Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Oncogene ; 33(45): 5295-302, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-24909177

ABSTRACT

Pleuropulmonary blastoma is a rare childhood malignancy of lung mesenchymal cells that can remain dormant as epithelial cysts or progress to high-grade sarcoma. Predisposing germline loss-of-function DICER1 variants have been described. We sought to uncover additional contributors through whole exome sequencing of 15 tumor/normal pairs, followed by targeted resequencing, miRNA analysis and immunohistochemical analysis of additional tumors. In addition to frequent biallelic loss  of TP53 and mutations of NRAS or BRAF in some cases, each case had compound disruption of DICER1: a germline (12 cases) or somatic (3 cases) loss-of-function variant plus a somatic missense mutation in the RNase IIIb domain. 5p-Derived microRNA (miRNA) transcripts retained abnormal precursor miRNA loop sequences normally removed by DICER1. This work both defines a genetic interaction landscape with DICER1 mutation and provides evidence for alteration in miRNA transcripts as a consequence of DICER1 disruption in cancer.


Subject(s)
DEAD-box RNA Helicases/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Mutation , Pulmonary Blastoma/genetics , Ribonuclease III/genetics , Tumor Suppressor Protein p53/genetics , Base Sequence , Chromosomes, Human, Pair 5/genetics , DEAD-box RNA Helicases/metabolism , DNA Copy Number Variations , Exome/genetics , Female , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/chemistry , Nucleic Acid Conformation , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pulmonary Blastoma/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III/metabolism , Sequence Analysis, DNA/methods , Tumor Suppressor Protein p53/metabolism
2.
J Clin Pharmacol ; 39(12): 1248-55, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10586390

ABSTRACT

The authors examined the pharmacokinetics of the CD19 receptor-directed tyrosine kinase inhibitor B43-Genistein in 17 patients (4 children, 13 adults) with B-lineage lymphoid malignancies, including 12 patients with acute lymphoblastic leukemia (ALL) and 5 patients with non-Hodgkin's lymphoma (NHL). The immunoconjugate was administered intravenously as a 1-hour continuous infusion at a dose level of either 0.1 mg/kg (N = 12) or 0.18 mg/kg (N = 5), and the plasma concentration-time data were modeled by using the WinNonlin program to estimate the pharmacokinetic parameters. Pharmacokinetic analyses revealed a plasma half-life of 19 +/- 4 hours, mean residence time of 22 +/- 4 hours, and a systemic clearance of 18 +/- 2 mL/h/kg. The average (mean +/- SEM) values for the maximum plasma concentration Cmax, volume of distribution at steady state (Vss), and area under curve (AUC) were 1092 +/- 225 ng/ml, 291 +/- 37 mL/kg, and 9987 +/- 2021 micrograms x h/L, respectively. The AUC values were higher at the 0.18 mg/kg dose level than at the 0.1 mg/kg dose level (16,848 +/- 5118 micrograms x h/L vs. 7128 +/- 1156 micrograms x h/L, p = 0.009). Patients with ALL had a significantly larger volume of distribution at steady state (332 +/- 47 mL/kg vs. 191 +/- 12 mL/kg, p = 0.04), faster clearance (21 +/- 3 mL/h/kg vs. 11 +/- 2 mL/h/kg, p = 0.03), and lower dose-corrected AUC than patients with NHL (6010 +/- 836 micrograms x h/L vs. 12,044 +/- 2707 micrograms x h/L, p = 0.006). There was a trend toward faster clearance rates (23 +/- 4 mL/h/kg vs. 16 +/- 3 mL/h/kg, p = 0.1), shorter elimination half-lives (5.7 +/- 3.6 hours vs. 13 +/- 8.8 hours, p = 0.1), and shorter mean residence times (11 +/- 3 hours vs. 25 +/- 5 hours, p = 0.08) for non-Caucasian patients as compared to Caucasian patients. When compared to adult patients, pediatric patients showed a significantly larger volume of distribution at steady state (418 +/- 82 mL/kg vs. 252 +/- 34 mL/kg, p = 0.02) and a longer elimination half-lives (18.4 +/- 13.6 hours vs. 8.7 +/- 6.7 hours, p = 0.04). The pharmacokinetics of B43-Genistein was not affected by the gender of the patients or by bone marrow transplantation in past medical history. Overall, B43-Genistein showed favorable pharmacokinetics in this heavily pretreated leukemia/lymphoma patient population, which is reminiscent of its recently reported favorable pharmacokinetics in cynomolgus monkeys. To our knowledge, this is the first clinical pharmacokinetics study of a tyrosine kinase inhibitor containing immunoconjugate.


Subject(s)
Antigens, CD19/metabolism , Antineoplastic Agents/pharmacokinetics , Genistein/pharmacokinetics , Lymphoma, Non-Hodgkin/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Adolescent , Adult , Age Factors , Child , Female , Humans , Immunoconjugates/pharmacokinetics , Lymphoma, Non-Hodgkin/ethnology , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/ethnology , Sex Characteristics
3.
J Pharmacol Exp Ther ; 291(3): 1301-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10565855

ABSTRACT

The purpose of the present study was to evaluate the toxicity and pharmacokinetics of TXU (anti-CD7)-pokeweed antiviral protein (PAP) in human immunodeficiency virus (HIV)-infected chimpanzees and adult patients. At a total dose of 100 microg/kg, TXU-PAP did not cause severe (grade >/= 3) toxicity in any of the four HIV type 1 (HIV-1)-infected or two healthy chimpanzees. The only side effects were a transient elevation of the liver enzyme alanine aminotransferase between days 2 and 14 without a concomitant rise in total bilirubin levels and a decrease in the serum albumin levels between days 1 and 5 without any concomitant weight gain or peripheral edema. TXU-PAP showed favorable pharmacokinetics in chimpanzees with a plasma elimination half-life of 5.1 to 12.0 h and a systemic clearance of 5.8 to 15.1 ml/h/kg. At 2 months after initiation of the TXU-PAP infusions, the HIV-1 burden was reduced to below-detection levels in three of the four chimpanzees, and in the remaining chimpanzee, the HIV burden was <500 RNA copies/ml at 2 weeks but returned to the pretreatment levels by 2 months. TXU-PAP was well tolerated by HIV-1-infected adult patients who received a single 5 microg/kg i.v. infusion of TXU-PAP. TXU-PAP showed very favorable pharmacokinetics in these patients with a relatively long plasma elimination half-life of 12.4 +/- 1.4 h, a mean residence time of 17.9 +/- 2.0 h, and a slow systemic clearance of 2.7 +/- 0.7 ml/h/kg. Concentrations of TXU-PAP required for effective inhibition of HIV-1 replication in preclinical models were achieved in HIV-1-infected patients at the 5 microg/kg dose level without any adverse reactions, and the mean value for AUC was 3059 +/- 721 ng. h/ml. The 1-h postinfusion plasma samples from TXU-PAP-treated patients showed potent anti-HIV activity in vitro and inhibited the replication of HIV in normal peripheral blood mononuclear cells (PBMCs) even at a 1:100 dilution. Although treatment with TXU-PAP at the 5 microg/kg dose level does not provide sustained therapeutic levels, it was capable of reducing the viral burden in six of six patients evaluated. To our knowledge, this is the first report of a clinical pharmacokinetics study of a PAP immunoconjugate in HIV-infected patients. The favorable long plasma elimination half-life of TXU-PAP in combination with its low toxicity provides the basis for further investigation of TXU-PAP as a potential anti-HIV agent.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/drug effects , Immunoconjugates/therapeutic use , Plant Proteins/therapeutic use , Adult , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacokinetics , Area Under Curve , CD4-CD8 Ratio/drug effects , CD56 Antigen/immunology , Female , Follow-Up Studies , HIV Infections/blood , HIV Infections/virology , Half-Life , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Male , Middle Aged , Pan troglodytes , Plant Proteins/administration & dosage , Plant Proteins/pharmacokinetics , Ribosome Inactivating Proteins, Type 1 , Viral Load , Virus Replication/drug effects
4.
Leuk Lymphoma ; 34(5-6): 415-32, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10492065

ABSTRACT

Although anthracyclines are associated with significant cardiac toxicity and their benefit remains unclear, they are included in nearly all current protocols for the treatment of childhood acute lymphoblastic leukemia (ALL). Currently open trials from most major groups use anthracyclines in the induction phase for all high-risk patients and in the delayed intensification phase for all patients regardless of risk classification. Our review of published randomized studies reveals no benefit for the addition of anthracyclines to induction phase of childhood ALL regimens consisting of vincristine, prednisone, and L-asparaginase (VPL), with or without a delayed intensification phase. No randomized studies have evaluated the use of anthracyclines in the delayed intensification phase of therapy. Furthermore, studies of relapsed patients indicated no benefit for the addition anthracyclines to maintenance regimens. Recent evidence from preclinical studies suggests that a combination of VPL with an anti-CD19 immunotoxin is more effective than VPL plus anthracyclines combination. Accumulated evidence exists that anthracyclines are associated with late-onset cardiac morbidity in about 25% of childhood ALL and other cancer survivors, and about 5% develop overt heart failure, with some requiring cardiac transplantation. Anthracycline-induced cardiotoxicity in children has no safe dose threshold and all doses are likely to cause significant myocardial damage. New data suggests that a unique cardiac mitochondrial exogenous NADH dehydrogenase is responsible for the anthracycline-induced oxygen radicals damage to the heart, and that chelators currently evaluated may not prevent late-onset cardiotoxicity in children. In view of these findings we urge extreme caution in using anthracyclines as part of multimodality ALL treatment programs, and strongly recommend reevaluation of what should be considered the best induction regimen for high-risk childhood ALL.


Subject(s)
Anthracyclines/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Anthracyclines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Drug Resistance, Neoplasm , Drug-Related Side Effects and Adverse Reactions/chemically induced , Drug-Related Side Effects and Adverse Reactions/prevention & control , Heart Diseases/chemically induced , Humans , Infant , Neoplasms, Second Primary/chemically induced , Recurrence , Remission Induction/methods , Risk Assessment
5.
Leuk Lymphoma ; 33(3-4): 289-93, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10221508

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Burkitt Lymphoma/drug therapy , Dacarbazine/analogs & derivatives , Immunotoxins/therapeutic use , N-Glycosyl Hydrolases , Plant Proteins/therapeutic use , Animals , Combined Modality Therapy , Dacarbazine/therapeutic use , Humans , Mice , Mice, SCID , Ribosome Inactivating Proteins, Type 1 , Temozolomide , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured
6.
Clin Cancer Res ; 5(12): 3906-13, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10632319

ABSTRACT

Seven children and eight adults with CD19+ B-lineage acute lymphoblastic leukemia, as well as one adult with chronic lymphocytic leukemia, were treated with the CD19 receptor-directed tyrosine kinase inhibitor B43-Genistein. All patients had failed previous chemotherapy regimens, and six patients had relapsed after bone marrow transplantation. B43-Genistein was administered as a 1-hour i.v. infusion at 0.1-0.32 mg/kg/day dose levels for 10 consecutive days or 3 consecutive days weekly for a total of nine doses. B43-Genistein was well tolerated by all patients with no life-threatening side effects. There were six episodes of grade 2-3 fever, two of which were clearly drug related, one episode each of grade 3 myalgia, grade 2 sinus tachycardia, and grade 2 vascular leak syndrome. There was one durable complete remission and two transient responses. Pharmacokinetic analyses in 12 patients revealed a plasma half-life of 20 +/- 5 h, mean residence time of 24 +/- 5 h, and a systemic clearance rate of 20 +/- 3 ml/h/kg. Moderate levels of human antimouse antibody (HAMA) ranging from 20-87 ng/ml were detected in the day 28 blood samples from three of nine cases examined. Treatment of these three HAMA-positive patients with a second course of B43-Genistein did not yield measurable immunoconjugate levels in the plasma, indicating that the administered B43-Genistein molecules were rapidly cleared from circulation due to the HAMA. On the basis of its acceptable toxicity profile and its ability to elicit objective responses at nontoxic dose levels, B43-Genistein may provide the basis for an effective treatment strategy for B-lineage acute lymphoblastic leukemia patients who have failed standard therapy.


Subject(s)
Antigens, CD19/immunology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/enzymology , Enzyme Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Adolescent , Adult , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/biosynthesis , Antigens, CD19/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Burkitt Lymphoma/pathology , Child , Child, Preschool , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Genistein/adverse effects , Genistein/pharmacokinetics , Genistein/therapeutic use , Humans , Immunotoxins/adverse effects , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Male , Middle Aged , Pilot Projects
7.
Clin Cancer Res ; 4(5): 1125-34, 1998 May.
Article in English | MEDLINE | ID: mdl-9607569

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , Genistein/pharmacology , Liver/drug effects , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Disease-Free Survival , Drug Combinations , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Epidermal Growth Factor/pharmacokinetics , Epidermal Growth Factor/therapeutic use , ErbB Receptors/antagonists & inhibitors , Female , Genistein/pharmacokinetics , Genistein/therapeutic use , Humans , Liver/pathology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, SCID , Subrenal Capsule Assay , Survival Analysis , Tumor Cells, Cultured
8.
Clin Cancer Res ; 4(1): 165-70, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9516966

ABSTRACT

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.


Subject(s)
Antigens, CD19/immunology , Antineoplastic Agents/toxicity , Enzyme Inhibitors/toxicity , Genistein/toxicity , Immunoconjugates/toxicity , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Female , Immunoconjugates/pharmacokinetics , Macaca fascicularis
9.
Clin Cancer Res ; 3(6): 881-90, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9815763

ABSTRACT

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.


Subject(s)
Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Immunotoxins/pharmacokinetics , Immunotoxins/toxicity , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Plant Proteins/pharmacokinetics , Plant Proteins/toxicity , Animals , Antibody Formation , Antigens, CD7/immunology , Heart/drug effects , Humans , Immunoconjugates/therapeutic use , Immunoglobulin G/biosynthesis , Immunotoxins/therapeutic use , Liver/drug effects , Liver/pathology , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Myocardium/pathology , Plant Proteins/therapeutic use , Ribosome Inactivating Proteins, Type 1 , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Clin Cancer Res ; 3(3): 325-37, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815689

ABSTRACT

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.


Subject(s)
Antigens, CD19/immunology , Antiviral Agents/pharmacokinetics , Immunotoxins/pharmacokinetics , N-Glycosyl Hydrolases , Plant Proteins/pharmacokinetics , Animals , Antiviral Agents/toxicity , Humans , Immunotoxins/blood , Immunotoxins/toxicity , Injections, Intravenous , Kidney/drug effects , Kidney/pathology , Kinetics , Macaca fascicularis , Mice , Models, Biological , Plant Proteins/blood , Plant Proteins/toxicity , Proteinuria , Ribosome Inactivating Proteins, Type 1
11.
Clin Cancer Res ; 3(12 Pt 1): 2217-27, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9815618

ABSTRACT

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.


Subject(s)
Diphtheria Toxin/pharmacokinetics , Diphtheria Toxin/therapeutic use , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Animals , Cytarabine/therapeutic use , Diphtheria Toxin/toxicity , Disease-Free Survival , Doxorubicin/therapeutic use , Female , HL-60 Cells , Humans , Immunotoxins/toxicity , Macaca fascicularis , Mice , Mice, SCID , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/toxicity , Tissue Distribution , Transplantation, Heterologous
12.
Leuk Lymphoma ; 23(1-2): 61-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-9021687

ABSTRACT

We used a SCID mouse xenograft model to study the in vivo growth patterns of primary leukemic cells from six patients with newly diagnosed B-cell precursor (BCP) acute lymphoblastic leukemia (ALL), including two patients with t(1;19) ALL, two patients with t(4;11) ALL, and two patients with t(9;22) ALL. Leukemic cells from these six patients caused overt leukemia in SCID mice with extensive multiple organ involvement. Leukemic BCP from SCID mice xenografted with leukemic cells from two t(9;22) ALL patients expressed very high levels of both VLA-4 and VLA-5 regardless of the tissue of origin. By comparison, in SCID mice xenografted with leukemic cells from the two patients with t(1;19) ALL and two patients with t(4;11) ALL, leukemic BCP from the bone marrow samples expressed high levels of VLA-4 as well as VLA-5, whereas the vast majority of leukemic BCP in the liver or spleen samples expressed neither of these adhesion molecules at significant levels. These results suggest that the expression of VLA-4 and VLA-5 on t(1;19) or t(4;11) leukemia cells likely determines their binding capacity to bone marrow stroma and may affect their migration to extramedullary tissues. Our findings are in accord with and extend previous studies which demonstrated that extracellular matrix and integrins influence development, compartmentalization, and migration of BCP during B-cell ontogeny. The described SCID mouse model system provides a unique opportunity to study the adhesion receptors which regulate the selective homing of human leukemic BCP to specific SCID mouse organs.


Subject(s)
Integrin beta1/physiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Receptors, Lymphocyte Homing/physiology , Receptors, Very Late Antigen/physiology , Severe Combined Immunodeficiency/immunology , Animals , B-Lymphocytes/pathology , Cell Adhesion Molecules , Child , Disease Models, Animal , Hematopoietic Stem Cells/pathology , Humans , Integrin alpha4beta1 , Integrins/physiology , Mice , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Fibronectin/physiology
13.
Clin Cancer Res ; 2(9): 1533-42, 1996 Sep.
Article in English | MEDLINE | ID: mdl-9816330

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , N-Glycosyl Hydrolases , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Carmustine/therapeutic use , Doxorubicin/therapeutic use , Etoposide/therapeutic use , Female , Humans , Immunotoxins/therapeutic use , Male , Mice , Mice, SCID , Neoplasm Transplantation , Plant Proteins/therapeutic use , Ribosome Inactivating Proteins, Type 1 , Specific Pathogen-Free Organisms , Treatment Outcome
14.
J Biol Chem ; 271(13): 7659-64, 1996 Mar 29.
Article in English | MEDLINE | ID: mdl-8631803

ABSTRACT

We demonstrate that the CD19 receptor associates with the beta1 family integrin receptors on human B-cell precursors as well as mature B-lymphocytes, and engagement of the beta1 family integrin receptors with monoclonal antibody homoconjugates leads to rapid activation of the CD19-associated protein-tyrosine kinases (PTK) and results in hyperphosphorylation of CD19 on tyrosine residues. Our findings prompt the hypothesis that homoconjugate-induced integrin clustering may effect the approximation and, by intermolecular cross-phosphorylation, activation of the CD19-associated PTK and subsequent tyrosine phosphorylation of the CD19 receptor. The ability of the beta1 family integrin receptors to transmit a biochemical signal triggering the CD19-linked multifunctional PTK pathway provides a possible explanation for the pleiotropic biologic responses generated though adhesive VLA-4- and VLA-5-mediated contacts.


Subject(s)
Antigens, CD19/physiology , B-Lymphocytes/immunology , Integrin beta1/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibronectin/physiology , Signal Transduction , B-Lymphocytes/physiology , Enzyme Activation , Enzyme Precursors/metabolism , Immunoblotting , Integrin alpha4beta1 , Integrins/biosynthesis , Integrins/physiology , Intracellular Signaling Peptides and Proteins , Phosphorylation , Phosphotyrosine/analysis , Protein-Tyrosine Kinases/metabolism , Receptors, Fibronectin/biosynthesis , Receptors, Lymphocyte Homing/biosynthesis , Receptors, Lymphocyte Homing/physiology , Receptors, Very Late Antigen/physiology , Syk Kinase
15.
Nucleic Acids Res ; 19(1): 117-23, 1991 Jan 11.
Article in English | MEDLINE | ID: mdl-2011492

ABSTRACT

Molecular cloning of genomic sequences altered in cancer cells is believed to lead to the identification of new genes involved in the initiation and progression of the malignant phenotype. DNA amplification is a frequent molecular alteration in tumor cells, and is a mode of proto-oncogene activation. The cytologic manifestation of this phenomenon is the appearance of chromosomal homogeneously staining regions (HSRs) or double minute bodies (DMs). The gastric carcinoma cell line KATO III is characterized by a large HSR on chromosome 11. In-gel renaturation analysis confirmed the amplification of DNA sequences in this cell line, yet none of 42 proto-oncogenes that we tested is amplified in KATO III DNA. We employed the phenol-enhanced reassociation technique (PERT) to isolate 21 random DNA fragments from the amplified domain, and used 6 of them to further clone some 150 kb from that genomic region. While in situ hybridization performed with some of these sequences indicated that in KATO III they are indeed amplified within the HSR on chromosome 11, somatic cell hybrid analysis and in situ hybridization to normal lymphocyte chromosomes showed that they are derived from chromosome 10, band q26. The same sequences were found to be amplified in another gastric carcinoma cell line, SNU-16, which contains DMs, but were not amplified in other 70 cell lines representing a wide variety of human neoplasms. One of these sequences was highly expressed in both KATO III and SNU-16. Thus, the cloned sequences supply a starting point for identification of novel genes which might be involved in the pathogenesis of gastric cancers, and are located in a relatively unexplored domain of the human genome.


Subject(s)
Chromosomes, Human, Pair 10 , DNA, Neoplasm/isolation & purification , Gene Amplification , Stomach Neoplasms/genetics , Adult , Base Sequence , Blotting, Northern , Blotting, Southern , Cloning, Molecular/methods , DNA, Neoplasm/biosynthesis , Humans , Phenols , Proto-Oncogene Mas , Proto-Oncogenes , Tumor Cells, Cultured
16.
Immunol Lett ; 17(3): 285-91, 1988 Mar.
Article in English | MEDLINE | ID: mdl-3372009

ABSTRACT

We examined the sera of 170 patients with various autoimmune diseases other than systemic lupus erythematosus (SLE) for the presence of an anti-DNA antibody idiotype termed 16/6 and known to occur with high frequency in sera of patients with SLE. The idiotype was found in 6/15 sera from patients with polymyositis (49%), 3/18 with multiple sclerosis (17%), 3/18 with primary Sjögren's syndrome (18%), 9/40 with autoimmune thyroid diseases (23%), 2/35 with myasthenia gravis (6%), and 3/42 patients with rheumatoid arthritis (7%). The idiotype was not detected among 12 patients with scleroderma or 77 normal controls. The presence of the 16/6 idiotype was associated with the presence of another anti-DNA idiotype termed 134-Id. Serum samples were also tested for activity against DNA, various synthetic polynucleotides, and cardiolipin. The serum activity against these antigens was found to be polyspecific, though overlap in reaction against the various polynucleotides was not absolute. The 16/6 idiotype is thought to be coded by a germline gene. The presence of this idiotype in various autoimmune diseases points to a pathophysiologic link between the diseases.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , DNA/immunology , Immunoglobulin Idiotypes/immunology , Cardiolipins/immunology , Humans , Lupus Erythematosus, Systemic/immunology , Polynucleotides/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...