Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Best Pract Res Clin Haematol ; 34(4): 101326, 2021 12.
Article in English | MEDLINE | ID: mdl-34865698

ABSTRACT

The traditional cytotoxic induction regimen for acute myeloid leukemia (AML) is seven days of standard-dose cytarabine and three days of an anthracycline antibiotic (such as daunorubicin or idarubicin), commonly known as "7 + 3." Many studies have been conducted to find an additional agent that might improve efficacy. Data from select studies has shown, in certain populations, benefit to adding cladribine, clofarabine and lomustine to a traditional backbone. For mutation-based chemotherapy regimens, midostaurin with 7 + 3 is the current standard of care for FLT3-mutant, younger AML patients. As we learn more about the synergism of molecular agents and traditional anti-cancer treatments, we can hopefully develop novel regimens without abandoning some of the benefits of these mutation agnostic historical therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, Myeloid, Acute , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Humans , Idarubicin/therapeutic use , Induction Chemotherapy , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Remission Induction
2.
Cancer ; 127(8): 1186-1207, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33734442

ABSTRACT

The unraveling of the pathophysiology of acute myeloid leukemia (AML) has resulted in rapid translation of the information into clinical practice. After more than 40 years of slow progress in AML research, the US Food and Drug Administration has approved nine agents for different AML treatment indications since 2017. In this review, we detail the progress that has been made in the research and treatment of AML, citing key publications related to AML research and therapy in the English literature since 2000. The notable subsets of AML include acute promyelocytic leukemia (APL), core-binding factor AML (CBF-AML), AML in younger patients fit for intensive chemotherapy, and AML in older/unfit patients (usually at the age cutoff of 60-70 years). We also consider within each subset whether the AML is primary or secondary (therapy-related, evolving from untreated or treated myelodysplastic syndrome or myeloproliferative neoplasm). In APL, therapy with all-trans retinoic acid and arsenic trioxide results in estimated 10-year survival rates of ≥80%. Treatment of CBF-AML with fludarabine, high-dose cytarabine, and gemtuzumab ozogamicin (GO) results in estimated 10-year survival rates of ≥75%. In younger/fit patients, the "3+7" regimen (3 days of daunorubicin + 7 days of cytarabine) produces less favorable results (estimated 5-year survival rates of 35%; worse in real-world experience); regimens that incorporate high-dose cytarabine, adenosine nucleoside analogs, and GO are producing better results. Adding venetoclax, FLT3, and IDH inhibitors into these regimens has resulted in encouraging preliminary data. In older/unfit patients, low-intensity therapy with hypomethylating agents (HMAs) and venetoclax is now the new standard of care. Better low-intensity regimens incorporating cladribine, low-dose cytarabine, and other targeted therapies (FLT3 and IDH inhibitors) are emerging. Maintenance therapy now has a definite role in the treatment of AML, and oral HMAs with potential treatment benefits are also available. In conclusion, AML therapy is evolving rapidly and treatment results are improving in all AML subsets as novel agents and strategies are incorporated into traditional AML chemotherapy. LAY SUMMARY: Ongoing research in acute myeloid leukemia (AML) is progressing rapidly. Since 2017, the US Food and Drug Administration has approved 10 drugs for different AML indications. This review updates the research and treatment pathways for AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Age Factors , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic Trioxide/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cladribine/therapeutic use , Core Binding Factors , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Gemtuzumab/therapeutic use , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/mortality , Maintenance Chemotherapy , Mutation , Myelodysplastic Syndromes/complications , Myeloproliferative Disorders/complications , Neoplasm, Residual , Sulfonamides/therapeutic use , Survival Rate , Translational Research, Biomedical , Tretinoin/therapeutic use , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
3.
Bogotá; IETS; oct. 2017.
Non-conventional in Spanish | BRISA | ID: biblio-1395388

ABSTRACT

INTRODUCCIÓN: El análisis de impacto presupuestal (AIP) de los medicamentos quimioterapéuticos para el tratamiento de cáncer en Colombia, se desarrolló en el marco del mecanismo técnico científico para la ampliación progresiva del Plan de Beneficios en Salud con cargo a la UPC (PBSUPC) y la definición de la lista de exclusiones, establecido en el artículo 15 de la Ley 1751 de 2015. La quimioterapia tiene un gran impacto en el tratamiento oncológico, la cual es indispensable por su valor terapéutico en varios tipos de cáncer. Esta tecnología puede ser usada sola o junto con otros tratamientos, tales como la cirugía o la radioterapia. La quimioterapia engloba a una gran variedad de fármacos y su objetivo es destruir las células tumorales con el fin de lograr la reducción de la enfermedad, los medicamentos empleados en este tipo de tratamiento se les denomina fármacos antineoplásicos. Cada tipo de tumor canceroso tiene una determinada sensibilidad a estos medicamentos, por lo tanto, es frecuente que el mismo fármaco se pueda emplear en el tratamiento de distintos tumores, variando las dosis o asociándolo a otros fármacos distintos. La quimioterapia puede ser administrada con fines curativos o para aliviar los síntomas y prolongar la supervivencia. La forma de administración de la quimioterapia es por ciclos y esto se logra alternando los periodos de tratamiento con periodos de descanso. Un ciclo es, por lo tanto, el periodo de administración del tratamiento y el de descanso hasta la siguiente administración. El objetivo de este análisis de impacto presupuestal (AIP) es estimar el esfuerzo financiero necesario para la adopción de la quimioterapia en el tratamiento de pacientes con cáncer en Colombia, en un horizonte temporal de tres años. Este documento está conformado por cuatro secciones: en la primera se identifican las tecnologías a evaluar, en la segunda sección se especifica la perspectiva, horizonte temporal y la población sobre la cual se realizó el AIP; en la sección tres se detallan los costos utilizados en el modelo, además de los escenarios planteados por los investigadores; por último, en la sección cuatro se exponen los resultados en los diferentes escenarios planteados Este documento describe la metodología desarrollada para realizar el análisis de impacto presupuestal de 21 tecnologías para el manejo quimioterapéutico del cáncer en Colombia Este informe, sigue los lineamientos propuestos en el Manual para la Elaboración de Análisis de Impacto Presupuestal y en Manual de Participación y Deliberación publicados por IETS. Insumos y método: Esta sección presenta los supuestos, parámetros y métodos utilizados para el modelo de estimación del impacto presupuestal describiendo la siguiente información: Perspectiva: La perspectiva de este AIP es la del tercer pagador el cual en nuestro contexto es el Sistema General de Seguridad Social en Salud (SGSSS). Horizonte temporal: El horizonte temporal de este AIP en el caso base corresponde a un año. Adicionalmente se reportan las estimaciones del impacto presupuestal para los años 2 y 3, bajo el supuesto de inclusión en el PBS en el año 1. Población total: Para el desarrollo de este AIP se parte de la población general afiliada al SGSSS colombiano sin distinción de sexo o edad. ESCENARIOS: Se consideró para la formulación de los escenarios de adopción de las tecnologías evaluadas los siguientes aspectos: 1. Los medicamentos evaluados no son alternativas terapéuticas para las patologías observadas, estas tecnologías sanitarias hacen parte de los protocolos de tratamiento con evidencia científica suficiente que garantizan su efectividad y seguridad clínica y que actualmente se encuentran en las opciones de tratamiento utilizados en la práctica clínica colombiana. 2. Al ser esquemas de tratamiento que hacen parte de protocolos estandarizados de aplicación, sí alguno de los medicamentos es sujeto de recobros ante ADRES, este trámite puede generar barreras de acceso al tratamiento hasta que se efectué la respectiva aprobación. Por lo tanto, no hay certeza de la efectividad clínica si los esquemas de tratamiento son suministrados de forma parcial o incompleta. 3. La elección del esquema de tratamiento obedece a criterios clínicos y a las características evaluadas en el paciente, no se espera una sustitución entre los diferentes esquemas sí se realiza un cambio en el mecanismo de financiamiento. 4. La adopción de las tecnologías evaluadas en este AIP no se espera que se modifiquen de manera importante, debido a que estas tecnologías hacen parte integral de los esquemas de tratamiento y su incorporación en la práctica clínica habitual en el contexto internacional y nacional, data de aproximadamente 10 a 5 años. Tambieén son parte de las opciones de primera línea de tratamiento para estadios tempranos, avanzados y localmente avanzados del paciente diagnosticado con câncer. De acuerdo a las anteriores consideraciones, al incorporar los medicamentos evaluados al PBS con cargo a la UPC, se espera la misma composición del mercado con la adopción de los nuevos medicamentos en el 100% de los tratamientos esperados en la siguiente anualidad. Los resultados esperados en el sistema de salud, en este cambio de financiamiento, se esperan obtener en dos puntos: a) En una mejor oportunidad de acceso a los esquemas de tratamiento en el SGSSS (25). b) En una mejora en la cobertura efectiva de los tratamientos de quimioterapia en pacientes con diagnóstico de cáncer. RESULTADOS: Se muestra el resultado consolidado para las ventiun tecnologías objeto del Análisis de Impacto Presupuestal. La tecnología que genera un mayor impacto es Oxaliplatino, con un valor por persona de $2.363.250,76 usada en 3170 pacientes, para un total de $7.491.504.923,90. El Megestrol es la tecnología con menor impacto, con un costo por persona de $ 383.791,06 y siendo usada en 34 pacientes, tiene un valor total de $ 13.048.896,00. La tretinoina es la tecnología más económica por paciente, con un valor de $ 97.996,50, es usada en 242 personas para un total de $ 23.715.153,00. DISCUSIÓN: En la práctica actual existe un volumen amplio de recobros en el caso de estos medicamentos por usos UNIRS. En algunos casos, los cambios en el mercado farmacéutico, ya sea por el retiro de medicamentos o la llegada de ellos, hace que se modifique indicaciones ya existentes en los registros y que pueden llegar a impactar estos. usos, por ejemplo aquellos casos en los que existe la indicación antineplásico y se cambian por indicaciones especificas, que pueden no considerar condiciones de salud de baja incidencia. Como se ha caracterizado con anterioridad, el mercado de tecnologías sanitarias que se encuentran incluidas al plan de beneficios en salud con cargo a la UPC difiere sustancialmente al mercado de tecnologías sanitarias aún no financiadas por dicho mecanismo. La existencia de las Empresas Administradoras de Planes de Beneficios (EAPB) presume la existencia de un actor que al maximizar su beneficio, es un buen negociador que en cumplimiento de los principios del SGSSS, llega a un precio de equilibrio que maximiza el beneficio social. En cambio, los medicamentos que son sujetos a recobros al ADRES presume un precio fuera de aquel nivel en donde se maximiza al beneficio social, en la medida que no hay una función clara de monopsonio que coteje y negocie un precio de adquisición. En algunos casos puede llegar asumir sobrecostos que las EAPB al ser intermediarias, no tienen incentivos para efectuar un adecuado control. Con el objetivo de estimar el resultado de la incorporación de estos medicamentos al PBS con cargo a la UPC, se asumieron dos escenarios en los cuales la población objetivo del AIP se consideró constante y se asumieron los siguientes supuestos: En el primer escenario se asume que los precios observados en recobros serán el promedio de todas las transacciones de compra en la siguiente anualidad. En el segundo escenario los precios promedio de adquisición de los medicamentos evaluados, corresponden al promedio observado en SISMED como predictor de los precios de equilibrio que pueden generar las EAPB como ente negociador. Se asume que, en promedio, las EAPB son negociadores eficientes que se acercan a un precio de equilibrio que maximiza el bienestar social. Se asume que la población objetivo corresponde al total de posibles pacientes que requieren las tecnologías sanitarias en evaluación, sin que exista demanda insatisfecha para estos esquemas de tratamiento. Para su cálculo, como se presenta en la tabla 09 de los servicios prestados durante el año 2015 y recobrados al FOSYGA en los años 2015 y 2016, se calculó un valor per-cápita de acuerdo con el identificador (cedula de ciudadanía anonimizada) registrado en cada recobro. Luego, este valor es indexado a precios 2016 con el IPC reportado por el DANE a diciembre 31 del año 2015. Este valor será el comparador del precio calculado para cada uno de los medicamentos a partir de SISMED 2016.


Subject(s)
Humans , Tretinoin/therapeutic use , Epirubicin/therapeutic use , Idarubicin/therapeutic use , Carmustine/therapeutic use , Daunorubicin/therapeutic use , Mitoxantrone/therapeutic use , Mitomycin/therapeutic use , Mesna/therapeutic use , Megestrol Acetate/therapeutic use , Dactinomycin/therapeutic use , Capecitabine/therapeutic use , Filgrastim/therapeutic use , Docetaxel/therapeutic use , Irinotecan/therapeutic use , Oxaliplatin/therapeutic use , Vinorelbine/therapeutic use , Hydroxyurea/therapeutic use , Ifosfamide/therapeutic use , Melphalan/therapeutic use , Neoplasms/drug therapy , Health Evaluation/economics , Efficacy , Colombia
4.
BMC Infect Dis ; 16(1): 621, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27809794

ABSTRACT

BACKGROUND: Bacillus anthracis, the causative agent of anthrax, is a spore forming and toxin producing rod-shaped bacterium that is classified as a category A bioterror agent. This pathogenic microbe can be transmitted to both animals and humans. Clinical presentation depends on the route of entry (direct contact, ingestion, injection or aerosolization) with symptoms ranging from isolated skin infections to more severe manifestations such as cardiac or pulmonary shock, meningitis, and death. To date, anthrax is treatable if antibiotics are administered promptly and continued for 60 days. However, if treatment is delayed or administered improperly, the patient's chances of survival are decreased drastically. In addition, antibiotics are ineffective against the harmful anthrax toxins and spores. Therefore, alternative therapeutics are essential. In this review article, we explore and discuss advances that have been made in anthrax therapy with a primary focus on alternative pre-approved and novel antibiotics as well as anti-toxin therapies. METHODS: A literature search was conducted using the University of Manitoba search engine. Using this search engine allowed access to a greater variety of journals/articles that would have otherwise been restricted for general use. In order to be considered for discussion for this review, all articles must have been published later than 2009. RESULTS: The alternative pre-approved antibiotics demonstrated high efficacy against B. anthracis both in vitro and in vivo. In addition, the safety profile and clinical pharmacology of these drugs were already known. Compounds that targeted underexploited bacterial processes (DNA replication, RNA synthesis, and cell division) were also very effective in combatting B. anthracis. In addition, these novel compounds prevented bacterial resistance. Targeting B. anthracis virulence, more specifically the anthrax toxins, increased the length of which treatment could be administered. CONCLUSIONS: Several novel and pre-existing antibiotics, as well as toxin inhibitors, have shown increasing promise. A combination treatment that targets both bacterial growth and toxin production would be ideal and probably necessary for effectively combatting this armed bacterium.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Antitoxins/therapeutic use , Alpha-Globulins/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, Bacterial , Bacillus anthracis , Bacterial Toxins , DNA Helicases/antagonists & inhibitors , Daunorubicin/analogs & derivatives , Daunorubicin/therapeutic use , Doxorubicin/therapeutic use , Drug Discovery , Fluoroquinolones , Humans , Interferon Inducers/therapeutic use , Levofloxacin , Linezolid , Moxifloxacin , Ofloxacin , Polyketides/therapeutic use , Serine Proteinase Inhibitors/therapeutic use , Tilorone/therapeutic use , Virulence
5.
Lancet Oncol ; 16(16): 1691-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26549589

ABSTRACT

BACKGROUND: Preclinical data and results from non-randomised trials suggest that the multikinase inhibitor sorafenib might be an effective drug for the treatment of acute myeloid leukaemia. We investigated the efficacy and tolerability of sorafenib versus placebo in addition to standard chemotherapy in patients with acute myeloid leukaemia aged 60 years or younger. METHODS: This randomised, double-blind, placebo-controlled, phase 2 trial was done at 25 sites in Germany. We enrolled patients aged 18-60 years with newly diagnosed, previously untreated acute myeloid leukaemia who had a WHO clinical performance score 0-2, adequate renal and liver function, no cardiac comorbidities, and no recent trauma or operation. Patients were randomly assigned (1:1) to receive two cycles of induction therapy with daunorubicin (60 mg/m(2) on days 3-5) plus cytarabine (100 mg/m(2) on days 1-7), followed by three cycles of high-dose cytarabine consolidation therapy (3 g/m(2) twice daily on days 1, 3, and 5) plus either sorafenib (400 mg twice daily) or placebo on days 10-19 of induction cycles 1 and 2, from day 8 of each consolidation, and as maintenance for 12 months. Allogeneic stem-cell transplantation was scheduled for all intermediate-risk patients with a sibling donor and for all high-risk patients with a matched donor in first remission. Computer-generated randomisation was done in blocks. The primary endpoint was event-free survival, with an event defined as either primary treatment failure or relapse or death, assessed in all randomised patients who received at least one dose of study treatment. We report the final analysis. This trial is registered with ClinicalTrials.gov, number NCT00893373, and the EU Clinical Trials Register (2008-004968-40). FINDINGS: Between March 27, 2009, and Nov 28, 2011, 276 patients were enrolled and randomised, of whom nine did not receive study medication. 267 patients were included in the primary analysis (placebo, n=133; sorafenib, n=134). With a median follow-up of 36 months (IQR 35·5-38·1), median event-free survival was 9 months (95% CI 4-15) in the placebo group versus 21 months (9-32) in the sorafenib group, corresponding to a 3-year event-free survival of 22% (95% CI 13-32) in the placebo group versus 40% (29-51) in the sorafenib group (hazard ratio [HR] 0·64, 95% CI; 0·45-0·91; p=0·013). The most common grade 3-4 adverse events in both groups were fever (71 [53%] in the placebo group vs 73 [54%] in the sorafenib group), infections (55 [41%] vs 46 [34%]), pneumonia (21 [16%] vs 20 [14%]), and pain (13 [10%] vs 15 [11%]). Grade 3 or worse adverse events that were significantly more common in the sorafenib group than the placebo group were fever (relative risk [RR] 1·54, 95% CI 1·04-2·28), diarrhoea (RR 7·89, 2·94-25·2), bleeding (RR 3·75, 1·5-10·0), cardiac events (RR 3·46, 1·15-11·8), hand-foot-skin reaction (only in sorafenib group), and rash (RR 4·06, 1·25-15·7). INTERPRETATION: In patients with acute myeloid leukaemia aged 60 years or younger, the addition of sorafenib to standard chemotherapy has antileukaemic efficacy but also increased toxicity. Our findings suggest that kinase inhibitors could be a useful addition to curative treatment for acute myeloid leukaemia. Overall survival after long-term follow-up and strategies to reduce toxicity are needed to determine the future role of sorafenib in treatment of this disease. FUNDING: Bayer HealthCare.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Neoadjuvant Therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Adult , Age Factors , Antibiotics, Antineoplastic/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Disease Progression , Disease-Free Survival , Double-Blind Method , Female , Germany , Hematopoietic Stem Cell Transplantation , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoadjuvant Therapy/mortality , Niacinamide/adverse effects , Niacinamide/therapeutic use , Phenylurea Compounds/adverse effects , Proportional Hazards Models , Protein Kinase Inhibitors/adverse effects , Recurrence , Risk Factors , Sorafenib , Time Factors , Transplantation, Homologous , Treatment Outcome
6.
Expert Rev Hematol ; 6(6): 767-84, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24219553

ABSTRACT

With a median age at diagnosis of approximately 65-70 years, acute myeloid leukemia (AML) represents a major therapeutic challenge in the elderly. Only 30-35% of elderly patients with AML are considered eligible for intensive chemotherapy and do actually receive it. However, the long-term benefit associated with intensive chemotherapy remains marginal, and the overall outcome for this population remains poor. The remaining 60-65% of elderly AML patients receives supportive care only. Nevertheless, several studies have indicated that patients who receive any therapy had a better outcome if compared with patients who receive supportive care only. Thus, the development of novel, less toxic, targeted agents is offering new options to older AML patients who are unfit for intensive approaches. In the present review, we will report on the results achieved using intensive chemotherapy and novel agents, and will describe some of the new strategies under development for treating older AML patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Antibodies, Monoclonal/therapeutic use , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Humans , Immunoconjugates/therapeutic use , Immunologic Factors/therapeutic use , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/surgery , Prognosis , Purine Nucleosides/chemistry , Purine Nucleosides/therapeutic use , Risk Factors , Stem Cell Transplantation , Transplantation, Autologous
7.
Hepatology ; 52(2): 703-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20683966

ABSTRACT

UNLABELLED: Human carbonyl reductase 1 (CBR1) converts the antitumor drug and anthracycline daunorubicin (DNR) into the alcohol metabolite daunorubicinol (DNROL) with significantly reduced antitumor activity and cardiotoxicity, and this limits the clinical use of DNR. Inhibition of CBR1 can thus increase the efficacy and decrease the toxicity of DNR. Here we report that (-)-epigallocatechin gallate (EGCG) from green tea is a promising inhibitor of CBR1. EGCG directly interacts with CBR1 and acts as a noncompetitive inhibitor with respect to the cofactor reduced nicotinamide adenine dinucleotide phosphate and the substrate isatin. The inhibition is dependent on the pH, and the gallate moiety of EGCG is required for activity. Molecular modeling has revealed that EGCG occupies the active site of CBR1. Furthermore, EGCG specifically enhanced the antitumor activity of DNR against hepatocellular carcinoma SMMC7721 cells expressing high levels of CBR1 and corresponding xenografts. We also demonstrated that EGCG could overcome the resistance to DNR by Hep3B cells stably expressing CBR1 but not by RNA interference of CBR1-HepG2 cells. The level of the metabolite DNROL was negatively correlated with that of EGCG in the cell extracts. Finally, EGCG decreased the cardiotoxicity of DNR in a human carcinoma xenograft model with both SMMC7721 and Hep3B cells in mice. CONCLUSION: These results strongly suggest that EGCG can inhibit CBR1 activity and enhance the effectiveness and decrease the cardiotoxicity of the anticancer drug DNR. These findings also indicate that a combination of EGCG and DNR might represent a novel approach for hepatocellular carcinoma therapy or chemoprevention.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Anticarcinogenic Agents/pharmacology , Anticarcinogenic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Catechin/analogs & derivatives , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Animals , Antibiotics, Antineoplastic/therapeutic use , Catechin/pharmacology , Catechin/therapeutic use , Daunorubicin/therapeutic use , Humans , Mice , Tumor Cells, Cultured
8.
Int J Mol Med ; 25(3): 465-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20127053

ABSTRACT

Chemotherapy remains the staple of treatment for many types of leukemia. Despite the positive impact on extending overall survival in patients with hematological malignancies, new treatment strategies are needed to reduce the nonspecific toxicity and improve the efficacy of treatment. Celastrol, derived from the 'Thunder God Vine' and Pro-EGCG, a pre-drug version of green tea polyphenol EGCG have shown potent biological activity in vitro and in vivo. Whether these natural products augment the efficacy of conventional chemotherapy in the treatment of leukemia cells has yet to be demonstrated. Here we demonstrate that these natural products could sensitize the effect of chemotherapy in both K-562 and Jurkat T human leukemia cells. Accordingly, this potent biological activity was associated with increased levels of leukemia cell killing, caspase 3 activation, and poly(ADP-ribose) polymerase cleavage. Furthermore, the higher levels of apoptotic indices were associated with decreased levels of Bcr-Abl oncoprotein in K-562 cells. Taken together, our findings present a compelling rationale for the development of combination strategies using natural products in the treatment of hematological malignancies.


Subject(s)
Antineoplastic Agents , Catechin/analogs & derivatives , Cell Line, Tumor/drug effects , Leukemia/drug therapy , Prodrugs , Triterpenes , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Catechin/chemistry , Catechin/pharmacology , Catechin/therapeutic use , Cytarabine/chemistry , Cytarabine/pharmacology , Cytarabine/therapeutic use , Daunorubicin/chemistry , Daunorubicin/pharmacology , Daunorubicin/therapeutic use , Fusion Proteins, bcr-abl/metabolism , Humans , Molecular Structure , Pentacyclic Triterpenes , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/therapeutic use , Triterpenes/chemistry , Triterpenes/pharmacology , Triterpenes/therapeutic use
9.
Chin J Integr Med ; 15(3): 193-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19568712

ABSTRACT

OBJECTIVE: To observe the clinical efficacy of Chinese drugs combined with chemotherapy in the treatment of acute myeloid leukemia (AML) and to investigate the prognostic relevance of the main parameters in AML treated with integrative medicine. METHODS: Forty AML patients hospitalized at the authors hospital were treated with Chinese drugs and chemotherapy. The routine examination, immunophenotype and karyotype analyses were carried out. The clinical efficacy was observed and the prognostic factors were analyzed. RESULTS: (1) Clinical efficacy: Twenty patients had complete remission (CR), with the CR rate being 50.0%. Among these patients, the CR rate was 73.9% (17/23) in de novo AML and 17.6% (3/17) in secondary or refractory AML, respectively. The median disease free survival (DFS) was 6 months (2-32 months) and median overall survival (OS) was 7 months (1-36 months). (2) Analysis of prognostic factors: Aging (> 60 years) and hepatosplenomegaly or extramedullary leukemia did not affect the treatment outcome. Patients with lower white blood cell (WBC) counts (<4.0x10(9)/L) had a significantly higher CR rate (P<0.01). Secondary or refractory AML was associated with a lower CR rate and shorter OS (P<0.01,P<0.05). Expression of CD34 was an adverse factor for obtaining CR (P<0.05) and survival in both DFS and OS (P<0.05,P<0.01). The expression of CD56 was significantly associated with a lower CR rate (P<0.05), but did not affect DFS and OS. Twenty-three (57.5%) out of 40 cases had chromosomal abnormalities. The CR rate was decreased and both DFS and OS shortened stepwise from the cases with favorable cytogenetics to those with intermediate and unfavorable cytogenetics (P<0.01). CONCLUSIONS: The combined treatment of Chinese drugs with chemotherapy has a predominant effect in de novo AML. Secondary or refractory AML, expression of CD34 and CD56, and unfavorable cytogenetics were the main factors of poor prognosis in AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Integrative Medicine , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Adult , Aged , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Child , Chromosome Aberrations , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Etoposide/therapeutic use , Female , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Mitoxantrone/therapeutic use , Prognosis , Treatment Outcome , Young Adult
10.
Article in English | MEDLINE | ID: mdl-19074116

ABSTRACT

Acute promyelocytic leukemia (APL) was initially described as a distinct clinical entity in 1957, one year before the first meeting of the American Society of Hematology. With routine administration of all-trans retinoic acid (ATRA) combined with chemotherapy in the early 1990s and arsenic trioxide (ATO) in the late 1990s, cure can now be expected in the majority of both newly diagnosed and relapsed patients. ATRA with anthracycline-based chemotherapy for induction and consolidation followed by ATRA plus low-dose chemotherapy maintenance is currently the standard of care for newly diagnosed patients. Early institution of ATRA before the diagnosis is confirmed by genetics and aggressive blood product support are important to reduce induction mortality, which remains approximately 10% among patients entered on clinical trials, but is certainly higher when all patients are considered. The relapse rate among high-risk patients is approximately 20%, and new strategies include administration of intensified anthracyclines, intermediate- or high-dose ara-C in either induction or consolidation, or ATO as early consolidation. Central nervous system (CNS) prophylaxis for such patients and those with relapsed disease may be important to prevent the development of extramedullary disease. New therapeutic strategies have focused on minimizing chemotherapy and administering ATRA plus ATO as initial therapy. Recent studies suggest that patients who are molecularly negative after intensive consolidation may not benefit from maintenance therapy. Most exciting is the combination of ATRA and ATO, given with minimal chemotherapy only for leukocytosis, which is a very effective new strategy for patients who are unable to tolerate anthracyclines or older adults and soon may replace conventional therapy for many patients. Patients with relapsed disease do well with ATO followed by autologous hematopoietic stem cell transplantation.


Subject(s)
Leukemia, Promyelocytic, Acute/drug therapy , Antineoplastic Agents/therapeutic use , Arsenic Trioxide , Arsenicals/therapeutic use , Clinical Trials as Topic , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Disease-Free Survival , Growth Inhibitors/therapeutic use , History, 20th Century , Humans , Leukemia, Promyelocytic, Acute/history , Middle Aged , Neoplasm Recurrence, Local/epidemiology , Oxides/therapeutic use , Tretinoin/therapeutic use
11.
J Steroid Biochem Mol Biol ; 110(1-2): 144-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18482833

ABSTRACT

The use of daunomycin against neoplasms is limited due to its severe cardiotoxicity. The cytotoxicity of daunomycin can be minimized by linking it to an affinity tag. Since ovarian cancer cells are sensitive to isoflavone action, we synthesized a daidzein daunomycin conjugate. In MLS human ovarian cancer cells, the conjugate was shown to have a larger cytotoxic effect than daunomycin per se at a low concentration. The conjugate was then tested in vivo in mice carrying MLS xenografts. Tumour growth in the groups of conjugate and daunomycin was inhibited by >50% as compared to vehicle treated mice. In contrast to daunomycin treated mice, no weight reduction or death was seen in mice treated with the conjugate. In vivo imaging of the fluorescence signal generated by daunomycin indicated uptake of both conjugate and daunomycin by the tumour. Tumour fluorescence was, however, higher in the conjugate treated mice than in the daunomycin treated mice, thus suggesting specific delivery of the drug to the tumour. Histological examination of myocardial tissue indicated that only the daunomycin, but not conjugate treated mice showed cardiac damage. These results indicate that targeting of daunomycin via carboxymethyldaidzein retains daunomycin's cytotoxic effects while averting its toxicity in an ovarian xenograft.


Subject(s)
Daunorubicin/pharmacology , Isoflavones/pharmacology , Ovarian Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Daunorubicin/chemistry , Daunorubicin/therapeutic use , Female , Humans , Isoflavones/chemistry , Isoflavones/therapeutic use , Mice , Mice, Nude , Molecular Structure , Ovarian Neoplasms/pathology , Phytoestrogens/chemistry , Phytoestrogens/pharmacology , Phytoestrogens/therapeutic use , Tumor Burden/drug effects
13.
Korean J Lab Med ; 27(2): 89-95, 2007 Apr.
Article in English | MEDLINE | ID: mdl-18094557

ABSTRACT

BACKGROUND: Cellular drug resistance is supposed to play a major role in chemotherapy failure or relapse. The purpose of this study was to analyze the relationship between in vitro chemosensitivity test results using a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and clinical response on chemotherapy, and to find the possibility of optimizing the treatment protocol for individual patients according to their actual drug resistance. METHODS: For MTT assay, we obtained bone marrow aspirates from 103 patients with acute leukemia at the time of initial diagnosis or relapse. The following drugs were tested: cytarabine, vincristine, methotrexate, daunorubicin, dexamethasone, L-asparaginase, and mitoxantrone. To evaluate clinical responses after induction chemotherapy, we followed up on their bone marrow study. RESULTS: In our study, in vitro chemosensitivity test with the MTT assay significantly predicted whether patients with AML remained continuous complete remission or went into relapse. It also predicted whether or not child patients with ALL would acquire complete remission after induction chemotherapy. CONCLUSIONS: Although it does not provide the insight into the mechanisms that cause drug resistance, the MTT assay may be a useful tool in individually optimizing the chemotherapy of patients with acute leukemia.


Subject(s)
Leukemia, Biphenotypic, Acute/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/therapeutic use , Child , Child, Preschool , Coloring Agents , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Female , Humans , Infant , Leukemia, Biphenotypic, Acute/diagnosis , Leukemia, Myeloid, Acute/diagnosis , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Tetrazolium Salts , Thiazoles , Treatment Outcome
14.
Article in English | WPRIM | ID: wpr-165132

ABSTRACT

BACKGROUND: Cellular drug resistance is supposed to play a major role in chemotherapy failure or relapse. The purpose of this study was to analyze the relationship between in vitro chemosensitivity test results using a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and clinical response on chemotherapy, and to find the possibility of optimizing the treatment protocol for individual patients according to their actual drug resistance. METHODS: For MTT assay, we obtained bone marrow aspirates from 103 patients with acute leukemia at the time of initial diagnosis or relapse. The following drugs were tested: cytarabine, vincristine, methotrexate, daunorubicin, dexamethasone, L-asparaginase, and mitoxantrone. To evaluate clinical responses after induction chemotherapy, we followed up on their bone marrow study. RESULTS: In our study, in vitro chemosensitivity test with the MTT assay significantly predicted whether patients with AML remained continuous complete remission or went into relapse. It also predicted whether or not child patients with ALL would acquire complete remission after induction chemotherapy. CONCLUSIONS: Although it does not provide the insight into the mechanisms that cause drug resistance, the MTT assay may be a useful tool in individually optimizing the chemotherapy of patients with acute leukemia.


Subject(s)
Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Antibiotics, Antineoplastic/therapeutic use , Coloring Agents , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Leukemia, Biphenotypic, Acute/diagnosis , Leukemia, Myeloid, Acute/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Tetrazolium Salts , Thiazoles , Treatment Outcome
15.
Leuk Res ; 30(3): 296-302, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16112192

ABSTRACT

Acute myeloid leukaemia (AML) is a heterogenous malignant disease with diverse biological features in which disease progression at the level of CD34+ cells has a major impact on the resistance to chemotherapy and relapse. The AML blast cells in these elderly patients are often characterised by several unfavourable covariates that predict the poor treatment outcome, including high stem cell marker CD34 expression, minimally or undifferentiated features, high P-glycoprotein expression, high bcl-2/bax ratio, unfavourable karyotype and more frequent internal tandem duplications (ITDs) and mutations of class III receptor-type tyrosine kinase for key haematopoietic cytokines: Flt-3 (receptor for Flt-ligand), c-kit (receptor for stem cell factor) and fms (receptor for M-CSF). Testing the new and more specific molecular-targeted therapeutic approaches in CD34+ AML cells can provide the basis for a more effective combined molecular/chemotherapy regimen and may consequently improve the treatment outcome in elderly AML patients. Therefore, the present study was performed to evaluate whether stem cell factor-antibody (anti-SCF) can enhance the efficacy of the two main chemotherapeutic drugs used in AML therapy: cytarabine and daunorubicin at low doses in human-resistant CD34+ AML cells, in an attempt to identify a novel effective regimen with tolerable side-effects for elderly AML patients. The effect of anti-SCF on each of the two chemotherapeutic drugs-induced apoptosis and necrosis was investigated in KG1a human-resistant CD34+ AML cells expressing P-glycoprotein to determine its enhancing activity. Anti-SCF has significantly enhanced the low dose cytarabine- and daunorubicin-induced apoptosis+necrosis in KG1a CD34+ AML cells from 12.0+/-1.7 to 40.9+/-5.9% and from 16.3+/-0.9 to 48.9+/-1.0%, respectively, p<0.01. It has also exerted its significant enhancement activity on the low dose cytarabine- and daunorubicin-induced apoptosis+necrosis in KG1a CD34+ AML cells in the presence of SCF, p<0.05. Anti-SCF has significantly enhanced the low dose cytarabine- and daunorubicin-induced bcl-2 reduction in KG1a CD34+ AML cells from 26.7+/-0.6 to 64.6+/-1.0% and from 59.8+/-3.1 to 80.1+/-7.9%, respectively, p<0.01. The addition of SCF has not altered the low dose cytarabine- and daunorubicin-induced bcl-2 reduction in KG1a CD34+ AML cells (Table 4). Anti-SCF has also significantly enhanced the low dose cytarabine- and daunorubicin-induced bcl-2 reduction in KG1a CD34+ AML cells in the presence of SCF, p<0.05. The unique potent enhancing activity of anti-SCF on low dose chemotherapy-induced apoptosis and necrosis in extremely resistant AML cells suggest a novel promising role for the treatment of elderly AML patients. Further studies are warranted to evaluate a similar enhancing effect for anti-SCF in blast cells from elderly AML patients in primary cultures before its introduction in a pilot clinical study. In conclusion, the combination of anti-SCF and the low dose cytarabine provides a promising solution for the dilemma of therapy in elderly AML patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, CD34 , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cytarabine/pharmacology , Daunorubicin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Stem Cell Factor/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Aged , Aged, 80 and over , Antibodies, Monoclonal/therapeutic use , Antigens, CD34/biosynthesis , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor/biosynthesis , Blast Crisis/drug therapy , Blast Crisis/metabolism , Blast Crisis/pathology , Cell Line, Tumor , Chromosome Aberrations , Clinical Trials as Topic , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Pilot Projects , Receptors, Cytokine/metabolism , Stem Cell Factor/metabolism , Treatment Outcome , bcl-2-Associated X Protein/biosynthesis
16.
J Fr Ophtalmol ; 27(9 Pt 1): 1051-9, 2004 Nov.
Article in French | MEDLINE | ID: mdl-15557870

ABSTRACT

Proliferative vitreoretinopathy (PVR) is the major cause of retinal detachment surgery failure. Many adjuvants were studied in vitro and on animals, some were studied on humans. Daunomycin seems to reduce PVR recidivism. 5-FU and steroids are nontoxic but their efficacy is not clear. Heparin reduced postoperative inflammation and seems to reduce PVR recidivism when associated with 5-FU. Associating heparin and steroids seems to reduce PVR in some groups of patients (aphakic, anterior PVR). Colchicine and retinoic acid per os are ineffective, silicone oil is effective as an internal tamponade but not as an adjuvant of PVR. Currently, no molecule has proven efficacy as an adjunctive treatment of PVR.


Subject(s)
Vitreoretinopathy, Proliferative/drug therapy , Vitreoretinopathy, Proliferative/surgery , Chemotherapy, Adjuvant , Clinical Trials as Topic , Daunorubicin/therapeutic use , Glucocorticoids/therapeutic use , Heparin/therapeutic use , Humans , Vitreoretinopathy, Proliferative/diagnosis
17.
J Antimicrob Chemother ; 45(1): 69-75, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10629015

ABSTRACT

Trovafloxacin, a new trifluoroquinolone, was evaluated for its therapeutic efficacy against Klebsiella pneumoniae lung infection in tumour (P388 murine leukaemia cells)-bearing mice, treated with or without a chemotherapeutic agent, daunorubicin (DNR) and in mice without tumour. Its activity was compared with ciprofloxacin and cephazolin. The effect on therapeutic efficacy of the addition of recombinant granulocyte colony stimulating factor (rGCSF) was also examined. Our study showed that both quinolones successfully cured pneumonia owing to infection with K. pneumoniae in mice without tumours but that all antibiotics failed in tumour-bearing mice if DNR was withheld. Substantial differences were noted in DNR-treated tumour-bearing mice with infection-the cure rate with trovafloxacin was 91% whereas the cure rate with ciprofloxacin or cephazolin was 57%. Addition of rGCSF to ciprofloxacin did not substantially improve its efficacy (when assessed by protection against death owing to infection; the survival rate was 41%). Trovafloxacin cure rates ranged from 80 to 90% whether or not rGCSF was added to the treatment regimen. Our results suggest that prior cancer chemotherapy had no adverse effect on the therapeutic efficacy of trovafloxacin, and that trovafloxacin may be a promising therapeutic agent for treatment of bacterial infections in the presence of leucopenia.


Subject(s)
Anti-Infective Agents/therapeutic use , Fluoroquinolones , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/drug effects , Leukemia P388/complications , Naphthyridines/therapeutic use , Pneumonia, Bacterial/drug therapy , Animals , Anti-Infective Agents/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Ciprofloxacin/therapeutic use , Daunorubicin/therapeutic use , Drug Evaluation, Preclinical , Drug Therapy, Combination , Granulocyte Colony-Stimulating Factor/therapeutic use , Klebsiella Infections/complications , Klebsiella Infections/metabolism , Klebsiella pneumoniae/metabolism , Leukemia P388/drug therapy , Leukemia P388/metabolism , Leukocyte Count/drug effects , Male , Mice , Mice, Inbred DBA , Naphthyridines/pharmacokinetics , Neoplasm Transplantation , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/metabolism , Recombinant Proteins
18.
Am J Ophthalmol ; 126(4): 550-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9780100

ABSTRACT

PURPOSE: To assess the efficacy and safety of adjunctive daunorubicin during vitrectomy surgery in eyes with idiopathic proliferative vitreoretinopathy (PVR). METHODS: Two hundred eighty-six eyes (286 patients) with stage C2 (Retina Society Classification, 1983) or more advanced preoperative PVR in which surgery with silicone oil was planned were enrolled in a multicenter, prospective, randomized, controlled clinical trial. Standardized surgery plus adjunctive daunorubicin perfusion was compared with surgery alone. Outcomes assessed were retinal attachment without additional vitreoretinal surgery 6 months after standardized surgery, number of and time until vitreoretinal reoperations within 1 year of standardized surgery, and change in visual acuity 1 year after standardized surgery, evaluated by photodocumentation, number of reoperations, and measurement of best-corrected visual function. Outcomes were determined 6 months after operation and reevaluated after 1 year of follow-up. RESULTS: Six months after standardized surgery, complete retinal reattachment without additional vitreoretinal surgery was achieved in 62.7% (89/142) of eyes in the daunorubicin group vs 54.1% (73/135) in the control group (P = .07, one-sided). However, in the daunorubicin group, significantly fewer vitreoretinal reoperations were performed within 1 year postoperatively (P = .005, one-sided) to achieve the same overall 1-year retinal reattachment rate (80.2% [105/131] vs 81.8% [103/126]). The rate of patients with no vitreoretinal reoperations was 65.5% (95/145) in the daunorubicin group vs 53.9% (76/141) in the control group. There was no difference in the best-corrected visual acuity. No severe adverse effect related to daunorubicin was identified. CONCLUSIONS: Although the rate of anatomic success after 6 months failed to show significance, some benefit of the adjunctive treatment exists, especially a tendency toward increased rate of reattachment and a significant reduction in the number of reoperations. This shows that human PVR is amenable to pharmacologic treatment.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Daunorubicin/therapeutic use , Vitrectomy , Vitreoretinopathy, Proliferative/drug therapy , Adult , Aged , Chemotherapy, Adjuvant , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies , Safety , Scleral Buckling , Treatment Outcome , Visual Acuity , Vitreoretinopathy, Proliferative/pathology , Vitreoretinopathy, Proliferative/surgery
19.
Anticancer Res ; 17(3C): 2083-8, 1997.
Article in English | MEDLINE | ID: mdl-9216668

ABSTRACT

Tumor cell resistance to many unrelated anticancer drugs is a major obstacle during cancer chemotherapy. One mechanism of drug resistance is thought to be due to the efflux of anticancer drugs caused by P-glycoprotein. In recent years, magnetic fields have been found to enhance the potency of anticancer drugs, with favorable modulation of cancer therapy. In this study, KB-ChR-8-5-11, a multidrug resistant (MDR) human carcinoma subline, was used as a model to evaluate the ability of pulsed magnetic fields (PMF) to modulate the potency of daunorubicin (DNR) in vivo and to determine the appropriate order of exposure to drugs and PMF using an in vitro cytotoxicity assay. Solenoid coils with a ramped pulse current source were used at 250 pulses per second for both in vivo and in vitro experiments. For the in vivo study, KB-ChR-8-5-11 cells were inoculated into thymic Balbc-nu/nu female mice. Treatment was begun when the average tumor volume reached 250-450 mm3. Treatment consisted of whole body exposure to PMF for one hour, followed immediately by intravenous (i.v.) injection of 8 mg/kg DNR designated as day 0, and repeated on days 7 and 14. Among the various groups, significant differences in the tumor volume were found between PMF + saline and PMF + DNR groups (p = 0.0107) at 39 days and 42 days (p = 0.0101). No mice died in the PMF alone group, and no toxicity attributable to PMF was found during the experimental period. For the in vitro studies, the sulforhodamine blue (SRB) cytotoxicity assay was used to determine the effect of the sequence which cells are exposed to PMF and/or DNR. Cells were exposed to PMF either before (pre-PMF) or after (post-PMF) drug was added. Results showed that the IC50 was significantly different between controls and pre-PMF + DNR groups (P = 0.0096, P = 0.0088). The IC50 of the post-PMF + DNR group was not found to be significantly different from control groups. Thus, the data in this report demonstrates that PMF enhanced the potency of DNR against KB-ChR-8-5-11 xenograft in vivo, while the efficacy of DNR was potentiated in vitro by PMF exposure only when PMF exposure occurred in the presence of drug. The data in vitro suggest that the mechanism by which PMFs modulate DNR's potency may be by inhibition of the efflux pump, P-glycoprotein. Further work to determine conditions for maximum modulation of drug potency by PMFs is warranted.


Subject(s)
Daunorubicin/toxicity , Drug Resistance, Multiple/radiation effects , Magnetics , Animals , Clone Cells , Combined Modality Therapy , Daunorubicin/therapeutic use , Female , Humans , KB Cells , Magnetics/therapeutic use , Mice , Mice, Nude , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/radiotherapy , Transplantation, Heterologous
20.
Planta Med ; 61(5): 409-13, 1995 Oct.
Article in English | MEDLINE | ID: mdl-7480200

ABSTRACT

Examined in vitro were the effects of some triterpenoids from Panax (Araliaceae) and Glycyrrhiza (Leguminosae) spp. on the sensitivity to daunomycin (DAU) and vinblastine (VBL) of adriamycin (ADM)-resistant P388 leukemia cells (P388/ADM), which were resistant to multiple anticancer drugs. Quasipanaxatriol, 20(S)-protopanaxatriol, ginsenoside Rh2, and compound K greatly enhanced the cytotoxicity of the anti-cancer drugs in P388/ADM cells. The extent of enhancement was different among the triterpene compounds; the 4- to 46-fold increase in DAU cytotoxicity was observed in P388/ADM cells in the presence of non-toxic or marginally toxic concentrations of individual compounds, while those for VBL were in the ratios of 2- to 37-fold. The maximum increase in cytotoxicity was observed with 50 microM quasipanaxatriol; the resistance indices defined to be the ratios of the IC50 values for P388/ADM and P388 parental cells decreased from 79 to 1.7 and from 180 to 4.9 in the cases of DAU and VBL, respectively. The reversal of DAU resistance in P388/ADM by quasipanaxatriol could be explained by the effective accumulation of the drugs mediated by the DAU-efflux blockage.


Subject(s)
Daunorubicin/pharmacology , Leukemia P388/pathology , Triterpenes/pharmacology , Vinblastine/pharmacology , Animals , Biological Transport , Daunorubicin/metabolism , Daunorubicin/therapeutic use , Drug Resistance, Neoplasm , Drug Synergism , Fabaceae/chemistry , Female , Leukemia P388/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Panax/chemistry , Plants, Medicinal , Tumor Cells, Cultured , Vinblastine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL