Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Sci Rep ; 10(1): 15399, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32958816

ABSTRACT

Graft-versus host disease (GVHD) remains one of the main causes of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (ASCT). Prophylactic T cell depletion via antithymocyte globulin (ATG) during ASCT conditioning is one of the standards of care for GVHD prophylaxis, although the optimal dosing strategy is still unclear. Recent studies have reported that absolute lymphocyte count at the time of ATG administration could predict survivals in ASCT from unrelated donors. Here this issue was examined in 116 patients receiving peripheral blood stem cells (PBSC) ASCT with purine analog/busulfan-based conditioning regimens between 2009 and 2019 in our department. The impact of lymphopenia at the time of ATG administration was evaluated in terms of overall survival, disease-free survival and GVHD-free/relapse-free survival. After a median follow-up of 4 years, no adverse effect of a profound lymphopenia was observed on patients' outcome. Notably, a reduced dose of ATG in patients with profound lymphopenia did not translate into better survivals. This study indicates that ATG can be administered whatever the recipient's lymphocyte counts in patients receiving a PBSC purine analog/busulfan-based conditioning regimen ASCT.


Subject(s)
Antilymphocyte Serum/therapeutic use , Lymphopenia/drug therapy , Peripheral Blood Stem Cell Transplantation/methods , Adult , Aged , Allografts , Antilymphocyte Serum/administration & dosage , Antilymphocyte Serum/metabolism , Busulfan/administration & dosage , Disease-Free Survival , Female , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/mortality , Humans , Lymphocyte Depletion , Lymphopenia/etiology , Male , Middle Aged , Peripheral Blood Stem Cells/drug effects , Purine Nucleosides/administration & dosage , Retrospective Studies , Transplantation Conditioning , Transplantation, Homologous
2.
Ann Hematol ; 98(1): 131-142, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29974231

ABSTRACT

Peripheral T cell lymphomas are an aggressive group of non-Hodgkin lymphomas with poor outcomes for most subtypes and no accepted standard of care for relapsed patients. This study evaluated the efficacy and safety of forodesine, a novel purine nucleoside phosphorylase inhibitor, in patients with relapsed peripheral T cell lymphomas. Patients with histologically confirmed disease, progression after ≥ 1 prior treatment, and an objective response to last treatment received oral forodesine 300 mg twice-daily. The primary endpoint was objective response rate (ORR). Secondary endpoints included duration of response, progression-free survival (PFS), overall survival (OS), and safety. Forty-eight patients (median age, 69.5 years; median of 2 prior treatments) received forodesine. In phase 1 (n = 3 evaluable), no dose-limiting toxicity was observed during the first 28 days of forodesine treatment. In phase 2 (n = 41 evaluable), the ORR for the primary and final analyses was 22% (90% CI 12-35%) and 25% (90% CI 14-38%), respectively, including four complete responses (10%). Median PFS and OS were 1.9 and 15.6 months, respectively. The most common grade 3/4 adverse events were lymphopenia (96%), leukopenia (42%), and neutropenia (35%). Dose reduction and discontinuation due to adverse events were uncommon. Secondary B cell lymphoma developed in five patients, of whom four were positive for Epstein-Barr virus. In conclusion, forodesine has single-agent activity within the range of approved therapies in relapsed peripheral T cell lymphomas, with a manageable safety profile, and may represent a viable treatment option for this difficult-to-treat population.


Subject(s)
Lymphoma, T-Cell, Peripheral/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacokinetics , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Administration, Oral , Adult , Aged , Female , Humans , Lymphoma, T-Cell, Peripheral/blood , Male , Middle Aged , Purine Nucleosides/adverse effects , Pyrimidinones/adverse effects , Recurrence
3.
Antiviral Res ; 156: 38-45, 2018 08.
Article in English | MEDLINE | ID: mdl-29864447

ABSTRACT

Rift Valley fever virus (RVFV) is a mosquito-borne pathogen endemic to sub-Saharan Africa and the Arabian Peninsula. There are no approved antiviral therapies or vaccines available to treat or prevent severe disease associated with RVFV infection in humans. The adenosine analog, galidesivir (BCX4430), is a broad-spectrum antiviral drug candidate with in vitro antiviral potency (EC50 of less than 50 µM) in more than 20 different viruses across eight different virus families. Here we report on the activity of galidesivir in the hamster model of peracute RVFV infection. Intramuscular and intraperitoneal treatments effectively limited systemic RVFV (strain ZH501) infection as demonstrated by significantly improved survival outcomes and the absence of infectious virus in the spleen and the majority of the serum, brain, and liver samples collected from infected animals. Our findings support the further development of galidesivir as an antiviral therapy for use in treating severe RVFV infection, and possibly other related phleboviral diseases.


Subject(s)
Antiviral Agents/administration & dosage , Purine Nucleosides/administration & dosage , Rift Valley Fever/drug therapy , Rift Valley fever virus/drug effects , Adenine/analogs & derivatives , Adenosine/analogs & derivatives , Animals , Disease Models, Animal , Injections, Intramuscular , Injections, Intraperitoneal , Liver/virology , Mesocricetus , Pyrrolidines , Spleen/virology , Survival Analysis , Treatment Outcome
4.
Br Poult Sci ; 58(5): 536-543, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28547997

ABSTRACT

1. This study was conducted to evaluate the effects of purine nucleosides on performance, gut morphology, intestinal enzymes and immunity functions in broiler chickens from 0 to 21 d of age. 2. A total of 360 1-d-old male chickens (Cobb 500) were randomly assigned to 4 treatments with 6 replications. Experimental diets consisted of a control without any additives and diets containing 0.1% pure adenosine, 0.1% pure guanosine and 0.1% equal aliquots of pure adenosine and guanosine. Two birds per cage (12 birds per treatment) were killed on d 11 and 21 in order to obtain serum samples for lipid profile, jejunal samples for morphology and mucosal immunity, digestive enzymes for epithelial maturation, and bursa and spleen samples for relative weight of immune organs to live body weight. 3. Birds receiving adenosine in their diets showed a significant increase in body weight and average daily gain and a significantly lower feed conversion ratio compared to the control birds. Villus height and width in jejunal samples also increased significantly in birds supplemented with adenosine. Although maltase was not affected by the experimental diets, adenosine increased alkaline phosphatase and aminopeptidase. Adenosine and its combination with guanosine boosted mucosal immunity as a result of increased IgA production. While there was no significant difference among treatments regarding the relative weight of the spleen, adenosine increased the relative weight of the bursa of Fabricius. Present results also showed that adding guanosine to broiler diets had no significant effects on growth, gut morphology, enzymes activity and immunological indices. 4. In conclusion, the improvement in growth performance, gut morphology and immunity in birds receiving adenosine demonstrated that pure adenosine could be a beneficial feed additive for the poultry industry, while guanosine showed no significant improvement.


Subject(s)
Chickens/physiology , Immunity, Mucosal/drug effects , Intestines/physiology , Purine Nucleosides/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Bursa of Fabricius/physiology , Chickens/anatomy & histology , Chickens/growth & development , Chickens/immunology , Diet/veterinary , Dietary Supplements/analysis , Intestinal Mucosa/immunology , Intestines/enzymology , Intestines/growth & development , Male , Organ Size , Purine Nucleosides/administration & dosage , Random Allocation , Spleen/physiology
5.
Expert Opin Investig Drugs ; 26(6): 771-775, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28447489

ABSTRACT

INTRODUCTION: Cutaneous T-cell lymphoma (CTCL) is characterized by the accumulation of neoplastic CD4+ T lymphocytes in the skin. Given the lack of curative treatments for CTCL, there is a significant need for new, superior therapies. Forodesine is a transition-state analogue that inhibits purine nucleoside phosphorylase. Because it selectively targets T lymphocytes, it represents a drug of interest for the treatment of CTCL. Areas covered: Phase I/II dose-ranging studies of intravenous (IV) and oral forodesine demonstrated its activity, safety, and tolerability for refractory CTCL. Response rates were 31% and 27%, respectively. No dose-limiting toxicities were observed. These studies were followed by a phase II trial of oral forodesine 200 mg daily. This oral formulation showed only partial activity, with a response rate of 11%, likely attributable to underdosing. Common adverse events in these trials included infection, fatigue, peripheral edema, nausea, pruritus, headache, and insomnia. Expert opinion: IV and oral formulations of forodesine have demonstrated partial activity and an acceptable safety profile in patients with refractory CTCL. A higher oral dose, or sequential therapy consisting of IV forodesine followed by maintenance oral forodesine, may be more effective. With proper dosing, forodesine may emerge as a safe and effective treatment for refractory CTCL.


Subject(s)
Lymphoma, T-Cell, Cutaneous/drug therapy , Purine Nucleosides/administration & dosage , Pyrimidinones/administration & dosage , Skin Neoplasms/drug therapy , Administration, Intravenous , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Dose-Response Relationship, Drug , Humans , Lymphoma, T-Cell, Cutaneous/pathology , Purine Nucleosides/adverse effects , Purine Nucleosides/pharmacology , Pyrimidinones/adverse effects , Pyrimidinones/pharmacology , Skin Neoplasms/pathology
6.
Antiviral Res ; 137: 14-22, 2017 01.
Article in English | MEDLINE | ID: mdl-27838352

ABSTRACT

Zika virus (ZIKV) is currently undergoing pandemic emergence. While disease is typically subclinical, severe neurologic manifestations in fetuses and newborns after congenital infection underscore an urgent need for antiviral interventions. The adenosine analog BCX4430 has broad-spectrum activity against a wide range of RNA viruses, including potent in vivo activity against yellow fever, Marburg and Ebola viruses. We tested this compound against African and Asian lineage ZIKV in cytopathic effect inhibition and virus yield reduction assays in various cell lines. To further evaluate the efficacy in a relevant animal model, we developed a mouse model of severe ZIKV infection, which recapitulates various human disease manifestations including peripheral virus replication, conjunctivitis, encephalitis and myelitis. Time-course quantification of viral RNA accumulation demonstrated robust viral replication in several relevant tissues, including high and persistent viral loads observed in the brain and testis. The presence of viral RNA in various tissues was confirmed by an infectious culture assay as well as immunohistochemical staining of tissue sections. Treatment of ZIKV-infected mice with BCX4430 significantly improved outcome even when treatment was initiated during the peak of viremia. The demonstration of potent activity of BCX4430 against ZIKV in a lethal mouse model warrant its continued clinical development.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Purine Nucleosides/pharmacology , Purine Nucleosides/therapeutic use , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Adenine/analogs & derivatives , Adenosine/analogs & derivatives , Animals , Antiviral Agents/administration & dosage , Brain/virology , Cell Line , Disease Models, Animal , Humans , Male , Mice , Purine Nucleosides/administration & dosage , Pyrrolidines , RNA, Viral , Testis/virology , Viral Load/drug effects , Viremia , Virus Replication/drug effects , Zika Virus Infection/virology
7.
Expert Opin Pharmacother ; 17(17): 2319-2330, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27759440

ABSTRACT

INTRODUCTION: Advances in acute lymphocytic leukemia (ALL) therapy has led to long-term survival rates in children. However, only 30%-40% of adults achieve long-term disease-free survival. After relapse, the outcome of salvage chemotherapy is very disappointing with less than 10% of long survival. Novel agents are therefore desperately required to improve response rates and survival, but also the quality of life of patients. Areas covered: The following review is a comprehensive summary of various novel options reported over the past few years in the therapeutic area of adult ALL. Expert opinion: Identifying key components involved in disease pathogenesis may lead to new approaches. In a near future, the incorporation of monoclonal antibodies and T-cell directed approaches including blinatumomab and chimeric antigen receptor T cells may increase the cure rates and may reduce the need for intensive therapy.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Drug Discovery , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Purine Nucleosides/therapeutic use , Pyrimidinones/therapeutic use , Salvage Therapy/methods , Adenine Nucleotides/administration & dosage , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/administration & dosage , Arabinonucleosides/administration & dosage , Clofarabine , Disease-Free Survival , Humans , Molecular Targeted Therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Purine Nucleosides/administration & dosage , Pyrimidinones/administration & dosage , Quality of Life , Recurrence
8.
Lima; s;n; ene. 2016.
Non-conventional in Spanish | LILACS, BRISA/RedTESA | ID: biblio-847773

ABSTRACT

IINTRODUCCIÓN: Antecedentes: El presente informe expone la evaluación del medicamento clofarabina respecto a su uso en combinación con ciclofosfamida y etopósido, en pacientes menores de 21 años con leucemia leucemia linfocítica aguda recurrente y/o refractaria a dos líneas de tratamiento con donante disponible. Aspectos Generales: La leucemia aguda es el cáncer más frecuente el la población pediátrica, comprendiendo aproximadamente el 30% de todas las enfermedades malignas pedátricas. De todas las leucemias, la leucemia linfoblástica aguda (LLA) ocurre cinco veces más frecuentemente que la leucemia mieloide aguda (LMA). Tecnología Sanitaria de Interés: Clofarabina: La clofarabina es un análogo nucleósido purínico de segunbda generación. Después de varias fosforilaciones consecutivas, la clofarabina es convertida en su metabolito activo trifosfato. Se cree que la actividad anticancerígena de clofarabina trifosfato se debe a tres mecanismos: (1) la incorporación del trifosfato en el DNA por intermedio de la DNA polimerasa que conduce a sua inhibición resultadno en la detención de la elongación del DNA y/o la reparación o sínstesis del DNA, (2) inhibición de la ribo nucleótido reductasa con reducción del pool de trifosfato deoxínucleotido (dNTP), e (3) inducción de la apoptosis a través de la acción directa e indirecta en la mitocondria produciendo la liberación de citocromo C y otros factores proapottóticos. METODOLOGÍA: Estratégia de Búsqueda: Se realizó una búsqueda con respecto a la eficacia y seguridad del esquema de clofarabina + ciclofosfamida + etopósido para el tratamiento de la leucemia linfocítica aguda en pacientes pediátricos con dos o más recaídas y/o recurriecias a líneas de quimioterapia en las bases de datos de Ovid Medline y Tripdatabase. También se hizo una búsqueda adicional en www.clinicaltrials.gov, para poder identificar ensayos en desarrollo. RESULTADOS: Se realizó la búsqueda bibliográfica y de la evidencia científica para el sustento del uso del esquema combinado de clofarabina con etocósido y ciclofosfamida como tratamiento de reinducción en pacientes pediátricos con LLA y con 2 o más recaídas o resistencias al tratamiento. CONCLUSIONES: En la presente evaluación de tecnología sanitaria la evidencia es escasa respecto a la eficacia y seguridad del esquema triple de clofarabina, etopósido y ciclofosfamida en el tratamiento de pacientes pediátricos con LLA y con recaídas y/o resistencia. Se identificaron dos ensayos fase II no comparativos y cuatro estudios de series de casos, que sugieren que el esquema triple pueden lograr la remissión general (RC y RCp) en cerca de la mitad de los pacientes tratados. Sin embargo, debido a las limitaciones metodológicas de los estudios, esta evidencia es de baja calidad, significando que la confianza en los estimados es aún muy baja respecto a pacientes peidátricos con LLA que requieren tratamiento de rescate, en quienes es un desafio realizar ensayos clínicos fase III. El Instituto de Evaluación de Tecnologías en Salud e Investigación-IETSI aprueba temporalmente el uso del esquema triple de clofarabina, etopósido y ciclofosfamida para el tratamiento de pacientes pediátricos con LLA y con recaídas o resistencia. Dado que la evidencia que respalda este uso del esquema triple de clofarabina, etopósido y ciclofosfamida es aún limitadam, se establece que el efecto del esquema triple se evaluará con los datos de los pacientes que hayan recibido el esquema por el lapso de un año para determinar el impacto de su uso en varios desenlaces clínicos. Esta información será tomada en cuenta en la reevaluación de este medicamento para efectos de un nuevo dictamen al terminar la vigencia del presente Dictamen Preliminar.


Subject(s)
Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Cyclophosphamide/administration & dosage , Drug Combinations , Etoposide/administration & dosage , Purine Nucleosides/administration & dosage , Technology Assessment, Biomedical , Treatment Outcome
9.
Ann Hematol ; 95(3): 425-35, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26692089

ABSTRACT

Recently, great progress has been achieved in the treatment of chronic lymphocytic leukemia (CLL). However, some patients, particularly older patients with comorbidities or with relapsed/refractory leukemia, still have limited therapeutic options. There is an urgent need to discover less toxic and more effective drugs for CLL patients. Applying new modalities or substances that are widely used for the treatment of other diseases has been reported to improve results in CLL treatment. This study aimed to assess the non-chemotherapeutic drug danazol for its potential to destroy leukemic cells. Leukemic cells, obtained from the peripheral blood and bone marrow of 23 CLL patients, were cultured in the presence of danazol and its combination with the purine nucleoside analogs fludarabine and cladribine and bendamustine. After 24 h of incubation, the rate of apoptosis indicated by active caspase-3 expression, and cytotoxicity indicated by forward light scatter and light scatter analysis, was assessed by flow cytometry. We also measured expression of apoptosis-regulating proteins of BCL family and active caspase 9 and active caspase 8 expressions in leukemic cells. Danazol had a caspase-dependent pro-apoptotic and cytotoxic effect on leukemic cells in a tumor-specific manner. The mechanisms of its action appear to be complex and should be precisely established; however, induction of apoptosis involving both mitochondrial and receptor cascades appears to be most probable. Danazol showed a synergic effect with cladribine, an additive effect with fludarabine, and an infra-additive effect with bendamustine. The rate of danazol-induced apoptosis and cytotoxicity did not differ between patients with better and worse prognostic markers. Our results indicate that danazol may be a potential therapeutic agent for CLL patients alone and in combination with purine analogs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Cytotoxins/administration & dosage , Danazol/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Purine Nucleosides/administration & dosage , Adult , Aged , Aged, 80 and over , Apoptosis/physiology , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Purine Nucleosides/chemistry , Tumor Cells, Cultured
10.
Front Med ; 9(1): 82-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25523750

ABSTRACT

Our preliminary studies demonstrated that Metacavir has potential to become a new anti-HBV agent. The main targets of the toxic effects of Metacavir, in rhesus monkeys, were gastrointestinal tracts, liver, blood, and kidneys, which were not related to mitochondrial effects. In this study, the maternal toxicity, embryo-fetal developmental toxicity and teratogenicity were studied in pregnant Sprague-Dawley rats after intragastric administration of Metacavir (200, 100, 50, 0 mg/kg body weight) during the first 6-15 days of pregnancy. Slower weight gain was observed in 5 out of 21 rats subjected to a 200 mg/kg dose, as well as 2 out of 20 subjected to a 100 mg/kg dose. Compared with the solvent control group, the calibration weight gain in the 200 mg/kg and 100 mg/kg dosage groups respectively, during first 6-20 pregnant days were significantly different (P < 0.01, P < 0.05). Significant dose related adverse effects to other reproductive parameters were not seen in F0 and F1, but the number of stillbirths in high dose group showed notably difference compared with the control group (P < 0.05), while the litter incidence showed no difference. No Metacavir-associated pathological changes were observed. The present research indicated that at a dose of 200 mg/(kg·d) (i.e., 40 times the effective dose in rats), Metacavir shows some maternal toxicity to SD rats. The embryotoxicity in the 200 mg/kg group encompass decreased fetal body weight, and higher fetal mortality rates, compared with the control group. However, the litter incidence showed no statistical difference. All the treated rats displayed normal bone development, no teratogenicity and without adverse effects on fetal development, thus indicating that below a dose of 200 mg/(kg·d) there is no teratogenic side effects.


Subject(s)
Hepatitis B/drug therapy , Purine Nucleosides , Reproduction/drug effects , Teratogenesis/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Pregnancy , Purine Nucleosides/administration & dosage , Purine Nucleosides/toxicity , Rats , Rats, Sprague-Dawley , Treatment Outcome , Weight Gain/drug effects
11.
Nature ; 508(7496): 402-5, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24590073

ABSTRACT

Filoviruses are emerging pathogens and causative agents of viral haemorrhagic fever. Case fatality rates of filovirus disease outbreaks are among the highest reported for any human pathogen, exceeding 90% (ref. 1). Licensed therapeutic or vaccine products are not available to treat filovirus diseases. Candidate therapeutics previously shown to be efficacious in non-human primate disease models are based on virus-specific designs and have limited broad-spectrum antiviral potential. Here we show that BCX4430, a novel synthetic adenosine analogue, inhibits infection of distinct filoviruses in human cells. Biochemical, reporter-based and primer-extension assays indicate that BCX4430 inhibits viral RNA polymerase function, acting as a non-obligate RNA chain terminator. Post-exposure intramuscular administration of BCX4430 protects against Ebola virus and Marburg virus disease in rodent models. Most importantly, BCX4430 completely protects cynomolgus macaques from Marburg virus infection when administered as late as 48 hours after infection. In addition, BCX4430 exhibits broad-spectrum antiviral activity against numerous viruses, including bunyaviruses, arenaviruses, paramyxoviruses, coronaviruses and flaviviruses. This is the first report, to our knowledge, of non-human primate protection from filovirus disease by a synthetic drug-like small molecule. We provide additional pharmacological characterizations supporting the potential development of BCX4430 as a countermeasure against human filovirus diseases and other viral diseases representing major public health threats.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/pharmacology , Filoviridae Infections/prevention & control , Filoviridae Infections/virology , Filoviridae/drug effects , Purine Nucleosides/pharmacology , Adenine/analogs & derivatives , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Disease Models, Animal , Ebolavirus/drug effects , Filoviridae/enzymology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Humans , Injections, Intramuscular , Macaca fascicularis/virology , Marburg Virus Disease/prevention & control , Marburg Virus Disease/virology , Marburgvirus/drug effects , Purine Nucleosides/administration & dosage , Purine Nucleosides/chemistry , Purine Nucleosides/pharmacokinetics , Pyrrolidines , RNA/biosynthesis , Time Factors
12.
PLoS One ; 8(1): e54044, 2013.
Article in English | MEDLINE | ID: mdl-23326571

ABSTRACT

Achieving an effective treatment of cancer is difficult, particularly when resistance to conventional chemotherapy is developed. P-glycoprotein (P-gp) activity governs multi-drug resistance (MDR) development in different cancer cell types. Identification of anti-cancer agents with the potential to kill cancer cells and at the same time inhibit MDR is important to intensify the search for novel therapeutic approaches. We examined the effects of sulfinosine (SF), a quite unexplored purine nucleoside analog, in MDR (P-gp over-expressing) non-small cell lung carcinoma (NSCLC) and glioblastoma cell lines (NCI-H460/R and U87-TxR, respectively). SF showed the same efficacy against MDR cancer cell lines and their sensitive counterparts. However, it was non-toxic for normal human keratinocytes (HaCaT). SF induced caspase-dependent apoptotic cell death and autophagy in MDR cancer cells. After SF application, reactive oxygen species (ROS) were generated and glutathione (GSH) concentration was decreased. The expression of key enzyme for GSH synthesis, gamma Glutamyl-cysteine-synthetase (γGCS) was decreased as well as the expression of gst-π mRNA. Consequently, SF significantly decreased the expression of hif-1α, mdr1 and vegf mRNAs even in hypoxic conditions. SF caused the inhibition of P-gp (coded by mdr1) expression and activity. The accumulation of standard chemotherapeutic agent--doxorubicin (DOX) was induced by SF in concentration- and time-dependent manner. The best effect of SF was obtained after 72 h when it attained the effect of known P-gp inhibitors (Dex-verapamil and tariquidar). Accordingly, SF sensitized the resistant cancer cells to DOX in subsequent treatment. Furthermore, SF decreased the experssion of vascular endothelial growth factor (VEGF) on mRNA and protein level and modulated its secretion. In conclusion, the effects on P-gp (implicated in pharmacokinetics and MDR), GSH (implicated in detoxification) and VEGF (implicated in tumor-angiogenesis and progression) qualify SF as multi-potent anti-cancer agent, which use must be considered, in particular for resistant malignancies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Transformation, Neoplastic/drug effects , Drug Resistance, Multiple/drug effects , Glioblastoma/drug therapy , Purine Nucleosides/administration & dosage , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Resistance, Multiple/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione/biosynthesis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/genetics
13.
J Pain Symptom Manage ; 43(4): 679-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21983265

ABSTRACT

CONTEXT: Neuropathic pain in patients with cancer can be difficult to treat effectively. OBJECTIVES: The purpose of the study was to determine safety and efficacy of KRN5500, a novel, spicamycin-derived, nonopioid analgesic agent, in patients with advanced cancer and neuropathic pain of any etiology. METHODS: The study was a Phase 2a, multicenter, double-blind, placebo-controlled, dose escalation clinical trial. Patients with refractory neuropathic pain and advanced cancer were randomly assigned 2:1 to receive a maximum of eight single escalating doses of KRN5500 or placebo, ranging from 0.6 to 2.2 mg/m(2). The primary objective was safety and tolerability. The secondary objective was efficacy, measured by change in average pain intensity on a 0-10 numeric rating scale administered one week after the patient's final dose. RESULTS: Nineteen patients received treatment (KRN5500 n=12; placebo n=7). The most frequently reported adverse events were gastrointestinal symptoms, which were more frequent and severe with KRN5500 than placebo; two (17%) KRN5500 patients discontinued the study because of nausea and vomiting. At study endpoint, KRN5500 exhibited a significant median decrease in pain intensity from baseline of 24% compared with 0% for placebo (P=0.03). The median for largest weekly reduction in target pain intensity was 29.5% for KRN5500 and 0% for placebo patients (P=0.02). CONCLUSION: This proof-of-concept study for KRN5500 in patients with advanced cancer and any type of neuropathic pain found gastrointestinal adverse events to be the predominant safety concern. The results also provided the first indication of clinical and statistical efficacy in reducing pain intensity.


Subject(s)
Neoplasms/complications , Neoplasms/drug therapy , Neuralgia/etiology , Neuralgia/prevention & control , Pain Measurement/drug effects , Spiramycin/analogs & derivatives , Terminal Care/methods , Adult , Analgesics/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/diagnosis , Neuralgia/diagnosis , Pilot Projects , Placebo Effect , Purine Nucleosides/administration & dosage , Purine Nucleosides/adverse effects , Spiramycin/adverse effects , Spiramycin/therapeutic use , Treatment Outcome
14.
Blood ; 116(6): 886-92, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20427701

ABSTRACT

Forodesine is a new and potent purine nucleoside phosphorylase (PNP) inhibitor. Patients with chronic lymphocytic leukemia (CLL) with primary resistance to fludarabine-based therapy or with progressive disease were eligible for oral forodesine (200 mg/d) for up to 24 weeks. Eight patients with median lymphocyte count of 35.9 x 10(9)/L and median serum beta2 microglobulin level of 6.45 mg/L were treated. Six had Rai stage III to IV and were previously heavily treated (median prior therapy = 5). Two had transient decrease in lymphocyte count to normal, whereas in 5, disease progressed. Adverse events were mild. Steady-state level of forodesine ranged from 200 to 1300 nM and did not reach desired 2 microM level. PNP inhibition ranged from 57% to 89% and steady-state 2'-deoxyguanosine (dGuo) concentration median was 1.8 microM. Intracellular deoxyguanosine triphosphate (dGTP) increase was very modest, from median of 6 microM to 10 microM. Compared with in vivo, in vitro incubations of CLL lymphocytes with 10 or 20 microM dGuo and forodesine (2 microM) resulted in accumulation of higher levels of dGTP (40-250 microM) which resulted in increase in apoptosis. Forodesine has biologic activity in CLL; pharmacodynamic parameters suggest that an alternate dosing schedule and/or higher doses to achieve greater intracellular dGTP may be beneficial in this patient population.


Subject(s)
Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacokinetics , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Vidarabine/analogs & derivatives , Administration, Oral , Aged , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Drug Administration Schedule , Drug Therapy, Combination , Enzyme Inhibitors/blood , Female , Follow-Up Studies , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphocyte Count , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Middle Aged , Phosphoric Monoester Hydrolases/metabolism , Purine Nucleosides/blood , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/metabolism , Pyrimidinones/blood , Severity of Illness Index , Vidarabine/administration & dosage
15.
Antivir Ther ; 15(2): 185-92, 2010.
Article in English | MEDLINE | ID: mdl-20386073

ABSTRACT

BACKGROUND: Amdoxovir acts synergistically with zidovudine in vitro and the combination prevents or delays the selection of thymidine analogue and K65R mutations. In silico studies have shown that a reduced dose of zidovudine (200 mg) results in decreased zidovudine-monophosphate levels, associated with toxicity, while maintaining zidovudine-triphosphate levels, which are associated with antiviral effects. Here, we aimed to assess the short-term tolerability and antiviral activity of amdoxovir in combination with reduced and standard doses of zidovudine. METHODS: The study was a double-blind, placebo-controlled study in HIV-1-infected patients not receiving antiretroviral therapy and with plasma HIV-1 RNA > or =5,000 copies/ml. Patients were randomized to 10 days of twice-daily treatment with 200 mg zidovudine, 300 mg zidovudine, 500 mg amdoxovir, 500 mg amdoxovir plus 200 mg zidovudine or 500 mg amdoxovir plus 300 mg zidovudine. The mean change in viral load (VL) log(10) and area under the virus depletion curve (AUC(VL)) from baseline to day 10 were determined. Laboratory and clinical safety monitoring were performed. RESULTS: Twenty-four patients were enrolled. The mean VL log(10) change was 0.10 with placebo, -0.69 with zidovudine 200 mg, -0.55 with zidovudine 300 mg, -1.09 with amdoxovir, -2.00 with amdoxovir plus zidovudine (200 mg) and -1.69 with amdoxovir plus zidovudine (300 mg). Amdoxovir plus zidovudine (200 mg) was significantly more potent than amdoxovir monotherapy in AUC(VL) and mean VL decline (P=0.019 and P=0.021, respectively), suggesting synergy. There was markedly decreased VL variability with the combination compared with amdoxovir alone. All adverse events were mild to moderate. CONCLUSION: The combination of amdoxovir plus zidovudine appeared synergistic with reduced VL variability. This combined therapy, including the use of a lower zidovudine dosage, warrants further development for the therapy of HIV infection.


Subject(s)
Anti-HIV Agents/therapeutic use , Dioxolanes/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , Purine Nucleosides/therapeutic use , Reverse Transcriptase Inhibitors/therapeutic use , Zidovudine/therapeutic use , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacology , Dioxolanes/administration & dosage , Dioxolanes/adverse effects , Dioxolanes/pharmacology , Double-Blind Method , Drug Therapy, Combination , Female , HIV Infections/virology , Humans , Male , Middle Aged , Purine Nucleosides/administration & dosage , Purine Nucleosides/adverse effects , Purine Nucleosides/pharmacology , RNA, Viral/blood , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/pharmacology , Treatment Outcome , Viral Load , Young Adult , Zidovudine/administration & dosage , Zidovudine/adverse effects , Zidovudine/pharmacology
16.
Antimicrob Agents Chemother ; 54(3): 1248-55, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20038617

ABSTRACT

Amdoxovir (AMDX) inhibits HIV-1 containing the M184V/I mutation and is rapidly absorbed and deaminated to its active metabolite, beta-D-dioxolane guanosine (DXG). DXG is synergistic with zidovudine (ZDV) in HIV-1-infected primary human lymphocytes. A recent in silico pharmacokinetic (PK)/enzyme kinetic study suggested that ZDV at 200 mg twice a day (b.i.d.) may reduce toxicity without compromising efficacy relative to the standard 300-mg b.i.d. dose. Therefore, an intense PK clinical study was conducted using AMDX/placebo, with or without ZDV, in 24 subjects randomized to receive oral AMDX at 500 mg b.i.d., AMDX at 500 mg plus ZDV at 200 or 300 mg b.i.d., or ZDV at 200 or 300 mg b.i.d. for 10 days. Full plasma PK profiles were collected on days 1 and 10, and complete urine sampling was performed on day 9. Plasma and urine concentrations of AMDX, DXG, ZDV, and ZDV-5'-O-glucuronide (GZDV) were measured using a validated liquid chromatography-tandem mass spectrometry method. Data were analyzed using noncompartmental methods, and multiple comparisons were performed on the log-transformed parameters, at steady state. Coadministration of AMDX with ZDV did not significantly change either of the plasma PK parameters or percent recovery in the urine of AMDX, DXG, or ZDV/GZDV. Larger studies with AMDX/ZDV, with a longer duration, are warranted.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Dioxolanes/pharmacokinetics , Drug Interactions , HIV Infections/drug therapy , Purine Nucleosides/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Zidovudine/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Dioxolanes/administration & dosage , Drug Administration Schedule , Female , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged , Purine Nucleosides/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage , Treatment Outcome , Young Adult , Zidovudine/administration & dosage
17.
Acta Pharmacol Sin ; 30(12): 1666-73, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19915582

ABSTRACT

AIM: To explore the potential mitochondrial toxicities and their severities of intravenously administered metacavir, a nucleoside analog, in rhesus monkeys. METHODS: Totally 21 rhesus monkeys were randomly divided into 4 groups: metacavir 120 mg/kg group, metacavir 40 mg/kg group, zidovudine(AZT) 50 mg/kg group, and blank control group. Animals were killed after the completion of dosing or further observed in a 4-week recovery phase. Changes of structure of mitochondria in liver, kidney, skeletal muscles, and cardiac muscles were observed under transmission electron microscope(TEM). Changes of the activities of mitochondrial respiratory chain complexes and mitochondrial DNA were also determined. RESULTS: In metacavir 120 mg/kg group, some mitochondrial injuries were found in skeletal muscle, cardiac muscle, and liver, including that some cristae was broken and became sparse in density in the skeletal muscle, the morphology and size of mitochondria remained unchanged. Metacavir decreased the activities of respiratory chain complexes I and II and the mtDNA contents in three tissues in a dose-dependent manner; however, the extent of such decrease was lower than that in AZT 50 mg/kg group. The mitochondrial injuries in metacavir 40 mg/kg group were mild in each tissue and no obvious change in mitochondrial function was noted. On week 4 in the recovery phase, results showed that all these injuries were reversible after drug withdrawal. CONCLUSION: These results suggest that metacavir has not a high risk for potential mitochondrial-related effects in rhesus monkeys.


Subject(s)
DNA, Mitochondrial/metabolism , Electron Transport Complex II/metabolism , Electron Transport Complex I/metabolism , Mitochondria/drug effects , Purine Nucleosides/agonists , Animals , Female , Heart/drug effects , Injections, Intravenous , Kidney/drug effects , Kidney/metabolism , Kidney/ultrastructure , Liver/drug effects , Liver/metabolism , Liver/ultrastructure , Macaca mulatta , Male , Mitochondria/physiology , Mitochondria/ultrastructure , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myocardium/metabolism , Myocardium/ultrastructure , Purine Nucleosides/administration & dosage , Zidovudine/pharmacology
18.
Blood ; 114(8): 1563-75, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19541822

ABSTRACT

Chronic lymphocytic leukemia (CLL) is an incurable disease derived from the monoclonal expansion of CD5(+) B lymphocytes. High expression levels of ZAP-70 or CD38 and deletions of 17p13 (TP53) and 11q22-q23 (ATM) are associated with poorer overall survival and shorter time to disease progression. DNA damage and p53 play a pivotal role in apoptosis induction in response to conventional chemotherapy, because deletions of ATM or p53 identify CLL patients with resistance to treatment. Forodesine is a transition-state inhibitor of the purine nucleoside phosphorylase with antileukemic activity. We show that forodesine is highly cytotoxic as single agent or in combination with bendamustine and rituximab in primary leukemic cells from CLL patients regardless of CD38/ZAP-70 expression and p53 or ATM deletion. Forodesine activates the mitochondrial apoptotic pathway by decreasing the levels of antiapoptotic MCL-1 protein and induction of proapoptotic BIM protein. Forodesine induces transcriptional up-regulation of p73, a p53-related protein able to overcome the resistance to apoptosis of CLL cells lacking functional p53. Remarkably, no differences in these apoptotic markers were observed based on p53 or ATM status. In conclusion, forodesine induces apoptosis of CLL cells bypassing the DNA-damage/ATM/p53 pathway and might represent a novel chemotherapeutic approach that deserves clinical investigation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/drug effects , DNA-Binding Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Membrane Proteins/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Purine Nucleosides/pharmacology , Pyrimidinones/pharmacology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/genetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Bendamustine Hydrochloride , Cyclophosphamide/administration & dosage , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondria/physiology , Nitrogen Mustard Compounds/administration & dosage , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Purine Nucleosides/administration & dosage , Purine Nucleosides/therapeutic use , Pyrimidinones/administration & dosage , Pyrimidinones/therapeutic use , Rituximab , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
19.
Invest New Drugs ; 27(2): 99-110, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18493718

ABSTRACT

A resistant non-small cell lung carcinoma cell line-NSCLC (NCI-H460/R) was established in order to investigate the potential of sulfinosine (SF) to overcome multidrug resistance (MDR). The cytotoxicity of doxorubicin (DOX) in NCI-H460/R cells was enhanced by interaction with SF. SF improved the sensitivity of resistant cells to DOX when NCI-H460/R cells were pretreated with SF. Synergism was accompanied by the accumulation of cells in S and G(2)/M phases. Pretreatment with SF was more potent in improving the sensitivity to DOX than verapamil (VER). The decrease of mdr1 and topo II alpha expression (assessed by RT-PCR), was consistent with the DOX accumulation assay and cell cycle analysis. Also, SF significantly decreased intracellular glutathione (GSH) concentration. These results point to SF as a potential agent of MDR reversal and a valuable drug for improving chemotherapy of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Lung Neoplasms/drug therapy , Purine Nucleosides/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Binding, Competitive , Cell Cycle/drug effects , Cell Line, Tumor , Cysteine/metabolism , DNA Topoisomerases, Type II/metabolism , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Glutathione/metabolism , Humans , Multidrug Resistance-Associated Proteins/metabolism , Purine Nucleosides/administration & dosage , Purine Nucleosides/chemistry
20.
Cancer Biol Ther ; 7(7): 1024-32, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18414057

ABSTRACT

Multidrug resistance (MDR) is the main obstacle to a successful chemotherapy of lung cancer. We tested the potential of sulfinosine and curcumin, alone and in combination, for modulating MDR in the human resistant, non-small cell lung carcinoma cell line (NCI-H460/R). First, we determined the mutational status of the p53 gene in NCI-H460/R cells by PCR-SSCP and DNA sequencing and identified mutations which could at least partially contribute to the development of the MDR phenotype. The effects of sulfinosine and curcumin were studied, both separately and in combination, at the level of cytotoxicity, cell cycle distribution and gene expression. Sulfinosine displayed dose-dependent growth inhibition in both resistant and control sensitive cell lines, whereas curcumin considerably inhibited their growth only at relatively high doses. When sulfinosine was combined with a low dose of curcumin the drugs exerted a synergistic cytotoxic effect in NCI-H460/R cells. The expression of MDR-related genes mdr1, gst-pi and topo IIalpha, was altered by sulfinosine and curcumin. The most pronounced effect was observed when the agents were applied together. Sulfinosine and curcumin caused perturbations in cell cycle distribution in the NCI-H460/R cell line. The combination of the two drugs induced a more pronounced cell cycle arrest in S and G(2)/M in NCI-H460/R cells. Our results show that sulfinosine and curcumin overcome MDR in non-small cell lung carcinoma cell line (NSCLC), especially in combination despite the presence of a mutated p53 gene.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Curcumin/administration & dosage , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Purine Nucleosides/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Exons , Genes, p53 , Humans , Mutation , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...