Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Med. infant ; 26(4): 364-367, dic. 2019. ilus, Tab
Article in Spanish | LILACS | ID: biblio-1047049

ABSTRACT

Introducción: La Oxigenoterapia de Alto Flujo (OAF) es una técnica de soporte respiratorio no invasiva, que ofrece un flujo de aire y oxígeno, caliente y humidificado, por encima del flujo pico inspiratorio del paciente, a través de una cánula nasal. En este artículo se presenta la experiencia con OAF en una sala de pediatría de mediana y baja complejidad para el tratamiento de bronquiolitis/ infección respiratoria aguda baja (IRAB). Materiales y métodos: Se diseñó un protocolo para la implementación de OAF. Criterios de inclusión: Pacientes cursando bronquiolitis/ IRAB con: Score de Tal modificado ≥6, Sat O2 < 92% y/o mala mecánica ventilatoria, a pesar de recibir más de 2 lt/ min de O2 por cánula nasal ó FiO2 >40%. Criterios de exclusión, pCO2 ≥55 mmHg; pH: < 7,20; Apneas ≥20 segundos; Glasgow ≤10; Peso >15 kg. Inestabilidad hemodinámica; Alteraciones craneofaciales. Resultados: En el periodo 2017- 2018 se internaron 441 pacientes con infección respiratoria aguda baja. Se administró OAF a 54 pacientes (12%). La mediana de edad mediana 7,4 meses (r: 27 días-36 meses). Los pacientes ingresados no presentaban comorbilidades asociadas. El 22,2% (12/54) fueron trasladados a UTIP (2,7% del total de los internados). El 64.8% de los pacientes que permanecieron en sala de internación, mostró mejoría en FC y FR a las 4 hs. Por el contrario, en el 75% de los pacientes que requirieron UTIP no se evidenció mejoría en estos parámetros. Conclusiones: La OAF es una alternativa terapéutica que podría disminuir el ingreso a UTIP en pacientes con dificultad respiratoria moderada. En nuestra experiencia resultó fácil de implementar, sin efectos adversos graves (AU)


Introduction: High-flow oxygen (HFO) therapy is a non-invasive oxygen support technique that provides hot and humidified air and oxygen flow above the peak inspiratory flow of the patient through a nasal cannula. In this study we present our experience with HFO on a intermediate and low complexity ward for the treatment of bronchiolitis/acute lower respiratory tract infection (LRTI). Material and methods: A protocol for the implementation of HFO was designed. Inclusion criteria: Patients with bronchiolitis/ALRI with: Modified Tal score ≥6, Sat O2 < 92%, and/or poor ventilatory mechanism, in spite of receiving more than 2 L/ min O2 by nasal cannula or FiO2 >40%. Exclusion criteria: pCO2 ≥55 mmHg; pH: < 7.20; Apnea ≥20 seconds; Glasgow score ≤10; Peso >15 kg. Hemodynamic instability; Craniofacial abnormalities. Results: During 2017- 2018, 441 patients were admitted with LRTI. HFO was administered to 54 patients (12%). Median age was 7.4 months (r: 27 days-36 months). The patients that were included in the study did not have associated morbidities. Overall, 22.2% (12/54) were transferred to the PICU (2.7% of all hospitalized patients). Of the patients who remained on the ward, 64.8% improved FC and FR after 4 hours. On the other hand, in 75% of the patients that required PICU admission these parameters did not improve. Conclusions: HFO is a therapeutic option to decrease PICU admission of patients with moderate respiratory difficulties. The protocol was easy to implement and was not associated with severe adverse effects (AU)


Subject(s)
Humans , Infant, Newborn , Infant , Child, Preschool , Oxygen Inhalation Therapy/instrumentation , Oxygen Inhalation Therapy/methods , Respiratory Tract Infections/therapy , Bronchiolitis/therapy , Retrospective Studies , Cannula
2.
Oncology ; 61(3): 234-42, 2001.
Article in English | MEDLINE | ID: mdl-11574780

ABSTRACT

OBJECTIVES: Systematic investigation of a novel series of intercalating agents, 9-aza-anthrapyrazoles, has led to the identification of a promising analogue, BBR 3438. This study describes the antitumour efficacy of the novel compound in human prostate carcinoma models and the molecular/cellular basis of its activity. METHODS AND RESULTS: The novel 9-aza-anthrapyrazole BBR 3438 was significantly more effective than doxorubicin and losoxantrone (DuP-941) in two of the three tested prostate carcinoma models. The superior activity was more evident in PC3 tumour, since BBR 3438 produced an appreciable rate of complete tumour regressions. Under these conditions, the drug-induced antiproliferative activity paralleled delayed apoptosis. Tumour response to in vivo drug treatment was associated with an early down-regulation of Bcl-2, which was somewhat more marked for the aza compound. In fact, the 9-aza-anthrapyrazole induced DNA cleavage in vitro with isolated DNA topoisomerase II (isoform alpha) and DNA strand breaks in prostatic carcinoma cells. Although the molecular effects of losoxantrone and the 9-aza analogue on the enzyme target were comparable, the cytotoxic effects of BBR 3438 could be enhanced by long-term exposure as a consequence of favourable cellular accumulation and prominent DNA-binding affinity. In addition, a lower reduction potential of the 9-aza-anthrapyrazole in comparison with classical anthrapyrazoles suggests an increased ability of the drug to induce oxidative stress following free radical production, which may be a contributing factor in determining the long-term response (i.e. delayed cell death) to genotoxic damage. CONCLUSIONS: BBR 3438 exhibited a unique profile of preclinical activity with a superior efficacy against prostatic carcinoma models compared to reference compounds (doxorubicin and losoxantrone). The antitumour efficacy of BBR 3438 against prostatic carcinoma could be the result of a combination of favourable events, including enhanced intracellular accumulation and an increased DNA-binding affinity favouring the accumulation of multiple sublethal or lethal damage. In spite of its enhanced cytotoxic potency, the 9-aza compound was better tolerated in vivo than losoxantrone, thus improving the therapeutic index. The preclinical profile of efficacy against prostatic carcinoma, a tumour resistant to conventional antitumour drugs, makes the novel 9-aza-anthrapyrazole BBR 3438 a promising candidate for clinical evaluation.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Ethanolamines/therapeutic use , Intercalating Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyrazolones , Animals , Anthraquinones/therapeutic use , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , DNA Damage , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/drug effects , DNA-Binding Proteins , Doxorubicin/therapeutic use , Ethanolamines/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genes, bcl-2 , Humans , Intercalating Agents/pharmacology , Male , Mice , Mice, Nude , Molecular Structure , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyrazoles/pharmacology , Remission Induction , Xenograft Model Antitumor Assays
3.
Int J Cancer ; 92(5): 738-47, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11340581

ABSTRACT

Loss of p53 function has been linked to increased responsiveness to taxane treatment of ovarian carcinoma in clinical studies. We recently reported that the acquisition of cisplatin resistance in an ovarian carcinoma cell line (IGROV-1) was associated with mutation of p53 and collateral sensitivity to paclitaxel. The increased sensitivity to paclitaxel of the cisplatin-resistant subline appeared to be pharmacologically relevant since it was reflected in an in vivo sensitization to taxanes. To investigate the cellular and molecular basis of this phenomenon, we performed a comparative study of cellular response to taxanes (paclitaxel and the novel analog IDN 5109) in the parental cell line, containing wild-type p53 and its cisplatin-resistant p53 mutant subline (IGROV-1/Pt1). IDN 5109 was included in this study because of its higher potency and efficacy compared with paclitaxel on both tumor systems. The pattern of cellular response of the two ovarian cell lines was different. In IGROV-1 cells, apoptosis was an early event consequent to a transient mitotic arrest. The cell death of IGROV-1/Pt1 cells was a somewhat slow and delayed event, following mitotic arrest and appearance of hyperploid cells. The increased cytotoxic effect of IDN 5109, compared with paclitaxel, was associated with more marked p34(cdc2) dephosphorylation in IGROV-1 cells and higher Bcl-2 phosphorylation in IGROV-1/Pt1 cells after 24 hr of treatment. In each cell line, these biochemical events were not correlated with parallel levels of mitotic cells. Attempts to reintroduce wild-type p53 in IGROV-1/Pt1 were unsuccessful. However, in other p53-deficient cells (osteosarcoma SAOS), taxane treatment was associated with hyperploid progression and the introduction of wild-type p53 resulted in a reduced sensivity. Although our approach does not allow definitive conclusions, these results suggest that loss of p53-dependent post-mitotic checkpoint results in a different time-course of taxane-induced cell death following DNA reduplication. These events, more evident after exposure to the potent analog IDN 5109, support the notion that the enhanced sensitivity of p53 mutant cells is closely related to the different mode of cell death.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Bridged-Ring Compounds/pharmacology , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Taxoids , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Cell Cycle/drug effects , DNA, Neoplasm/analysis , Female , Humans , Ovarian Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin/metabolism , Tumor Cells, Cultured
4.
Eur J Cancer ; 37(3): 431-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11239767

ABSTRACT

MEN10755 is a disaccharide analogue of doxorubicin (DXR) endowed with a broader spectrum of activity compared with DXR in a panel of human tumour xenografts. In an attempt to investigate the pharmacological basis of the improvement of therapeutic efficacy of the analogue, a comparative pharmacokinetic (tissue and tumour distribution) and pharmacodynamic (antitumoral activity and ability to induce apoptosis) study of MEN10755 and DXR was performed in athymic nude mice bearing a human ovarian carcinoma xenograft (A2780). Drug level was quantified by high performance liquid chromatography (HPLC) with fluorimetric detection after a single intravenous (i.v.) injection of 7 mg/kg of MEN10755 or DXR. The results indicated a reduced accumulation of MEN10755 compared with DXR in all tissues investigated (tumour, heart, kidney and liver). The reduction was more marked in normal than tumour tissues. Moreover, in spite of the reduced drug uptake by tumour tissues, the new disaccharide anthracycline given in its optimal regimen showed an enhanced antitumour efficacy, compared with DXR. The drug effects on tumour growth paralleled a marked activation of apoptosis. In conclusion, the pattern of tissue distribution and the pharmacokinetic behaviour were consistent with a better tolerability of the novel analogue which allowed a higher cumulative dose to be delivered. The superior therapeutic efficacy of the analogue over DXR, in spite of a reduced tumour accumulation, supports an increased tumour selectivity.


Subject(s)
Antineoplastic Agents/therapeutic use , Disaccharides/therapeutic use , Doxorubicin/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Disaccharides/pharmacokinetics , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Female , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Transplantation, Heterologous
5.
Prostate ; 46(4): 327-35, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11241556

ABSTRACT

BACKGROUND: RORalpha is a transcription factor which belongs to the family of orphan nuclear receptors. The regulatory functions of this receptor are still poorly understood. However, response elements for RORalpha are present on the promoter of cell cycle-related genes suggesting that it might be involved in the control of cell proliferation. In this study, we investigated the expression and the possible function of RORalpha in a human androgen-independent prostate cancer cell line (DU 145). The thiazolidinedione-derivative CGP 52608 has been utilized as the specific ligand and activator of RORalpha. METHODS: The effects of CGP 52608 on DU 145 cell proliferation and cell cycle distribution were analyzed by hemocytometer and by FACS analysis, respectively. The expression of RORalpha as well as the effects of RORalpha activation on the expression of cell cycle-related genes were evaluated by RT-PCR. To clarify whether RORalpha activation might affect the proliferation of prostate cancer cells also in vivo, nude mice bearing DU 145 tumor xenografts were treated with CGP 52608 at different doses and the growth of the tumors was followed by caliper measurement. RESULTS: RORalpha is expressed in DU 145 cells and the treatment of the cells with the thiazolidinedione-derivative CGP 52608 brought about a dose-dependent and significant decrease of cell proliferation. Ligand-induced activation of RORalpha affected cell cycle distribution, inducing an accumulation in the G(0)/G(1) phase and a decrease in the S phase. This effect was accompanied by an increased expression of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) and a decreased expression of cyclin A. The growth of DU 145 tumors in nude mice was significantly reduced by treatment with CGP 52608. CONCLUSIONS: These data indicate that, in androgen-independent DU 145 prostate cancer cells, activation of the orphan nuclear receptor RORalpha inhibits cell growth, both in vitro and in vivo. RORalpha also induces cell cycle arrest, possibly through the modulation of the expression of cell cycle-related genes.


Subject(s)
Antineoplastic Agents/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacology , Thiosemicarbazones/pharmacology , Trans-Activators/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Male , Mice , Mice, Nude , Nuclear Receptor Subfamily 1, Group F, Member 1 , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/administration & dosage , Thiosemicarbazones/administration & dosage , Trans-Activators/drug effects , Trans-Activators/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
6.
Curr Med Chem ; 8(1): 31-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11172690

ABSTRACT

Cellular resistance to anthracyclines is a major limitation of their clinical use in the treatment of human tumors. Resistance to doxorubicin is described as a multifactorial phenomenon involving the overexpression of defense factors and alterations in drug-target interactions. Such changes do not account for all manifestations of drug resistance, in particular intrinsic resistance of solid tumors. Since anthracyclines can induce apoptotic cell death, an alternative promising approach to drug resistance has focused on the study of cellular response to drug-induced DNA damage, with particular reference to the relationship between cytotoxicity/antitumor efficacy and apoptotic response. The evidence that a novel disaccharide analog (MEN 10755), endowed with an improved preclinical activity over doxorubicin, was also more effective as an inducer of apoptosis provided additional insights to better understand the cellular processes that confer sensitivity to anthracyclines. Although the presence or alteration of a single apoptosis-related factor (e.g., p53, bcl-2) is not predictive of the sensitivity/resistance status, the complex interplay among DNA damage-activated pathways is likely an important determinant of tumor cell sensitivity to anthracyclines


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Apoptosis/genetics , Apoptosis/physiology , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Humans , Neoplasms/pathology
7.
Clin Cancer Res ; 6(7): 2626-34, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914703

ABSTRACT

Multinuclear platinum complexes represent a new class of anticancer agents, distinct in terms of DNA binding features and the profile of antitumor activity from their mononuclear counterparts, in particular cisplatin. Among complexes of this class, BBR 3464, a trinuclear platinum compound has been selected for preclinical development. In the present study, we describe the preclinical evaluation of BBR 3464 in a series of human tumor cell lines and tumor xenografts, with special emphasis on tumor types known to be resistant to cisplatin. In a panel of seven human tumor cell lines naturally resistant to cisplatin (three ovarian and four melanomas), BBR 3464 was extremely potent with IC50 values at least 20-fold lower than cisplatin. Against eight human tumor xenografts including four tumors refractory to cisplatin, BBR 3464 was confirmed to be very active with a tumor weight inhibition >80% in seven of them. The efficacy of BBR 3464 against cisplatin-resistant tumors was consistent with the ability of the drug to completely overcome resistance in three cell systems characterized by acquired resistance to cisplatin. Moreover, BBR 3464 caused a more prolonged effect than cisplatin, which was reflected by higher specific growth delay values. This prolonged effect is likely to be related to a more persistent perturbation of the cell cycle induced by BBR 3464 than by cisplatin, as shown in one ovarian tumor cell line. Finally, the profile of sensitivity to BBR 3464 within the 60-cell-lines screening panel of the National Cancer Institute, NIH (Bethesda, MD) differed from those of established drugs, thus supporting the hypothesis of a distinct mechanism of cytotoxic activity of BBR 3464. The novel trinuclear platinum complex, in light of its innovative antitumor activity profile, has the potential to become a useful clinical agent for the treatment of unresponsive tumors.


Subject(s)
Antineoplastic Agents/toxicity , Cell Survival/drug effects , Cisplatin/toxicity , Organoplatinum Compounds/toxicity , Ovarian Neoplasms/drug therapy , Animals , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/pathology , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Biochem Pharmacol ; 59(12): 1539-47, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10799650

ABSTRACT

Targeting of deregulated protein tyrosine kinases has been proposed as a new approach in the therapeutic intervention against pathological processes including proliferative disorders and cancer. Using a screening approach based on a comparative evaluation of antiproliferative effects in a panel of tumor cells with differential expression of protein tyrosine kinases, three benzoquinoid macrolidic fungal metabolites produced by Clitocybe clavipes, clavilactones A, B, and D (CA, CB, and CD) and two semisynthetic derivatives of these products, diacetyl-CA and dimethyl-CA, were identified as inhibitors of protein tyrosine kinases. Naturally occurring CA, CB, and CD showed inhibitory activity in kinase assays against the Ret/ptc1 and epidermal growth factor receptor (EGF-R) tyrosine kinases, while being less effective against the v-Abl tyrosine kinase and p34(cdc2) serine/threonine kinase (IC(50) 2.8, 5.5, 81.3, and 128 microM respectively, for the most potent compound CD). CB was shown to be a non-competitive inhibitor of EGF-R with respect to ATP or poly(Glu(6)Ala(3)Tyr). CD also preferentially inhibited the growth of A431 cells, which overexpress a constitutively active EGF-R, as opposed to IGROV-1 and SKOV-3 cells, which express low levels of the receptor. Further, EGF-R was shown to be a target for clavilactones in A431 cells, since EGF-induced receptor autophosphorylation was inhibited in the presence of CB, CD, and diacetyl-CA. Both CD and diacetyl-CA displayed weak activity when administered daily (i.p.) to mice bearing ascitic A431 tumor. These findings indicate that clavilactones represent the prototypes of a new structural class of tyrosine kinase inhibitors deserving further investigation.


Subject(s)
Agaricales/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Lactones/pharmacology , Macrolides/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Cell Division/drug effects , Cell Line, Transformed , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , ErbB Receptors/metabolism , Humans , Lactones/therapeutic use , Macrolides/therapeutic use , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Phosphorylation , Protein Kinases/metabolism , Tumor Cells, Cultured
9.
Biochem Pharmacol ; 60(1): 77-82, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10807947

ABSTRACT

A variety of cytotoxic agents effective as antitumor drugs are known to kill tumor cells through induction of apoptosis as the most relevant modality of cell death. A specific role for the protein Bcl-2 in the cell death pathway induced by antimicrotubule agents has been proposed, because Bcl-2 phosphorylation occurs in response to microtubule damage. In this study, we compared efficacy, apoptosis, and Bcl-2 phosphorylation in the Bcl-2-overexpressing MX-1 human breast carcinoma xenograft after treatment with cytotoxic agents characterized by different mechanisms of action. We demonstrated that, in addition to antimicrotubule agents, effective DNA-damaging agents were also able to induce Bcl-2 phosphorylation irrespective of the type of genotoxic lesion. A comparison of effects of drugs belonging to the same class but endowed with a different antitumor activity (i.e. cisplatin versus a novel multinuclear platinum complex and doxorubicin versus a disaccharide analogue) showed a correlation between drug efficacy, apoptotic response, and Bcl-2 phosphorylation. In conclusion, overexpression of Bcl-2 did not counteract the apoptotic effects of a number of cytotoxic agents and could not be regarded as a mechanism of cellular resistance. Since Bcl-2 phosphorylation is a common event in response to different types of cytotoxic damage and is not only related to microtubule dysfunction, we suggest that many cell death pathways converge on Bcl-2 and protein phosphorylation is a step of the signaling cascade activated by diverse stimuli and likely related to the onset of drug-induced apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Breast Neoplasms/genetics , DNA Damage/drug effects , Disease Models, Animal , Female , Humans , Mice , Mice, Nude , Microtubules/drug effects , Neoplasm Transplantation , Phosphorylation , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Clin Cancer Res ; 6(5): 2070-4, 2000 May.
Article in English | MEDLINE | ID: mdl-10815934

ABSTRACT

A novel taxane (IDN 5109), originally selected for its ability to overcome P-glycoprotein-mediated drug resistance, is characterized by an improved preclinical profile in terms of efficacy and tolerability. Because P-glycoprotein may critically influence intestinal absorption and oral bioavailability of taxanes, the purpose of the study was to evaluate the bioavailability, the pharmacokinetic behavior, and the antitumor activity of the new taxane after oral administration. A comparative study of antitumor activity of Taxol and IDN 5109 given orally was performed in a human breast carcinoma model, MX-1, which is highly responsive to i.v. treatment with both of the taxanes. In contrast to Taxol, which was completely ineffective after administration to MX-1-bearing mice, oral IDN 5109 exhibited an activity comparable with that of i.v. treatment (ie., 100% cures). Again, the maximal tolerated doses were comparable (90 mg/kg, every 4 days for four doses) after i.v. and oral treatment. Three other tumor models (LoVo, IGROV/DDP, and U87) with a variable sensitivity to the drug were used to compare the antitumor effects of i.v. and oral treatment with IDN 5109. The efficacy after oral administration was only slightly lower than that found after i.v. treatment at equivalent doses; but optimal effects were comparable likely as a consequence of the long (>6 h) terminal half-life of oral IDN 5109. The bioavailability of IDN 5109 assessed by comparing area-under-the-curve values after oral and i.v. administrations was approximately 50%. The oral efficacy of the novel taxane, likely related to the inability of the P-glycoprotein to recognize the drug, which allowed an adequate intestinal absorption, is a unique feature among the taxanes and may represent a pharmacological breakthrough in their clinical use.


Subject(s)
Bridged-Ring Compounds/pharmacokinetics , Bridged-Ring Compounds/therapeutic use , Taxoids , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Area Under Curve , Biological Availability , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Intravenous , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Transplantation, Heterologous , Treatment Outcome , Tumor Cells, Cultured
11.
Carcinogenesis ; 21(12): 2203-10, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11133809

ABSTRACT

In human fibroblasts, N:-phosphoacetyl-L-aspartate (PALA) and gamma-radiation induce reversible and irreversible p53-mediated G(1) cell cycle arrest, respectively. By coupling the premature chromosome condensation technique to fluorescence in situ hybridization, we found no evidence of DNA damage after PALA treatment. We used representational difference analysis (cDNA-RDA) to study changes in gene expression after PALA treatment and gamma-radiation in normal human fibroblasts. The mammary-derived growth inhibitor (MDGI) gene was expressed in PALA-treated cells. Ectopic MDGI expression arrested PALA-treated but not irradiated RKO cells. Expression of an antisense RNA against MDGI resulted in partial G(1) escape of PALA-treated human fibroblasts. The tumor necrosis factor stimulated gene 6, TSG-6, seems to be under the control of p53 and is only and specifically induced upon PALA treatment. In irradiated cells we have identified 'novel' genes that are differentially expressed, along with known genes not previously linked to cell cycle control. Some of these 'novel' genes correspond to clones in the expressed sequence tag (EST) database; one of them shows identity with ESTs mapping to a region on chromosome 7, where gene(s) involved in replicative senescence and frequently deleted in tumors are located. Thus, PALA treatment and gamma-irradiation elicit a pattern of differential gene expression that could contribute to a quiescence or senescence-like phenotype.


Subject(s)
Aspartic Acid/analogs & derivatives , Carrier Proteins/genetics , Cell Adhesion Molecules/genetics , Fibroblasts/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Phosphonoacetic Acid/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Aspartic Acid/toxicity , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 7 , Expressed Sequence Tags , Fatty Acid Binding Protein 3 , Fatty Acid-Binding Proteins , Fibroblasts/cytology , G1 Phase , Gamma Rays , Growth Inhibitors/genetics , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Metaphase , Phosphonoacetic Acid/toxicity , Skin/cytology , Skin/drug effects , Skin/radiation effects
12.
Br J Cancer ; 80(12): 1912-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10471039

ABSTRACT

Multinuclear platinum compounds were rationally designed to bind to DNA in a different manner from that of cisplatin and its mononuclear analogues. A triplatinum compound of the series (BBR 3464) was selected for preclinical development, since, in spite of its charged nature, it was very potent as cytotoxic agent and effective against cisplatin-resistant tumour cells. Anti-tumour efficacy studies were performed in a panel of human tumour xenografts refractory or poorly responsive to cisplatin. The novel platinum compound exhibited efficacy in all tested tumours and an impressive efficacy (including complete tumour regressions) was displayed in two lung carcinoma models, CaLu-3 and POCS. Surprisingly, BBR 3464 showed a superior activity against p53-mutant tumours as compared to those carrying the wild-type gene. The involvement of p53 in tumour response was investigated in an osteosarcoma cell line, SAOS, which is null for p53 and is highly sensitive to BBR 3464, and in the same cells following introduction of the wild-type p53 gene. Thus the pattern of cellular response was investigated in a panel of human tumour cells with a different p53 gene status. The results showed that the transfer of functional p53 resulted in a marked (tenfold) reduction of cellular chemosensitivity to the multinuclear platinum complex but in a moderate sensitization to cisplatin. In addition, in contrast to cisplatin, the triplatinum complex was very effective as an inducer of apoptosis in a lung carcinoma cell line carrying mutant p53. The peculiar pattern of anti-tumour activity of the triplatinum complex and its ability to induce p53-independent cell death may have relevant pharmacological implications, since p53, a critical protein involved in DNA repair and induction of apoptosis by conventional DNA-damaging agents, is defective in several human tumours. We suggest that the peculiar DNA binding properties of the triplatinum complex may contribute to the striking profile of anti-tumour efficacy. Taken together, the available information supports that anti-tumour activity of the novel compound is mediated by a mechanism different from that of conventional platinum complexes, and compounds of this series could represent a new class of promising anti-tumour agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Genes, p53 , Mutation , Neoplasms/drug therapy , Organoplatinum Compounds/therapeutic use , Animals , Cell Cycle/drug effects , Female , Humans , Male , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Neoplasms/genetics , Neoplasms/pathology , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/toxicity , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/pathology , Polymorphism, Single-Stranded Conformational , Transplantation, Heterologous
13.
Cancer Res ; 59(5): 1036-40, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10070960

ABSTRACT

Clinically available taxanes represent one of the most promising class of antitumor agents, despite several problems with their solubility and toxicity. In an attempt to improve the pharmacological profile of taxanes, a new series of analogues was synthesized from 14beta-hydroxy-10-deacetylbaccatin III and tested in a panel of human tumor cell lines. On the basis of the pattern of cytotoxicity and lack of cross-resistance in tumor cell lines expressing the typical multidrug-resistant phenotype, a compound (IDN5109) was selected for preclinical development. A comparative efficacy study of IDN5109 and paclitaxel was performed using a large panel of human tumor xenografts, characterized by intrinsic (seven tumors) or acquired (four tumors) resistance to cisplatin or doxorubicin, including four ovarian, one breast, one cervical, three lung, one colon, and one prostatic carcinoma. Drugs were delivered i.v. according to the same schedule (four times every 4th day). IDN5109 achieved a very high level of activity (percentage tumor weight inhibition >70%; log10 cell kill >1) in all but one of the tested tumors. Compared to paclitaxel, IDN5109 exhibited a significantly superior activity in six tumors (including the four tumors that were resistant to paclitaxel) and a comparable activity against the other five paclitaxel-responsive tumors. Additional advantages of IDN5109 over paclitaxel were also suggested by its toxicity profile. IDN5109 was not only less toxic (maximal tolerated doses were 90 and 54 mg/kg for IDN5109 and paclitaxel, respectively), but it also appeared to be endowed with a reduced neurotoxic potential and an improved profile of tolerability compared to the parent drug. Furthermore, the best antitumor efficacy was often already reached with doses lower than the maximal tolerated dose, suggesting an improved therapeutic index for the new drug. In conclusion, the results support the preclinical interest of IDN5109 in terms of the toxicity profile and of the efficacy with particular reference to the ability to overcome multiple mechanisms of drug resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/toxicity , Animals , Antineoplastic Agents, Phytogenic/toxicity , Breast Neoplasms/drug therapy , Cell Division/drug effects , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Drug Resistance, Multiple , Female , Humans , Male , Mice , Mice, Nude , Paclitaxel/therapeutic use , Prostatic Neoplasms/drug therapy , Transplantation, Heterologous , Tumor Cells, Cultured , Uterine Cervical Neoplasms/drug therapy
14.
Clin Cancer Res ; 4(11): 2833-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829750

ABSTRACT

On the basis of a structure-activity study of a new series of anthracycline disaccharides, we recently identified a doxorubicin analogue (MEN 10755) with a promising antitumor activity. In the present study, to better support the pharmacological interest of MEN 10755, we extended the preclinical evaluation of antitumor efficacy to a large panel of 16 human tumor xenografts, which originated from different clinicopathological types. Tumors with typical multidrug-resistant phenotype were excluded because MEN 10755 was found unable to overcome resistance mediated by transport systems. In the doxorubicin-responsive series, MEN 10755 exhibited a higher activity in three of five tumors, as documented by a more marked tumor growth inhibition and an increased value of log-cell kill. In the series of doxorubicin-resistant tumors, MEN 10755 was found effective in 6 of 11 tumors (1 breast, 3 lung, and 2 prostate carcinomas). The overall response rates were 31% and 69% for doxorubicin and MEN 10755, respectively. The improvement in drug efficacy was also supported by a substantial increase in the long-term survivor rate of animals implanted with responsive tumors. Most of the tumors refractory to doxorubicin and responsive to MEN 10755 were characterized by overexpression of the antiapoptotic protein Bcl-2. In one of these tumors (MX-1 breast carcinoma), we examined the ability of MEN 10755 to induce phosphorylation of Bcl-2 after a single treatment with therapeutic doses. The results indicated that, unlike doxorubicin, MEN 10755 induced protein phosphorylation. A similar modification was produced by Taxol, which is known to be very effective against the tumor. The correlation between drug efficacy and Bcl-2 phosphorylation may underly a peculiar feature related to improvement of efficacy of the disaccharide analogue. In conclusion, the present study supports some favorable features of the novel doxorubicin analogue in terms of both efficacy and tolerability with comparison to doxorubicin, although the improvement is somewhat tumor- and schedule-dependent.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Disaccharides/therapeutic use , Doxorubicin/analogs & derivatives , Animals , Blotting, Western , Carcinoma/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
15.
Int J Cancer ; 76(4): 506-11, 1998 May 18.
Article in English | MEDLINE | ID: mdl-9590126

ABSTRACT

Experiments have been performed to clarify whether LHRH agonists might decrease growth of hormone-unresponsive prostate cancer in vivo. Male nude mice were injected s.c. with the human androgen-independent prostate tumor DU 145 cells; osmotic minipumps releasing the LHRH agonist Zoladex (LHRH-A) for 14 days were simultaneously implanted under the skin. Treatment with LHRH-A induced a significant decrease in tumor growth up to the end of the treatment. In subsequent experiment, minipumps releasing LHRH-A were implanted in nude mice either 7 or 14 days after cell inoculation. When the treatment was started 7 days after inoculation of the cells, tumor growth was significantly decreased up to 28 days; thereafter, tumor volume remained lower than in controls, although not significantly. When LHRH-A was administered beginning 14 days after cell inoculation, tumor growth was not significantly affected at any time interval considered. LHRH-A did not appear to induce apoptosis in DU 145 cells, at least on the basis of the apoptotic index and immunohistochemical staining of the p53 protein. On the other hand, treatment with LHRH-A was accompanied by a significant decrease of the concentration of epidermal growth factor receptors in DU 145 prostate cancer specimens. Our results show that the LHRH agonist used significantly inhibits the growth of DU 145 androgen-independent prostate tumor xenografts in nude mice.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Goserelin/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Apoptosis , Goserelin/pharmacology , Male , Mice , Mice, Nude , Mitotic Index , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/drug therapy , Time Factors , Tumor Cells, Cultured
16.
Br J Cancer ; 77(3): 434-9, 1998.
Article in English | MEDLINE | ID: mdl-9472640

ABSTRACT

The pharmacological interest in lonidamine is related to its ability to enhance the cytotoxic effects of several DNA-damaging anti-tumour agents. This study was undertaken to better understand the in vivo interaction between lonidamine and cisplatin in the treatment of human tumour xenografts, including three carcinoma models characterized by a different responsiveness to cisplatin, in spite of the presence of a wild-type p53 gene in all tumours. The drug combination was more effective in tumour growth inhibition than cisplatin alone against MX-1 breast carcinoma and A2780 ovarian carcinoma, both highly responsive to cisplatin, whereas no influence of ionidamine was observed on anti-tumour activity of cisplatin in the treatment of the relatively resistant IGROV-1 ovarian carcinoma. As cisplatin activity is related to induction of apoptosis, the modulation of drug-induced apoptosis by lonidamine was investigated. Under conditions in which lonidamine itself had negligible effects on tumour growth and apoptosis, the modulating agent stimulated the apoptotic response induced by cisplatin in the responsive but not in the resistant tumours. Tumour response was dependent not only on the drug activation of apoptosis, but mainly on the persistence over time of the event. In the breast carcinoma MX-1, hypersensitive to cisplatin and to the lonidamine+cisplatin combination, the efficacy of drug treatment was associated with phosphorylation of bcl-2 followed by down-regulation of the protein. Lonidamine itself caused a delayed phosphorylation of bcl-2. These results are consistent with the interpretation that lonidamine is effective in modulating biochemical factors involved in regulation of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Indazoles/pharmacology , Animals , DNA Damage , Drug Synergism , Female , Humans , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transplantation, Heterologous
17.
J Natl Cancer Inst ; 89(16): 1217-23, 1997 Aug 20.
Article in English | MEDLINE | ID: mdl-9274917

ABSTRACT

BACKGROUND: Although doxorubicin remains one of the most effective agents for the treatment of solid tumors, there is an intensive effort to synthesize doxorubicin analogues (compounds with similar chemical structures) that may have improved antitumor properties. We have synthesized a novel doxorubicin disaccharide analogue (MEN 10755) and have characterized some of its relevant biochemical, biologic, and pharmacologic properties. METHODS: The antitumor activity of this compound (MEN 10755) was studied in a panel of human tumor xenografts, including xenografts of A2780 ovarian tumor cells, MX-1 breast carcinoma cells, and POVD small-cell lung cancer cells. MEN 10755 was compared with doxorubicin according to the optimal dose and schedule for each drug. The drug's cytotoxic effects, induction of DNA damage, and intracellular accumulation were studied in A2780 cells. DNA cleavage mediated by the enzyme topoisomerase II was investigated in vitro by incubating fragments of simian virus 40 DNA with the purified enzyme at various drug concentrations and analyzing the DNA cleavage-intensity patterns. Drug-induced apoptosis (programmed cell death) in tumors was determined with the use of MX-1 and POVD tumor-bearing athymic Swiss nude mice. RESULTS: MEN 10755 was more effective than doxorubicin as a topoisomerase II poison and stimulated DNA fragmentation at lower intracellular concentrations. In addition, MEN 10755 exhibited striking antitumor activity in the treatment of human tumor xenografts, including those of the doxorubicin-resistant breast carcinoma cell line MX-1. CONCLUSIONS: The high antitumor activity of MEN 10755 in human tumor xenografts, including doxorubicin-resistant xenografts, and its unique pharmacologic and biologic properties make this disaccharide analogue a promising candidate for clinical evaluation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA Topoisomerases, Type II/drug effects , DNA, Neoplasm/drug effects , Doxorubicin/analogs & derivatives , Neoplasms, Experimental/drug therapy , Animals , Breast Neoplasms/drug therapy , Carcinoma, Small Cell/drug therapy , DNA Damage , Disaccharides/chemical synthesis , Doxorubicin/chemical synthesis , Female , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Ovarian Neoplasms/drug therapy , Time Factors , Transplantation, Heterologous
18.
Anticancer Drug Des ; 12(8): 649-57, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9448704

ABSTRACT

Vascularization is an important step in tumor growth and metastasis. Tumor neovascularization can be considered, therefore, as a good target for antineoplastic therapy. In order to target saporin, a powerful plant toxin, in proximity of the tumor we fused the saporin coding sequence to that for placental growth factor-2 (P1GF-2). P1GF is an angiogenic factor involved in tumor neovascularization. The fusion protein P1GF-2-saporin was obtained by transient transfection of mammalian cells and released in the culture medium as a 57.5 kDa polypeptide. Selectivity and cytotoxic activity are reported as a preliminary step towards the evaluation of its in vivo antitumor activity.


Subject(s)
Endothelium, Vascular/drug effects , Immunotoxins , N-Glycosyl Hydrolases , Plant Proteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta/pharmacology , 3T3 Cells/drug effects , Animals , Blotting, Western , COS Cells/drug effects , Endothelium, Vascular/cytology , Glycosylation , Mice , Neovascularization, Pathologic/prevention & control , Plant Proteins/genetics , Plant Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosome Inactivating Proteins, Type 1 , Saporins , Transfection , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...