorter duration of grade IV neutropenia (median, 2 days versus 3 days; P = 0.0004), antibiotic therapy (median, 5 days versus 6 days; P = 0.013), and hospital stay (median, 5 days versus 7 days; P = 0.015) than patients in the control arm. The incidence of serious medical complications not present at the initial clinical evaluation was 10% in the G-CSF group and 17% in the control group (P = 0.12), including five deaths in each study arm. The median cost of hospital stay and the median overall cost per patient admission were reduced by 17% (P = 0.01) and by 11% (P = 0.07), respectively, in the G-CSF arm compared with the control arm. CONCLUSIONS: Adding G-CSF to antibiotic therapy shortens the duration of neutropenia, reduces the duration of antibiotic therapy and hospitalization, and decreases hospital costs in patients with high-risk febrile neutropenia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Fever/etiology , Granulocyte Colony-Stimulating Factor/economics , Granulocyte Colony-Stimulating Factor/therapeutic use , Neutropenia/chemically induced , Neutropenia/drug therapy , Aged , Anti-Bacterial Agents/therapeutic use , Cost-Benefit Analysis , Drug Administration Schedule , Female , Fever/chemically induced , Fever/microbiology , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Length of Stay , Male , Middle Aged , Neoplasms/drug therapy , Neutropenia/complications , Proportional Hazards Models , Prospective Studies , Spain , Survival Analysis , Time Factors , Treatment Outcome
20.
J Clin Oncol ; 18(10): 2126-34, 2000 May.
Article in English | MEDLINE | ID: mdl-10811678

ABSTRACT

PURPOSE: To determine whether the addition of rifampin to a quinolone-based antibacterial prophylactic regimen in patients undergoing high-dose chemotherapy (HDC) with peripheral-blood stem-cell transplantation (PBSCT) decreases the incidence of neutropenia and fever, Gram-positive bacteremia, and infection-related morbidity. PATIENTS AND METHODS: Patients with solid tumors undergoing HDC with PBSCT were randomized to receive prophylactic antibiotics with either ciprofloxacin 500 mg orally every 8 hours or the same ciprofloxacin regimen with rifampin 300 mg orally every 12 hours. Prophylaxis was started 48 hours before stem-cell reinfusion. Patients were monitored to document the occurrence of neutropenia and fever, incidence and cause of bacterial infection, time to onset and duration of fever, requirement for intravenous antimicrobials, and length of hospital admission. RESULTS: Sixty-five patients were randomized to receive ciprofloxacin and 65 to receive ciprofloxacin plus rifampin, and from these groups, 62 and 61 were assessable, respectively. The proportion of patients who developed neutropenia and fever was 87% in the group treated with ciprofloxacin and 78% in the group treated with ciprofloxacin and rifampin (P =.25). Although there was a trend toward a reduction in the overall incidence of bacteremia (12 v 4 patients), and Gram-positive bacteremia (8 v 2 patients) with the addition of rifampin, none of these comparisons was statistically significant (P =.05 and P =.09, respectively). CONCLUSION: The results of this study, which demonstrate that rifampin does not improve ciprofloxacin antibacterial prophylaxis in cancer patients undergoing HDC with PBSCT support but that it does increase the occurrence of undesirable side effects, do not support the routine use of rifampin in this setting.


Subject(s)
Anti-Infective Agents/therapeutic use , Antibiotics, Antitubercular/therapeutic use , Antineoplastic Agents/adverse effects , Ciprofloxacin/therapeutic use , Gram-Positive Bacterial Infections/prevention & control , Neutropenia/prevention & control , Rifampin/therapeutic use , Adult , Anti-Bacterial Agents , Drug Therapy, Combination/therapeutic use , Female , Fever/chemically induced , Fever/prevention & control , Hematopoietic Stem Cell Transplantation , Humans , Male , Neoplasms/drug therapy , Neoplasms/therapy , Neutropenia/chemically induced , Prospective Studies , Statistics, Nonparametric , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...