Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
2.
Clin Cancer Res ; 4(7): 1641-7, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9676838

RESUMO

TP3-pokeweed antiviral protein (PAP) immunotoxin is directed against the p80 antigen on osteosarcoma cells. Previous studies have demonstrated that TP3-PAP kills clonogenic human osteosarcoma cells in vitro and shows significant antitumor activity in a murine soft tissue sarcoma model (P. M. Anderson, et al, Cancer Res., 55: 1321-1327, 1995.) In this study, we demonstrate that TP3-PAP elicits potent in vivo antitumor activity in a hamster cheek pouch model of human osteosarcoma. Furthermore, treatment with TP3-PAP at nontoxic dose levels significantly delayed the emergence and progression of leg tumors and markedly improved tumor-free survival in severe combined immunodeficient mice challenged with OHS human osteosarcoma cells. Thus, TP3-PAP may be useful in the treatment of poor risk osteosarcoma.


Assuntos
Imunotoxinas/uso terapêutico , N-Glicosil Hidrolases , Osteossarcoma/terapia , Fragmentos de Peptídeos/uso terapêutico , Proteínas de Plantas/uso terapêutico , Timopoietinas/uso terapêutico , Animais , Bochecha , Cricetinae , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Mesocricetus , Camundongos , Camundongos SCID , Mucosa Bucal , Proteínas Inativadoras de Ribossomos Tipo 1 , Transplante Heterólogo , Células Tumorais Cultivadas
3.
Clin Cancer Res ; 5(10): 2954-62, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10537365

RESUMO

4-(4'Hydroxyphenyl)-amino-6,7-dimethoxyquinazoline (WHI-P131) is a potent and selective inhibitor of the Janus kinase 3, which triggers apoptosis in human acute lymphoblastic leukemia (ALL) cells. In this preclinical study, we evaluated the pharmacokinetics and toxicity of WHI-P131 in rats, mice, and cynomolgus monkeys. Following i.v. administration, the terminal elimination half-life of WHI-P131 was 73.2 min in rats, 103.4 min in mice, and 45.0 min in monkeys. The i.v. administered WHI-P131 showed a very wide tissue distribution in mice. Following i.p. administration, WHI-P131 was rapidly absorbed in both rats and mice, and the time to reach the maximum plasma concentration (tmax) was 24.8 min in rats and 10.0 min in mice. Subsequently, WHI-P131 was eliminated with a terminal elimination half-life of 51.8 min in rats and 123.6 min in mice. The estimated i.p. bioavailability was 95% for rats, as well as for mice. WHI-P131 was quickly absorbed after oral administration in mice with a tmax of 5.8 min, but its oral bioavailability was relatively low (29.6%). The elimination half-life of WHI-P131 after oral administration was 297.6 min. WHI-P131 was not acutely toxic to mice at single i.p. bolus doses ranging from 0.5-250 mg/kg. Two cynomolgus monkeys treated with 20 mg/kg WHI-P131 and one cynomolgus monkey treated with 100 mg/kg WHI-P131 experienced no side effects. Plasma samples from WHI-P131-treated monkeys exhibited potent antileukemic activity against human ALL cells in vitro. To our knowledge, this is the first preclinical toxicity and pharmacokinetic study of a Janus kinase 3 inhibitor. Further development of WHI-P131 may provide the basis for new and effective treatment programs for relapsed ALL in clinical settings.


Assuntos
Antineoplásicos/farmacocinética , Inibidores Enzimáticos/farmacocinética , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinazolinas/farmacocinética , Animais , Humanos , Janus Quinase 3 , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Quinazolinas/toxicidade , Ratos , Ratos Endogâmicos Lew , Distribuição Tecidual , Células Tumorais Cultivadas
4.
Clin Cancer Res ; 2(9): 1533-42, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9816330

RESUMO

Combined immunochemotherapy regimens using the investigational biotherapeutic agent B43(anti-CD19)-poke-weed antiviral protein (PAP) immunotoxin may offer an effective treatment for refractory B-cell precursor leukemias. The purpose of the present study was to explore and identify effective combinations of B43-PAP with standard chemotherapeutic drugs, including the anthracyclin doxorubicin, the epipodophyllotoxin etoposide, the nitrosurea carmustine, and the antimetabolite cytosine arabinoside. Here, we report that the B43-PAP plus cytosine arabinoside combination has potent antileukemic activity against human B-cell precursor leukemia in SCID mice and leads to 100% long-term event-free survival from an otherwise invariably fatal leukemia. Surprisingly, none of the other treatment protocols tested, including combinations of B43-PAP with carmustine, doxorubicin, or etoposide, proved more effective than B43-PAP alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Citarabina/uso terapêutico , N-Glicosil Hidrolases , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Carmustina/uso terapêutico , Doxorrubicina/uso terapêutico , Etoposídeo/uso terapêutico , Feminino , Humanos , Imunotoxinas/uso terapêutico , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Proteínas de Plantas/uso terapêutico , Proteínas Inativadoras de Ribossomos Tipo 1 , Organismos Livres de Patógenos Específicos , Resultado do Tratamento
5.
Clin Cancer Res ; 4(5): 1125-34, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9607569

RESUMO

Epidermal growth factor receptor (EGFR)-associated protein tyrosine kinase (PTK) complexes have vital anti-apoptotic functions in human breast cancer cells. We have shown previously that targeting the naturally occurring PTK inhibitor genistein to the EGFR-associated PTK complexes using the EGF-Genistein (Gen) conjugate triggers rapid apoptotic cell death in human breast cancer cells and abrogates their in vitro clonogenic growth. In the present study, we examined the in vivo toxicity profile, pharmacokinetics, and anticancer activity of EGF-Gen. No toxicities were observed in mice treated with EGF-Gen at dose levels as high as 40 mg/kg administered i.p. as a single dose or 140 mg/kg administered i.p. over 28 consecutive days. EGF-Gen significantly improved tumor-free survival in a severe combined immune deficiency (SCID) mouse xenograft model of human breast cancer, when it was administered 24 h after inoculation of tumor cells. At 100 microg/kg/day x 10 days (1 mg/kg total dose), which is >100-fold less than the highest tested and nontoxic cumulative dose (ie., 140 mg/kg) in mice, EGF-Gen was more effective than cyclophosphamide (50 mg/kg/day x 2 days), Adriamycin (2.5 mg/kg x 1 day), or methotrexate (0.5 mg/kg x 1 day), the most widely used standard chemotherapeutic drugs for breast cancer, and resulted in 60% long-term tumor-free survival. Furthermore, treating SCID mice with established s.c. human breast cancer xenografts of 0.5-cm diameter with EGF-Gen at this dose level resulted in disappearance of the tumors in two of five mice and >50% shrinkage in three of five mice within 10 days, whereas all of the control tumors in five PBS-treated mice as well as five mice treated with unconjugated Gen (1 mg/kg/day x 10 days) showed >200% increase in diameter during the same observation period. EGF-Gen treatment reduced the growth rate of breast cancer xenografts of 1.0-cm diameter, but unlike with tumors of 0.5-cm diameter, it failed to cause shrinkage or disappearance of these larger tumors. The level of EGF-Gen systemic exposure that was effective in SCID mice was achieved in cynomolgus monkeys without any significant side effects detectable by clinical observation, laboratory studies, or histopathological examination of multiple organs. EGF-Gen might be useful in the treatment of breast cancer as well as other EGFR-positive malignancies.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/efeitos dos fármacos , Genisteína/farmacologia , Fígado/efeitos dos fármacos , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Intervalo Livre de Doença , Combinação de Medicamentos , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/uso terapêutico , Fator de Crescimento Epidérmico/farmacocinética , Fator de Crescimento Epidérmico/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Feminino , Genisteína/farmacocinética , Genisteína/uso terapêutico , Humanos , Fígado/patologia , Macaca fascicularis , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Ensaio de Cápsula Sub-Renal , Análise de Sobrevida , Células Tumorais Cultivadas
6.
Clin Cancer Res ; 4(1): 165-70, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9516966

RESUMO

B43 (anti-CD19)-genistein immunoconjugate targets genistein, a naturally occurring protein tyrosine kinase-inhibitory isoflavone to the membrane-associated antiapoptotic CD19-LYN complexes and triggers apoptotic cell death. In this preclinical study, the toxicity profiles of B43-genistein as well as unconjugated genistein were evaluated in cynomolgus monkeys. B43-genistein and genistein were administered either as single bolus injections or daily injections for 5-10 consecutive days via the i.v. route to monkeys. Neither genistein nor B43-genistein was toxic to cynomolgus monkeys, and no test article-related histopathological lesions were found in any of the two genistein-treated or five B43-genistein-treated cynomolgus monkeys. B43-genistein showed a favorable pharmacokinetics in monkeys, with a plasma half-life of 10-23 h. Plasma samples from B43-genistein-treated monkeys elicited potent and CD19 antigenspecific antileukemic activity against human CD19+ leukemia cells in vitro. To our knowledge, this is the first preclinical toxicity and pharmacokinetic study of a tyrosine kinase inhibitor-containing immunoconjugate in nonhuman primates.


Assuntos
Antígenos CD19/imunologia , Antineoplásicos/toxicidade , Inibidores Enzimáticos/toxicidade , Genisteína/toxicidade , Imunoconjugados/toxicidade , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Feminino , Imunoconjugados/farmacocinética , Macaca fascicularis
7.
Clin Cancer Res ; 3(12 Pt 1): 2217-27, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9815618

RESUMO

Acute myeloid leukemia (AML) is the most common form of acute leukemia. Contemporary chemotherapy regimens fail to cure most patients with AML. We have genetically engineered a recombinant diphtheria toxin human granulocyte macrophage colony-stimulating factor (GMCSF) chimeric fusion protein (DTctGMCSF) that specifically targets the GMCSF receptor on fresh human AML cells and myeloid leukemia cell lines. At a nontoxic dose level, DTctGMCSF therapy was superior to the standard chemotherapeutic agents 1-beta-D-arabinofuranosylcytosine and Adriamycin, resulting in 60% long-term event-free survival of severe combined immunodeficient mice challenged with an otherwise invariably fatal cell dose of the human HL-60 myeloid leukemia. Notably, systemic exposure levels of DTctGMCSF, which were found to be therapeutic in the severe combined immunodeficient mouse xenograft model of human HL-60 myeloid leukemia, could be achieved in cynomolgus monkeys without any significant nonhematological toxicities. The recombinant DTctGMCSF fusion toxin might be useful in the treatment of AML patients whose leukemias have recurred and developed resistance to contemporary chemotherapy programs.


Assuntos
Toxina Diftérica/farmacocinética , Toxina Diftérica/uso terapêutico , Imunotoxinas/farmacocinética , Imunotoxinas/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Animais , Citarabina/uso terapêutico , Toxina Diftérica/toxicidade , Intervalo Livre de Doença , Doxorrubicina/uso terapêutico , Feminino , Células HL-60 , Humanos , Imunotoxinas/toxicidade , Macaca fascicularis , Camundongos , Camundongos SCID , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes de Fusão/toxicidade , Distribuição Tecidual , Transplante Heterólogo
8.
Clin Cancer Res ; 3(3): 325-37, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9815689

RESUMO

We studied the pharmacokinetic features, immunogenicity, and toxicity of B43-pokeweed antiviral protein (PAP) immunotoxin in 13 cynomolgus monkeys. The disposition of B43-PAP in two monkeys, when administered as a single i.v. bolus dose, was characterized by a slow clearance (1-2 ml/h/kg) with a very discrete peripheral distribution. B43-PAP was retained and distributed largely in the blood as the sole compartment with no significant equilibration with the extravascular compartment. The circulating B43-PAP immunotoxin detected in monkey plasma samples by ELISA and protein immunoblotting was both immunoreactive with, and active against, human leukemic cells in vitro. In systemic immunogenicity and toxicity studies, which involved 11 cynomolgus monkeys, each monkey received a total of seven i.v. doses of B43-PAP at a specific dose level of the dose escalation schedule. B43-PAP-treated monkeys mounted a dose-dependent humoral immune response against both the mouse IgG and PAP moieties of the immunotoxin. When administered i.v. either on an every-day or every-other-day schedule, B43-PAP was very well tolerated, with no significant clinical or laboratory signs of toxicity at total dose levels ranging from 0.007 to 0.7 mg/kg. A transient episode of a mild capillary leak with a grade 2 hypoalbuminemia and 2+ proteinuria was observed at total dose levels equal to or higher than 0.35 mg/kg. At total dose levels of 3.5 and 7.0 mg/kg, B43-PAP caused dose-limiting renal toxicity due to severe renal tubular necrosis. The present study completes the preclinical evaluation of B43-PAP and provides the basis for its clinical evaluation in children with therapy-refractory B-lineage acute lymphoblastic leukemia.


Assuntos
Antígenos CD19/imunologia , Antivirais/farmacocinética , Imunotoxinas/farmacocinética , N-Glicosil Hidrolases , Proteínas de Plantas/farmacocinética , Animais , Antivirais/toxicidade , Humanos , Imunotoxinas/sangue , Imunotoxinas/toxicidade , Injeções Intravenosas , Rim/efeitos dos fármacos , Rim/patologia , Cinética , Macaca fascicularis , Camundongos , Modelos Biológicos , Proteínas de Plantas/sangue , Proteínas de Plantas/toxicidade , Proteinúria , Proteínas Inativadoras de Ribossomos Tipo 1
9.
Clin Cancer Res ; 3(6): 881-90, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9815763

RESUMO

We evaluated the TXU (anti-CD7)-pokeweed antiviral protein (PAP) immunotoxin in both murine and nonhuman primate models. TXU-PAP caused dose-limiting cardiac toxicity in BALB/c mice. In a SCID mouse model of invariably fatal human T-lineage acute lymphoblastic leukemia (ALL), TXU-PAP therapy resulted in a marked improvement of leukemia-free survival without any side effects. Whereas 100% of control mice treated with PBS, unconjugated TXU antibody, or B43-PAP (an immunotoxin that does not react with T-lineage ALL cells) died of disseminated human leukemia within 80 days (median survival, 37 days), 80 +/- 13% of SCID mice treated with 15 microgram of TXU-PAP (median survival, >120 days) and 100% of mice treated with 30 microgram of TXU-PAP (median survival, > 120 days) remained alive and free of leukemia for >120 days. In cynomolgus monkeys, TXU-PAP showed favorable pharmacokinetics with an elimination half-life of 8.1-8.7 h. The monkeys treated with TXU-PAP at dose levels of 0.05 mg/kg/day x 5 days and 0.10 mg/kg/day x 5 days tolerated the therapy very well, without any significant clinical compromise or side effects, and at necropsy, no gross or microscopic lesions were found. This study provides a basis for further evaluation of TXU-PAP as an investigational biotherapeutic agent in the treatment of T-lineage ALL.


Assuntos
Imunoconjugados/farmacocinética , Imunoconjugados/toxicidade , Imunotoxinas/farmacocinética , Imunotoxinas/toxicidade , Leucemia-Linfoma de Células T do Adulto/tratamento farmacológico , Proteínas de Plantas/farmacocinética , Proteínas de Plantas/toxicidade , Animais , Formação de Anticorpos , Antígenos CD7/imunologia , Coração/efeitos dos fármacos , Humanos , Imunoconjugados/uso terapêutico , Imunoglobulina G/biossíntese , Imunotoxinas/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/patologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Miocárdio/patologia , Proteínas de Plantas/uso terapêutico , Proteínas Inativadoras de Ribossomos Tipo 1 , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas
10.
Leuk Lymphoma ; 33(3-4): 289-93, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10221508

RESUMO

We used a SCID mouse model of human B-lineage acute lymphoblastic leukemia to examine the antileukemic activity of temozolomide in comparison to as well as in combination with B43-PAP anti-CD19 immunotoxin. One hundred percent of the 20 PBS-treated control mice died of disseminated human B-lineage ALL at 32 to 64 days after the inoculation of 1x10(6) NALM-6 cells, with a median event free survival time of 43 +/- 1 days. Temozolomide, when administered i.p. for 5 consecutive days at a dose level of 411 mg/m2 or as a single 750 mg/m2 bolus dose, elicited significant antileukemic activity and improved survival in this SCID mouse model of human B-lineage ALL. The median survival times were 43 +/- 1 days for PBS-treated mice, 56 +/- 16 days for mice injected with the 5-day temozolomide program, and 64 +/- 15 days for mice treated with a single bolus dose of temozolomide. However, temozolomide was not as effective as B43-PAP. Whereas only 40 +/- 21% of mice treated with temozolomide survived beyond 120 days, B43-PAP treatment resulted in 74 +/- 7% survival in the same model system. The combination of temozolomide with B43-PAP was well tolerated by mice but it was not significantly more effective than B43-PAP alone. Temozolomide may have very limited potential as an antileukemic agent for treatment of B-lineage ALL.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Linfoma de Burkitt/tratamento farmacológico , Dacarbazina/análogos & derivados , Imunotoxinas/uso terapêutico , N-Glicosil Hidrolases , Proteínas de Plantas/uso terapêutico , Animais , Terapia Combinada , Dacarbazina/uso terapêutico , Humanos , Camundongos , Camundongos SCID , Proteínas Inativadoras de Ribossomos Tipo 1 , Temozolomida , Fatores de Tempo , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA